• Rezultati Niso Bili Najdeni

View of Epidermolysis bullosa, wound management and new therapeutic approaches

N/A
N/A
Protected

Academic year: 2022

Share "View of Epidermolysis bullosa, wound management and new therapeutic approaches"

Copied!
11
0
0

Celotno besedilo

(1)

1 Institute of Biophysics, Faculty of medicine, University of Ljubljana, Ljubljana, Slovenia

2 Department of Dermatovenereology, University Medical Centre Ljubljana, Ljubljana, Slovenia

3 Medical Center for Molecular Biology, Faculty of medicine, University of Ljubljana, Ljubljana, Slovenia

Correspondence/

Korespondenca:

Mirjana Liovic, e: mirjana.

liovic@mf.uni-lj.si Key words:

epidermolysis bullosa;

mutation; therapy Ključne besede:

bulozna epidermoliza;

mutacija; zdravljenje Received: 12. 3. 2019 Accepted: 5. 8. 2019

en article-lang

10.6016/ZdravVestn.2937 doi

12.3.2019 date-received

5.8.2019 date-accepted

Metabolic and hormonal disorders Metabolne in hormonske motnje discipline

Professional article Strokovni članek article-type

Epidermolysis bullosa, wound management

and new therapeutic approaches Dedne bulozne epidermolize, oskrba ran in nove možnosti zdravljenja

article-title

Epidermolysis bullosa Dedne bulozne epidermolize alt-title

epidermolysis bullosa, mutation, therapy bulozna epidermoliza, mutacija, zdravljenje kwd-group The authors declare that there are no conflicts

of interest present. Avtorji so izjavili, da ne obstajajo nobeni

konkurenčni interesi. conflict

year volume first month last month first page last page

2020 89 1 2 107 117

name surname aff email

Mirjana Liovic 3 mirjana.liovic@mf.uni-lj.si

name surname aff

Špela Zemljič-Jokhadar 1

Vlasta Dragoš 2

eng slo aff-id

Institute of Biophysics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia

Inštitut za biofiziko, Medicinska fakulteta, Univerza v ljubljani, Ljubljana, Slovenija

1

Department of

Dermatovenereology, University Medical Centre Ljubljana, Ljubljana, Slovenia

Dermatovenerološka klinika, Univerzitetni klinični center ljubljana, Ljubljana, Slovenija

2

Medical Center for Molecular Biology, Faculty of medicine, University of Ljubljana, Ljubljana, Slovenia

Medicinski center za molekularno biologijo, Medicinska fakulteta, Univerza v Ljubljani, Ljubljana, Slovenija

3

Epidermolysis bullosa, wound management and new therapeutic approaches

Dedne bulozne epidermolize, oskrba ran in nove možnosti zdravljenja

Špela Zemljič-Jokhadar,1 Vlasta Dragoš,2 Mirjana Liovic3

Abstract

Epidermolysis bullosa is a hereditary skin fragility disorder, which is linked to mutations in 18 genes that are expressed in the skin. Today we distinguish 4 main EB types, which include about 30 different disease subtypes with a variety of clinical symptoms. The main types are classified according to the skin layer that is affected: EB simplex linked to the epidermis; junctional EB linked to the basal membrane, and dystrophic EB linked to the dermis. Kindler syndrome is the fourth and very rare type of EB. In all cases, the patient’s skin is very fragile, so the basic clin- ical symptoms are skin blisters and wounds that heal with difficulty. EB affects patients both physically and psychologically, and the chronic inflammation accompanying the disease often leads to aggressive forms of squamous cell carcinoma. This is a group of rare genetic diseases with an incidence of 1:40000. In Slovenia there are 60 patients with EB. In this article we present the current standard of care of patients with this still incurable disorder, as well as the newest experimental methodologies aiming at the development of possible genetic, regenerative and pharmacological treatments of EB.

Izvleček

Bulozna epidermoliza (EB) je dedna bolezen krhkosti kože, povezana z mutacijami 18 različnih genov, ki se izražajo v koži. Zaradi raznovrstnosti simptomov se razvršča v 4 osnovne tipe s 30 različnimi podtipi. Osnovni tipi EB so razdeljeni glede na plast kože, ki je z mutacijo prizadeta:

EB simpleks, ki je vezana na epidermis, junkcijska EB, ki je vezana na bazalno membrano, ter distrofična EB, ki je vezana na dermis. Poznamo še Kindlerjev sindrom, ki je izredno redka ob- lika te bolezni. Pri vseh primerih je koža bolnikov zelo krhka, zato so osnovni simptomi mehurji in odprte rane, ki se težko celijo. Bolezen prizadene bolnike telesno in duševno, kronični vnet- ni procesi pa pogosto povzročijo nastanek agresivnih oblik ploščatoceličnega karcinoma kože.

Gre za skupino redkih bolezni z incidenco okoli 1 : 40.000. V Sloveniji imamo 60 bolnikov z EB. V članku predstavljamo trenutno uveljavljen način oskrbe kroničnih ran bolnikov z EB in pregled najnovejših in najsodobnejših genetskih, regenerativnih in farmakoloških pristopov za razvoj načina zdravljenja te še vedno neozdravljive bolezni.

Cite as/Citirajte kot: Zemljič-Jokhadar Š, Dragoš V, Liovic M. Epidermolysis bullosa, wound management and new therapeutic approaches. Zdrav Vestn. 2020;89(1–2):107–17.

DOI: https://doi.org/10.6016/ZdravVestn.2937

Copyright (c) 2020 Slovenian Medical Journal. This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.

Slovenian Medical

Journal

(2)

1 Introduction

Epidermolysis bullosa (epidermolysis bullosa – EB) is a hereditary skin disor- der with characteristic blisters, erosions and wounds arising from a mild mechan- ic skin stimulation (Figure 1). EB patients frequently have numerous comorbidities, depending on the type of epidermolysis bullosa, such as dehydration, secondary lesion infections, onset of granulated tis- sues, atrophic scarring, post-inflammato- ry pigmentation, as well as poikiloderma, hypertrophic scars, milia, palmoplantar keratodermas, cutis aplasia. Late conse- quences of the basic diseases are skin car- cinoma, both basal cell carcinoma (BCC), as well as squamous cell carcinoma (SCC) (1). Accessory structures of the skin can manifest dystrophy or a complete loss of nails, scarring alopecia or alopecia univer- salis or hypotrichosis. In the mucosa of the mouth, patients may develop microsto- mia, obliteration of the vestibulum, enam- el hypoplasia, numerous cases of caries, missing teeth, or periodontitis. There may be an onset of corneal erosions, eyelid in- fections, corneal scarring, symblepharon,

ectropion, corneal infection and vision impairment with blindness, and conjunc- tival granulomata. In the area of the inner ear, there is a tightening of the outer ear canal, conductive hearing loss and, in the area of the nose, a tightening of the nos- trils resulting from granulations.

Other possible complications on oth- er organ systems with some types of epi- dermolysis bullosa occur in the digestive tract: pyloric atresia, oesophageal stric- ture, chronic constipation, gastroesoph- ageal reflux, anal fissures, protein losing enteropathy, colitis. In the genitourinary system, there can be urethral strictures, malformation of the female genital tract, vesicoureteral obstructions and stenoses, scarring in the vulva, kidney failure. In the upper respiratory tract, there are la- ryngotracheal stenoses. In the musculo- skeletal system, the following may occur:

osteopenia, osteoporosis, flexural limb contractures, finger contractures and fu- sion (pseudosyndactyly), webbed fingers and toes, muscle atrophy. Patients may also suffer from anaemia from different conditions, most often from lack of iron.

There are also cases of dilated cardiomy-

Figure 1: Some clinical symptoms of EB. a) aplasia cutis congenita; b) blood-fluid-filled blisters; c) erosions.

the functional and structural integrity of the epidermis, basal membrane or dermis.

EB phenotypes are expressed in a broad spectrum of symptoms (4). We differenti- ate between 4 basic EB types: EB simplex (EBS), junctional EB (JEB), dystrophic EB (DEB) and the Kindler syndrome (Table 1). This classification is based on the lay- er in which the skin tissue cleavages. In the scope of basic types, there are at least 30 clinically different EB subtypes, differ- entiated by scope and severity of changes to the skin and mucous membranes, lo- cation and impact on the internal organs and tissues (2). The disease can be inher- ited either dominantly or recessively. EBS is characteristically inherited dominantly, however there are 18 known families that have the recessive type of the disease in the world (5-13). It is mostly tied to the muta- tions in the genes for keratin 5 or 14. On the other hand, JEB is exclusively recessive in nature and is linked to the mutations in the genes for α6β4 integrin, collagen 17 or laminin 5. Unlike those, DEB is inherited both along the dominant and the recessive path, and is tied to mutations in the genes which encode collagen 7. Kinder syn- drome is an exceptionally rare type of the disease, which is linked to the mutations in the KIND1 (or FERMT1) gene, which encodes the kindlin protein. Kindlin has a special role in connecting actin filaments with the extracellular matrix. The disease is inherited recessively.

The estimated extent of EB in USA (the National Epidermolysis Bullosa Register – NEBR, which has analysed the data for 16 years) is estimated at 11 cases per mil- lion, with a frequency of approximately 20 cases per million children born (15). In Slovenia, the incidence is approximately 30 cases per million. It is estimated that currently there are approximately 500,000 patients suffering from EB globally.

In 2014, a new classification system for diagnosing EB was developed, which in- cludes the EB type, method of inheritance, phenotype characteristic (scope of wounds and various extracutaneous symptoms), Table 1: Main types and subtypes of hereditary epidermolysis bullosa (14).

Level of skin

cleavage Major EB

type Major EB

subtype Targeted protein(s)

Intraepidermal EBS Suprabasal EBS transglutaminase 5; plakophilin 1;

desmoplakin; plakoglobin Basal EBS transglutaminase 5; plakophilin 1;

desmoplakin; plakoglobin Intralamina

lucida JEB JEB,

generalised laminin-332, collagen XVII; α6β4 integrin; α3 integrin subunit

JEB, localised collagen XVII; laminin-332; α6β4 integrin Sublamina

densa DEB DDEB collagen VII

RDEB collagen VII

Mixed Kindler

syndrome Fermitin family homolog 1 (kindlin-1)

DDEB – Dominant dystrophic epidermolysis bullosa; DEB – dystrophic epidermolysis bullosa; EB – epidermolysis bullosa; EBS – simplex epidermolysis bullosa, JEB – junction epidermolysis bullosa;

RDEB – recessive dystrophic epidermolysis bullosa

(3)

ectropion, corneal infection and vision impairment with blindness, and conjunc- tival granulomata. In the area of the inner ear, there is a tightening of the outer ear canal, conductive hearing loss and, in the area of the nose, a tightening of the nos- trils resulting from granulations.

Other possible complications on oth- er organ systems with some types of epi- dermolysis bullosa occur in the digestive tract: pyloric atresia, oesophageal stric- ture, chronic constipation, gastroesoph- ageal reflux, anal fissures, protein losing enteropathy, colitis. In the genitourinary system, there can be urethral strictures, malformation of the female genital tract, vesicoureteral obstructions and stenoses, scarring in the vulva, kidney failure. In the upper respiratory tract, there are la- ryngotracheal stenoses. In the musculo- skeletal system, the following may occur:

osteopenia, osteoporosis, flexural limb contractures, finger contractures and fu- sion (pseudosyndactyly), webbed fingers and toes, muscle atrophy. Patients may also suffer from anaemia from different conditions, most often from lack of iron.

There are also cases of dilated cardiomy-

Figure 1: Some clinical symptoms of EB. a) aplasia cutis congenita; b) blood-fluid-filled blisters; c) erosions.

opathy. Among endocrine disorders, there may be a delay in the onset of puberty and amenorrhea. Other system complications include sepsis, delayed growth and de- velopment, and resulting from any of the numerous above comorbidities also a dis- ability (2).

The reason for the onset of fragility of the skin and epithelial tissue is one of 18 different genes which encode 18 structural and adhesive proteins of the cells and ex- tracellular matrix. They are manifested in a part of skin and have a width of just a few hundred micrometres (Figure 2).

The impacted layer includes the basal part of the epidermis, the basal membrane and partially also dermis. Mutations in tar- get genes most often cause the change in one amino acid in a protein (missense) or premature termination of gene expression, which results in a protein folding problem and consequently incorrect expression or lack of protein (3). Mutations can lead to irregularities in these proteins that impact

the functional and structural integrity of the epidermis, basal membrane or dermis.

EB phenotypes are expressed in a broad spectrum of symptoms (4). We differenti- ate between 4 basic EB types: EB simplex (EBS), junctional EB (JEB), dystrophic EB (DEB) and the Kindler syndrome (Table 1). This classification is based on the lay- er in which the skin tissue cleavages. In the scope of basic types, there are at least 30 clinically different EB subtypes, differ- entiated by scope and severity of changes to the skin and mucous membranes, lo- cation and impact on the internal organs and tissues (2). The disease can be inher- ited either dominantly or recessively. EBS is characteristically inherited dominantly, however there are 18 known families that have the recessive type of the disease in the world (5-13). It is mostly tied to the muta- tions in the genes for keratin 5 or 14. On the other hand, JEB is exclusively recessive in nature and is linked to the mutations in the genes for α6β4 integrin, collagen 17 or laminin 5. Unlike those, DEB is inherited both along the dominant and the recessive path, and is tied to mutations in the genes which encode collagen 7. Kinder syn- drome is an exceptionally rare type of the disease, which is linked to the mutations in the KIND1 (or FERMT1) gene, which encodes the kindlin protein. Kindlin has a special role in connecting actin filaments with the extracellular matrix. The disease is inherited recessively.

The estimated extent of EB in USA (the National Epidermolysis Bullosa Register – NEBR, which has analysed the data for 16 years) is estimated at 11 cases per mil- lion, with a frequency of approximately 20 cases per million children born (15). In Slovenia, the incidence is approximately 30 cases per million. It is estimated that currently there are approximately 500,000 patients suffering from EB globally.

In 2014, a new classification system for diagnosing EB was developed, which in- cludes the EB type, method of inheritance, phenotype characteristic (scope of wounds and various extracutaneous symptoms), Table 1: Main types and subtypes of hereditary epidermolysis bullosa (14).

Level of skin

cleavage Major EB

type Major EB

subtype Targeted protein(s)

Intraepidermal EBS Suprabasal EBS transglutaminase 5; plakophilin 1;

desmoplakin; plakoglobin Basal EBS transglutaminase 5; plakophilin 1;

desmoplakin; plakoglobin Intralamina

lucida JEB JEB,

generalised laminin-332, collagen XVII; α6β4 integrin; α3 integrin subunit

JEB, localised collagen XVII; laminin-332; α6β4 integrin Sublamina

densa DEB DDEB collagen VII

RDEB collagen VII

Mixed Kindler

syndrome Fermitin family homolog 1 (kindlin-1)

DDEB – Dominant dystrophic epidermolysis bullosa; DEB – dystrophic epidermolysis bullosa; EB – epidermolysis bullosa; EBS – simplex epidermolysis bullosa, JEB – junction epidermolysis bullosa;

RDEB – recessive dystrophic epidermolysis bullosa

(4)

discoveries in mapping immunofluores- cent tagged antigens and mutations pres- ents in an individual patient (14).

2 Diagnosing EB

The first step in diagnosing EB type in a patient is to determine the level of the location of the occurrence of blisters in the skin, usually by immunofluorescent anti- gen mapping and/or transmission elec- tronic microscopy (1). Using monoclonal antibodies focused on the components of the epidermal basement membrane and an epidermal antigen, we can additionally improve subclassification.

At the level of the skin cleavage, we can classify them into one of the four EB groups. Further diagnostics require a ge- netic analysis of the mutation, as it makes it possible to classify the disease in more detail and to provide further genetic con-

Figure 2: A schematic of a part of the skin (frame) with EB. This is a thin layer encompassing the lower part of the epidermis, basal membrane and the surface part of the dermis in which 18 proteins are express – potential generators of hereditary EB. Some of the most frequent ones are marked in the schematic.

sultation (14). If genetic analysis is not available, EB is classified by phenotype characteristics, such as dispersion (local- ised or general), severity and the presence of extracutaneous complications. The pa- tient’s family history can provide an in- sight into how it was inherited (1).

3 Wound care

Pope et al. (16) developed a procedure for treating wounds, consisting of four steps:

1. EB type, comorbidities, age, nutritional status, systemic comorbidities of other organ systems.

2. Care is focused on the patient and man- agement of their pain, itching, day-to- day activities and consulting regarding therapy.

3. Local treatment of the wound: deter- mining the location and the charac-

(5)

teristics of the wounds, regular gentle cleaning, removing necrotic tissue so that the lesion heals sooner, treating colonisation/inflammation/infection, selecting the appropriate local therapy and dressing, assessment of healing.

4. With wounds that heal poorly or not at all, we conduct a biopsy and a histolog- ical verification when suspecting skin carcinoma.

It is important to have the support of various medical providers, such as special- ised nurses or cooperation with various clinics when treating patients.

Local treatment of wounds depends on the type of lesion we are dealing with.

There are numerous dressings available, and we divide them into three groups:

non-occlusive, partially occlusive and ac- tive or biological dressings (16,17). Select- ing the appropriate dressing depends on the level the wound is healing. A necrot- ic wound must be treated with a dressing that uses hydrogel and in combination with polyurethane film or alginate. When a wound is inflamed, an alginate dressing will provide appropriate conditions for the wound to heal. In the proliferative phase and in the maturation phase, a dressing is needed to regulate moisture by soaking in the excess exudation (polyurethane or silicon foam and other). With an infect- ed wound, topical antimicrobial drugs and antimicrobial dressings (with silver, honey, charcoal, polyaminopropyl bigua- nide, dialkylcarbomoyl chloride) must be used (16,17). To prevent the onset of new blisters, silicon dressings are often used (16).

4 Patient care in Slovenia

The rare occurrence of EB and the im- pact on numerous other organs and sys- tems (beyond the skin) are a special chal- lenge for appropriate treatment of these patients (2). In the past few decades in Slo- venia, an interdisciplinary patient treat- ment has been developed, where groups

include dermatologists as well as other medical specialists. These include neo- natologists, paediatricians and internists, pathologists, medical geneticist, otorhino- laryngologists, ophthalmologists, plastic surgeons, orthopaedic surgeons, paedi- atric gastroenterologists, haematologists, dentists, anaesthesiologists, endocrinolo- gists, neurologists, radiologists, cardiolo- gists, nephrologists, specialists of physical therapy. The treatment also includes dis- trict nurses and a dietician from early on.

The main concern of the group is to reg- ularly monitor the skin and the wounds, diagnosing skin carcinoma early, regularly monitoring the complications on other organ systems, itching relief management and pain prevention.

5 Development of new therapy approaches

The main reason for the onset of EB is genetic changes to key structural genes which encode different structural proteins that connect the three layers of skin. For a complete recovery, the mutation would have to be fixed. Skin in our biggest organ by size. A therapeutic approach for treat- ing this disease would have to fix all the cells that for the damaged proteins. Unlike other organs, such as lungs, heart, liver, etc., skin is readily accessible. Therefore, therapy of hereditary skin diseases could potentially utilize an approach that would mitigate the clinical signs. In the past ten years, we have seen a major breakthrough in this area, even though none of numer- ous promising and tested approaches has proven to provide routine EB therapy.

Besides the long-term procedure of intro- ducing new medications and clinical stud- ies, one of the reasons is probably the high price of genetic therapy: up to 6 million euros per patient.

Past new therapeutic approaches are divided into: cellular therapy, gene ther- apy, protein therapy and system therapy (Figure 3). Below, we present a few of the most interesting approaches.

(6)

5.1 Cellular therapies

Cellular therapies include past trials with allogeneic fibroblasts, mesenchymal stem cells, stem cells from the umbili- cal cord and bone marrow transplant. In 2009, Kern et al. (18) did a study on more than 50 mice with recessive dystrophic EB; in separate experiments, they subder- mally injected healthy mouse and human fibroblasts. They established that the pro- cedure was safe (there was a mild tempo- rary inflammation). The level of collagen 7 (which is tied to the dystrophic EB type) increased at the dermo-epidermal con- nection by 3.5- to 4.7-fold. It is interesting that the injected cells produced healthy collagen 7 for a month, while the protein was stable for longer. Consequently, the skin of the mice maintained the improved phenotype for more than 100 days after the procedure. Injected cells remained at the injection point, did not divide, form

tumours or fibrotic tissue. In 2010, a study was published (10) in which two patients with RDEB were subcutaneously injected allogenic mesenchymal stem cells, isolated from bone marrow. After a while, the cells disappeared; however, their effect was visi- ble already after one week (the dermo-epi- dermal barrier was re-established), and lasted up to 4 months. In 2013, another study was conducted (20), in which a sim- ilar approach to the one performed on the mice was tested on patients with RDEB.

Allogenic fibroblasts were injected into the skin around the wounds. Cells survived a few weeks in the tissue, then disappeared, while their positive effect lasted up to 28 days after the procedure. In 2015, Petrof et al. (21) published a new study based on ten RDEB patients. They delivered allo- genic mesenchymal stem cells from bone marrow by infusion. The effect was visible in two months after several repeated pro- cedures. The surface of the affected skin

grew back faster, and the skin inflamma- tion was significantly reduced. Patients said that the positive effect was present for 4 – 6 months. El-Darouti et al. (22) published a study in 2016, in which they conducted a cellular infusion of non-he- matopoietic bone marrow cells, which are proven to be able to differentiate into fi- broblasts. They had two groups of RDEB patients: the first received an infusion of stem cells with added ciclosporin, while the second group received stem cells with- out ciclosporin. In both groups, the effect was very similar (there were no statistical- ly significant differences between them):

the patients’ health condition apparently improved, their wounds grew back faster, and one year after the test was completed, there were no major negative consequenc- es. It has been established that in proce- dures which capture mesenchymal stem cells, they do not divide or differentiate into healthy cells after the transplant, but operate indirectly by secreting numerous growth factors and cytokines which affect the surrounding tissue and thereby accel- erate wound healing and inflammation re- duction. Unfortunately, their effect, albeit very positive, was again just transitory and not long-lasting. We also should mention an exceptional experiment in which a se- lect group of children with RDEB received bone marrow transplants (23). Among the seven patients, one died before the transplant because of exceptionally ag- gressive therapy (immuno-myeloablative therapy). After bone marrow transplant, another patient died as a result of the re- jection of the implant and an infection.

With the remaining 5 patients, the phy- sicians established that within half a year after the procedure, there was a significant improvement to their condition, resulting from the build-up of healthy collagen 7 in the basal membrane area. Two years after the procedure, there was still a presence of foreign cells in the skin (chimerism) and an improved health condition.

Figure 3: Basic principles of some new therapeutic approaches for EB.

Protein therapy Cellular therapy

Gene therapy

Growing donor cells in culture

Tissue biopsy

Healthy donor

Isolation and multiplication of donor stem cells

Delivery of healthy donor

stem cells to the patient Transfection

Delivery of the healthy protein to the patient Bioreactor,

expression of the recombined

protein Cleaning

the protein Recombined construct

Slicing the mutated gene and replacing it with a healthy copy of the gene

Gene editing technology

Retrovirus with a healthy copy of the gene

Growing and multiplication of repaired cells

Delivery of the patient’s repaired cells

Patient’s cells grown in culture

Tissue biopsy Retrovirus with a

healthy copy of the gene Healthy copy of the gene Target cells for in vitro

expression of the healthy gene

Vector

Patient

(7)

grew back faster, and the skin inflamma- tion was significantly reduced. Patients said that the positive effect was present for 4 – 6 months. El-Darouti et al. (22) published a study in 2016, in which they conducted a cellular infusion of non-he- matopoietic bone marrow cells, which are proven to be able to differentiate into fi- broblasts. They had two groups of RDEB patients: the first received an infusion of stem cells with added ciclosporin, while the second group received stem cells with- out ciclosporin. In both groups, the effect was very similar (there were no statistical- ly significant differences between them):

the patients’ health condition apparently improved, their wounds grew back faster, and one year after the test was completed, there were no major negative consequenc- es. It has been established that in proce- dures which capture mesenchymal stem cells, they do not divide or differentiate into healthy cells after the transplant, but operate indirectly by secreting numerous growth factors and cytokines which affect the surrounding tissue and thereby accel- erate wound healing and inflammation re- duction. Unfortunately, their effect, albeit very positive, was again just transitory and not long-lasting. We also should mention an exceptional experiment in which a se- lect group of children with RDEB received bone marrow transplants (23). Among the seven patients, one died before the transplant because of exceptionally ag- gressive therapy (immuno-myeloablative therapy). After bone marrow transplant, another patient died as a result of the re- jection of the implant and an infection.

With the remaining 5 patients, the phy- sicians established that within half a year after the procedure, there was a significant improvement to their condition, resulting from the build-up of healthy collagen 7 in the basal membrane area. Two years after the procedure, there was still a presence of foreign cells in the skin (chimerism) and an improved health condition.

Figure 3: Basic principles of some new therapeutic approaches for EB.

Protein therapy Cellular therapy

Gene therapy

Growing donor cells in culture

Tissue biopsy

Healthy donor

Isolation and multiplication of donor stem cells

Delivery of healthy donor

stem cells to the patient Transfection

Delivery of the healthy protein to the patient Bioreactor,

expression of the recombined

protein Cleaning

the protein Recombined construct

Slicing the mutated gene and replacing it with a healthy copy of the gene

Gene editing technology

Retrovirus with a healthy copy of the gene

Growing and multiplication of repaired cells

Delivery of the patient’s repaired cells

Patient’s cells grown in culture

Tissue biopsy Retrovirus with a

healthy copy of the gene Healthy copy of the gene Target cells for in vitro

expression of the healthy gene

Vector

Patient

5.2 Gene therapies

Experiments with gene therapy in- clude: delivery of a healthy copy of the gene through a virus under ex vivo con- ditions, retroviral and lentiviral vectors (24-28), and non-viral experiments. The latter include: RNA interference or in- hibition of the damaged gene expression (29), using antisense oligoribonucleotides (AON) for suppressing mutated exons of a gene when copying RNA into the pro- tein, aminoglycosides for suppressing the premature termination codons (30), as well as using various proteins for gene ed- iting, such as zing finger nucleases (ZNF), Talens, Crispr/Cas9 (31).

With ex vivo gene therapy, the patient’s cells are removed in a skin biopsy, then grown under in vitro conditions. Repairing the mutated gene in the cells is also done in vitro in a culture. Then the repaired cells are inserted back into the patient. All these approaches are detailed in review articles (32-34); however, we will provide more information on the recently published first successful gene therapy procedure, which encompassed nearly all of skin sur- face (80%) of a patient with JEB, related to the mutations in the gene for laminin 332 (LAMB3) (35). After skin biopsy, isolating patient’s keratinocytes, growing cells and in vitro repair of the broken gene by deliv- ering a copy of a healthy gene through the lentiviral vector, the authors also complet- ed clonal tracing of autologous repaired cells. It has been proven that human epi- dermis regenerates thanks to progenitor cells, called holoclones. These are long-liv- ing stem cells of the epidermis of the skin, which can be successfully multiplied in vivo and in vitro. They differentiate into other groups of progenitor cells, which in the end differentiate into epidermis kera- tinocytes. After the selection, they were used as basis for growing epidermal sheets of healthy, autologous cells of the patient, which are then planted into the most dam-

(8)

aged parts of the skin. Using this proce- dure, an eight-year-old boy had 80% of his skin replaced (transplanted), and now has no more problems, except in the areas that were not included in the transplanta- tion. This is an exceptional accomplish- ment, as the boy was in very poor medical condition, and has had a normal life for a few years now since the procedure. Even though the study was exceptional and highly successful, the chances for using a similar procedure for other EB patients are currently low. Besides the very high costs of such treatment, there is still the concern related to possible side effects related to the viral delivery of the healthy gene, as it could result in malignant changes even later in life, which opens a set of different ethical questions related to using such EB therapy.

5.3 Protein therapy

Protein therapy includes intradermal or intravenous delivery ex vivo and un- der in vitro conditions of recombinantly obtained healthy protein (36,37). Healthy collagen 7 was generally delivered topi- cally, which yielded good results (38). All such studies have been conducted at the level of pre-clinical trials and on mouse models; however, in spite of positive ef- fects, they are still only experimental in character.

5.4 Systemic therapies

Systemic therapy encompasses using existing medications for treating oth- er health issues with the objective that some of its effects to improve the health of EB patients. This is especially related to reducing the inflammation response, reducing wound scarring, faster wound healing, etc. The drugs tested so far in- clude the effects of minocycline (39,40), which has anticollagenase action; howev- er, it unfortunately also causes issues with skin hyperpigmentation. Ciclosporin (41) has anti-inflammatory action; but it can

also cause skin cancer. Etanercept (42), which inhibits TNFaplha (anti-inflam- matory action) and Losartan (43), which inhibits TGFbeta (reduced scarring), can affect all the processes, related to these factors in the body. Researchers also tried bone marrow stimulation with the HMG protein (44) in order to increase the num- ber of stem cells in the skin and improve wound growth, as they believe that with EB patients the number of skin stem cells decreases during their lifetime because of constant activation of wound growth.

6 Research in Slovenia

At the Medical Centre for Molecular Biology (Institute of Biochemistry of the Faculty of Medicine, University of Ljublja- na), we have been conducting EB studies at the genetic, protein and cellular levels for nearly 20 years. We collaborate with the Dermatology & Venereology Clinic of the University Medical Centre Ljubljana and numerous international research and medical centres (Guy’s Hospital, King’s College London, United Kingdom; Miller School of Medicine, University of Miami, USA; Institute of Medical Biology, AStar, Singapore; Department of Medical Scienc- es, Uppsala University, Sweden, etc.). In the past few years, our research has been focused on studying the mechanism of the development of the disease and new deliv- ery systems that can be useful especially for delivering therapeutic molecules into the skin (45-48). An especially important achievement was the breakthrough in re- generative medicine and the preparation of the first induced EBS pluripotent stem cell line (iPSC) with the assistance of re- programming (dedifferentiation) of the keratinocytes of an EBS patient (49). In future studies, this cell line will be used as a cellular model and the basis for in vitro preparation of a tissue equivalent of EBS skin. This will then be used for transplant tests of numerous substances that could contribute to faster wound healing or to reducing certain clinical symptoms of EB

(9)

(e.g. inflammation).

7 Conclusion

In the past decade, we have witnessed an exceptional breakthrough in EB re- search mechanisms, understanding of the disease itself and the development of nu- merous strategies for experimental ther- apy approaches that mainly had positive effects. The clinical treatment of these patients has also changed, which contrib- utes to a better quality of living despite the severity of the disease. Support groups play in important contribution, especially the umbrella organization DEBRA Inter- national (dystrophic epidermolysis bullo- sa research association, http://www.deb-

ra-international.org/about-debra.html), which is present in more than 50 coun- tries through local DEBRA organizations, including in Slovenia (Debra Slovenia, http://www.debra-slovenia.si/).

8 Acknowledgment

We would like to thank the Slovenian Research Agency (programme P1–0390), the Ministry of Education, Science and Sports of the Republic of Slovenia, and the EraCoSysMed tender (project 4D-HEAL- ING), the Development Fund of the Uni- versity of Ljubljana, and the institution CELSA Alliance (project Molecular basis of keratopathies) for supporting and fi- nancing our research.

References

1. Fine JD, Johnson LB, Weiner M, Suchindran C. Cause-specific risks of childhood death in inherited epidermolysis bullosa. J Pediatr. 2008;152(2):276-80. DOI: 10.1016/j.jpeds.2007.06.039 PMID: 18206702 2. El Hachem M, Zambruno G, Bourdon-Lanoy E, Ciasulli A, Buisson C, Hadj-Rabia S, et al. Multicentre

consensus recommendations for skin care in inherited epidermolysis bullosa. Orphanet J Rare Dis.

2014;9(1):76. DOI: 10.1186/1750-1172-9-76 PMID: 24884811

3. Has C, Fischer J. Inherited epidermolysis bullosa: new diagnostics and new clinical phenotypes. Exp Dermatol. 2018;28(1146):52. DOI: 10.1111/exd.13668 PMID: 29679399

4. Fine JD, Mellerio JE. Extracutaneous manifestations and complications of inherited epidermolysis bullosa:

part II. Other organs. J Am Acad Dermatol. 2009;61(3):387-402. DOI: 10.1016/j.jaad.2009.03.053 PMID:

19700011

5. Corden LD, Mellerio JE, Gratian MJ, Eady RA, Harper JI, Lacour M, et al. Homozygous nonsense mutation in helix 2 of K14 causes severe recessive epidermolysis bullosa simplex. Hum Mutat. 1998;11(4):279-85. DOI:

10.1002/(SICI)1098-1004(1998)11:4<279::AID-HUMU5>3.0.CO;2-E PMID: 9554744

6. Ciubotaru D, Bergman R, Baty D, Indelman M, Pfendner E, Petronius D, et al. Epidermolysis bullosa simplex in Israel: clinical and genetic features. Arch Dermatol. 2003;139(4):498-505. DOI: 10.1001/

archderm.139.4.498 PMID: 12707098

7. Rugg EL, McLean WH, Lane EB, Pitera R, McMillan JR, Dopping-Hepenstal PJ, et al. A functional “knockout”

of human keratin 14. Genes Dev. 1994;8(21):2563-73. DOI: 10.1101/gad.8.21.2563 PMID: 7525407 8. Jonkman MF, Heeres K, Pas HH, van Luyn MJ, Elema JD, Corden LD, et al. Effects of keratin 14 ablation

on the clinical and cellular phenotype in a kindred with recessive epidermolysis bullosa simplex. J Invest Dermatol. 1996;107(5):764-9. DOI: 10.1111/1523-1747.ep12365805 PMID: 8875963

9. Batta K, Rugg EL, Wilson NJ, West N, Goodyear H, Lane EB, et al. A keratin 14 ‘knockout’ mutation in recessive epidermolysis bullosa simplex resulting in less severe disease. Br J Dermatol. 2000;143(3):621-7.

DOI: 10.1111/j.1365-2133.2000.03722.x PMID: 10971341

10. Lanschuetzer CM, Klausegger A, Pohla-Gubo G, Hametner R, Richard G, Uitto J, et al. A novel homozygous nonsense deletion/insertion mutation in the keratin 14 gene (Y248X; 744delC/insAG) causes recessive epidermolysis bullosa simplex type Köbner. Clin Exp Dermatol. 2003;28(1):77-9. DOI: 10.1046/j.1365- 2230.2003.01218.x PMID: 12558637

11. Has C, Chang YR, Volz A, Hoeping D, Kohlhase J, Bruckner-Tuderman L. Novel keratin 14 mutations in patients with severe recessive epidermolysis bullosa simplex. J Invest Dermatol. 2006;126(8):1912-4. DOI:

10.1038/sj.jid.5700312 PMID: 16614722

(10)

12. Yiasemides E, Trisnowati N, Su J, Dang N, Klingberg S, Marr P, et al. Clinical heterogeneity in recessive epidermolysis bullosa due to mutations in the keratin 14 gene, KRT14. Clin Exp Dermatol. 2008;33(6):689- 97. DOI: 10.1111/j.1365-2230.2008.02858.x PMID: 18713255

13. García M, Santiago JL, Terrón A, Hernández-Martín A, Vicente A, Fortuny C, et al. Two novel recessive mutations in KRT14 identified in a cohort of 21 Spanish families with epidermolysis bullosa simplex. Br J Dermatol. 2011;165(3):683-92. DOI: 10.1111/j.1365-2133.2011.10428.x PMID: 21623745

14. Fine JD, Bruckner-Tuderman L, Eady RA, Bauer EA, Bauer JW, Has C, et al. Inherited epidermolysis bullosa:

updated recommendations on diagnosis and classification. J Am Acad Dermatol. 2014;70(6):1103-26. DOI:

10.1016/j.jaad.2014.01.903 PMID: 24690439

15. Fine JD. Epidemiology of Inherited Epidermolysis Bullosa Based on Incidence and Prevalence Estimates From the National Epidermolysis Bullosa Registry. JAMA Dermatol. 2016;152(11):1231-8. DOI: 10.1001/

jamadermatol.2016.2473 PMID: 27463098

16. Pope E, Lara-Corrales I, Mellerio J, Martinez A, Schultz G, Burrell R, et al. A consensus approach to wound care in epidermolysis bullosa. J Am Acad Dermatol. 2012;67(5):904-17. DOI: 10.1016/j.jaad.2012.01.016 PMID: 22387035

17. Denyer J, Pillay E, Clapham J. International Consensus Best Practice Guidelines for Skin and Wound Care in Epidermolysis Bullosa. London: Wounds International; 2017.

18. Kern JS, Loeckermann S, Fritsch A, Hausser I, Roth W, Magin TM, et al. Mechanisms of fibroblast cell therapy for dystrophic epidermolysis bullosa: high stability of collagen VII favors long-term skin integrity.

Mol Ther. 2009;17(9):1605-15. DOI: 10.1038/mt.2009.144 PMID: 19568221

19. Conget P, Rodriguez F, Kramer S, Allers C, Simon V, Palisson F, et al. Replenishment of type VII collagen and re-epithelialization of chronically ulcerated skin after intradermal administration of allogeneic mesenchymal stromal cells in two patients with recessive dystrophic epidermolysis bullosa. Cytotherapy.

2010;12(3):429-31. DOI: 10.3109/14653241003587637 PMID: 20230217

20. Petrof G, Martinez-Queipo M, Mellerio JE, Kemp P, McGrath JA. Fibroblast cell therapy enhances initial healing in recessive dystrophic epidermolysis bullosa wounds: results of a randomized, vehicle-controlled trial. Br J Dermatol. 2013;169(5):1025-33. DOI: 10.1111/bjd.12599 PMID: 24032424

21. Petrof G, Lwin SM, Martinez-Queipo M, Abdul-Wahab A, Tso S, Mellerio JE, et al. Potential of Systemic Allogeneic Mesenchymal Stromal Cell Therapy for Children with Recessive Dystrophic Epidermolysis Bullosa. J Invest Dermatol. 2015;135(9):2319-21. DOI: 10.1038/jid.2015.158 PMID: 25905587

22. El-Darouti M, Fawzy M, Amin I, Abdel Hay R, Hegazy R, Gabr H, et al. Treatment of dystrophic epidermolysis bullosa with bone marrow non-hematopoeitic stem cells: a randomized controlled trial. Dermatol Ther (Heidelb). 2016;29(2):96-100. DOI: 10.1111/dth.12305 PMID: 26439431

23. Wagner JE, Ishida-Yamamoto A, McGrath JA, Hordinsky M, Keene DR, Woodley DT, et al. Bone marrow transplantation for recessive dystrophic epidermolysis bullosa. N Engl J Med. 2010;363(7):629-39. DOI:

10.1056/NEJMoa0910501 PMID: 20818854

24. Chen M, Kasahara N, Keene DR, Chan L, Hoeffler WK, Finlay D, et al. Restoration of type VII collagen expression and function in dystrophic epidermolysis bullosa. Nat Genet. 2002;32(4):670-5. DOI: 10.1038/

ng1041 PMID: 12426566

25. Woodley DT, Krueger GG, Jorgensen CM, Fairley JA, Atha T, Huang Y, et al. Normal and gene-corrected dystrophic epidermolysis bullosa fibroblasts alone can produce type VII collagen at the basement membrane zone. J Invest Dermatol. 2003;121(5):1021-8. DOI: 10.1046/j.1523-1747.2003.12571.x PMID:

14708601

26. Titeux M, Pendaries V, Zanta-Boussif MA, Décha A, Pironon N, Tonasso L, et al. SIN retroviral vectors expressing COL7A1 under human promoters for ex vivo gene therapy of recessive dystrophic epidermolysis bullosa. Mol Ther. 2010;18(8):1509-18. DOI: 10.1038/mt.2010.91 PMID: 20485266

27. Mavilio F, Pellegrini G, Ferrari S, Di Nunzio F, Di Iorio E, Recchia A, et al. Correction of junctional epidermolysis bullosa by transplantation of genetically modified epidermal stem cells. Nat Med.

2006;12(12):1397-402. DOI: 10.1038/nm1504 PMID: 17115047

28. Chen M, O’Toole EA, Muellenhoff M, Medina E, Kasahara N, Woodley DT. Development and characterization of a recombinant truncated type VII collagen “minigene”. Implication for gene therapy of dystrophic epidermolysis bullosa. J Biol Chem. 2000;275(32):24429-35. DOI: 10.1074/jbc.M003440200 PMID: 10821839 29. Morgan CP, Allen DS, Millington-Ward S, O’Dwyer GE, Palfi A, Jane Farrar G. A mutation-independent

therapeutic strategy for dominant dystrophic epidermolysis bullosa. J Invest Dermatol. 2013;133(12):2793- 6. DOI: 10.1038/jid.2013.241 PMID: 23743647

30. Zingman LV, Park S, Olson TM, Alekseev AE, Terzic A. Aminoglycoside-induced translational read- through in disease: overcoming nonsense mutations by pharmacogenetic therapy. Clin Pharmacol Ther.

2007;81(1):99-103. DOI: 10.1038/sj.clpt.6100012 PMID: 17186006

31. Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science. 2012;337(6096):816-21. DOI: 10.1126/

science.1225829 PMID: 22745249

(11)

32. Gorell E, Nguyen N, Lane A, Siprashvili Z. Gene therapy for skin diseases. Cold Spring Harb Perspect Med.

2014;4(4):a015149. DOI: 10.1101/cshperspect.a015149 PMID: 24692191

33. Abdul-Wahab A, Qasim W, McGrath JA. Gene therapies for inherited skin disorders. Semin Cutan Med Surg.

2014;33(2):83-90. DOI: 10.12788/j.sder.0085 PMID: 25085667

34. Rashidghamat E, McGrath JA. Novel and emerging therapies in the treatment of recessive dystrophic epidermolysis bullosa. Intractable Rare Dis Res. 2017;6(1):6-20. DOI: 10.5582/irdr.2017.01005 PMID:

28357176

35. Hirsch T, Rothoeft T, Teig N, Bauer JW, Pellegrini G, De Rosa L, et al. Regeneration of the entire human epidermis using transgenic stem cells. Nature. 2017;551(7680):327-32. DOI: 10.1038/nature24487 PMID:

29144448

36. Hovnanian A. Systemic protein therapy for recessive dystrophic epidermolysis bullosa: how far are we from clinical translation? J Invest Dermatol. 2013;133(7):1719-21. DOI: 10.1038/jid.2013.137 PMID: 23760051 37. Woodley DT, Wang X, Amir M, Hwang B, Remington J, Hou Y, et al. Intravenously injected recombinant human type VII collagen homes to skin wounds and restores skin integrity of dystrophic epidermolysis bullosa. J Invest Dermatol. 2013;133(7):1910-3. DOI: 10.1038/jid.2013.10 PMID: 23321924

38. Wang X, Ghasri P, Amir M, Hwang B, Hou Y, Khalili M, et al. Topical application of recombinant type VII collagen incorporates into the dermal-epidermal junction and promotes wound closure. Mol Ther.

2013;21(7):1335-44. DOI: 10.1038/mt.2013.87 PMID: 23670575

39. White JE. Minocycline for dystrophic epidermolysis bullosa. Lancet. 1989;1(8644):966. DOI: 10.1016/S0140- 6736(89)92555-5 PMID: 2565464

40. Golub LM, Wolff M, Lee HM, McNamara TF, Ramamurthy NS, Zambon J, et al. Further evidence that tetracyclines inhibit collagenase activity in human crevicular fluid and from other mammalian sources. J Periodontal Res. 1985;20(1):12-23. DOI: 10.1111/j.1600-0765.1985.tb00405.x PMID: 2983061

41. del-Río E. Prevention of blisters in dystrophic recessive epidermolysis bullosa with cyclosporine. J Am Acad Dermatol. 1993;29(6):1038-9. DOI: 10.1016/S0190-9622(08)82040-3 PMID: 8245240

42. Gubinelli E, Angelo C, Pacifico V. A case of dystrophic epidermolysis bullosa improved with etanercept for concomitant psoriatic arthritis. Am J Clin Dermatol. 2010;11:53-4. DOI: 10.2165/1153427-S0-000000000- 00000 PMID: 20586512

43. Fritsch A, Loeckermann S, Kern JS, Braun A, Bösl MR, Bley TA, et al. A hypomorphic mouse model of dystrophic epidermolysis bullosa reveals mechanisms of disease and response to fibroblast therapy. J Clin Invest. 2008;118(5):1669-79. DOI: 10.1172/JCI34292 PMID: 18382769

44. Tamai K, Yamazaki T, Chino T, Ishii M, Otsuru S, Kikuchi Y, et al. PDGFRalpha-positive cells in bone marrow are mobilized by high mobility group box 1 (HMGB1) to regenerate injured epithelia. Proc Natl Acad Sci USA. 2011;108(16):6609-14. DOI: 10.1073/pnas.1016753108 PMID: 21464317

45. Liovic M, Ozir M, Zavec AB, Peternel S, Komel R, Zupancic T. Inclusion bodies as potential vehicles for recombinant protein delivery into epithelial cells. Microb Cell Fact. 2012;11(1):67. DOI: 10.1186/1475-2859- 11-67 PMID: 22624805

46. Zavec AB, Ota A, Zupancic T, Komel R, Ulrih NP, Liovic M. Archaeosomes can efficiently deliver different types of cargo into epithelial cells grown in vitro. J Biotechnol. 2014;192 Pt A:130-5. DOI: 10.1016/j.

jbiotec.2014.09.015 PMID: 25270023

47. Zemljič Jokhadar Š, Klančnik U, Grundner M, Švelc Kebe T, Vrhovec Hartman S, Liović M, et al. GPMVs in variable physiological conditions: could they be used for therapy delivery? BMC Biophys. 2018;11(1):1. DOI:

10.1186/s13628-017-0041-x PMID: 29308185

48. Zupancic T, Sersa G, Törmä H, Lane EB, Herrmann H, Komel R, et al. Keratin gene mutations influence the keratinocyte response to DNA damage and cytokine induced apoptosis. Arch Dermatol Res.

2017;309(7):587-93. DOI: 10.1007/s00403-017-1757-9 PMID: 28647894

49. Kolundzic N, Khurana P, Hobbs C, Rogar M, Ropret S, Törmä H, et al. Induced pluripotent stem cell (iPSC) line from an epidermolysis bullosa simplex patient heterozygous for keratin 5 E475G mutation and with the Dowling Meara phenotype. Stem Cell Res (Amst). 2019;37:101424. DOI: 10.1016/j.scr.2019.101424 PMID:

30933721

Reference

POVEZANI DOKUMENTI

The stress induced by the electroporation and/or cryopreservation increased the expression of the SOD2 and HSPA1A genes; however, expression of the main immunosuppressive genes IDO1

We used CHO cells embedded in 3-D collagen gels to study electropermeabilization with uptake of propidium iodide (PI) and the effect of different plasmid concentrations and

The use of contact electrodes after drug or DNA intradermal injection were validated by clinical treatment of large surface skin tumors and by in vivo imaging of permeabilization or

The regression of tumor growth after electrochemotherapy, proven also in clinical environments, and a suc- cessful gene electrotransfer to skin cells had shown that deeper

After considering a number of two-, three- and four- parameter models, the delayed exponential model was found to offer better ®t and handling of missing wound area data over time

Regarding the time span of experiences of sound therapies with gongs, the results testify that the agreement with statements on beneficent effect of sound baths on the desire

In this way it is socially

This research, which covered 1400 respondents from a target group of young people, aged between 15 and 29, begins by providing answers to questions about the extent to which