Skip to main content
Erschienen in: Molecular Diagnosis & Therapy 3/2020

Open Access 01.06.2020 | Review Article

Molecular Therapeutics in Development for Epidermolysis Bullosa: Update 2020

verfasst von: Cristina Has, Andrew South, Jouni Uitto

Erschienen in: Molecular Diagnosis & Therapy | Ausgabe 3/2020

Abstract

Epidermolysis bullosa (EB) is a group of rare genetic disorders for which significant progress has been achieved in the development of molecular therapies in the last few decades. Such therapies require knowledge of mutant genes and specific mutations, some of them being allele specific. A relatively large number of clinical trials are ongoing and ascertaining the clinical efficacy of gene, protein or cell therapies or of repurposed drugs, mainly in recessive dystrophic EB. It is expected that some new drugs may emerge in the near future and that combinations of different approaches may result in improved treatment outcomes for individuals with EB.
Hinweise
Cristina Has and Andrew South contributed equally.
Key Points
Remarkable progress has been made in understanding the molecular genetics and underlying pathomechanisms of epidermolysis bullosa (EB) forming the platform for development of treatments.
Gene-replacement approaches, particularly delivery of COL7A1 to the skin of patients with severe dystrophic EB, type VII collagen replacement, skipping of exons and read-through of premature termination codons are currently in clinical trials.
Preclinical research explores the applicability of new strategies in regenerative medicine (e.g., induced pluripotent stem cells) and genome editing (e.g., CRISPR/Cas9).
Particular effort is focused on severe dystrophic EB, characterized by extensive scarring and aggressive squamous cell carcinomas. Small molecules repurposed to reduce fibrosis, and the multikinase inhibitor rigosertib—for the treatment of recessive dystrophic EB squamous cell carcinomas—are being tested in clinical trials.

1 Introduction

Epidermolysis bullosa (EB) comprises a group of genetic disorders characterized by fragility of the skin and mucosal membranes. The molecular basis involves pathogenic variants in genes encoding structural proteins of the dermal–epidermal junction zone (DEJZ) [1]. As a consequence of missing or dysfunctional molecules (e.g., keratins 5/14, integrin α6β4, type XVII and VII collagens), reduced epidermal–dermal cohesion results in blisters after minimal mechanical trauma. The clinical severity of EB covers a broad spectrum, ranging from minor skin or nail involvement and minimal disease burden in localized subtypes to early lethality or life-long progressive systemic disease in severe subtypes [2].
EB is a prototypic disorder for which molecular therapies have been under development in the last few decades. Significant progress has been achieved in understanding the molecular pathogenesis of EB and the potential benefits and limitations of different therapeutic approaches [3]. Considering that EB is a rare disease, a relatively large number of clinical trials are ongoing and ascertaining the clinical efficacy of gene, protein or cell therapies or of repurposed drugs (Table 1). In parallel, preclinical research explores the applicability of new strategies in regenerative medicine (e.g., induced pluripotent stem cells [iPSCs]) and genome editing (e.g., CRISPR/Cas9) (Table 2). However, the initial hope of rapid translation from bench to bedside has been tempered by multiple hurdles and challenges, including the complexity of EB itself. Thus, instead of attempting to cure EB, researchers are increasingly aiming at “symptom-relieving” or “disease-modifying” therapies.
Table 1
Gene-replacement therapies for epidermolysis bullosa in clinical trials
Gene therapy trial
EB type/protein
Approach
Participants (N)
Results
References
Phase I
JEB/laminin β3 chain
Ex vivo genetically corrected (retroviral) autologous epidermal grafts
2
One 7-year-old child treated in wounds covering 80% of the total body surface resulted in regeneration of entire epidermis by transgenic stem cells stable over several years. One 49-year-old woman was successfully treated on an 80 cm2 chronic wound
[35, 36]
Phase I/II; NCT03490331
(HOLOGENE17)
JEB/C17
Ex vivo grafting of gene‐corrected epidermal sheets with a gamma‐retroviral vector carrying COL17A1 cDNA
12
Ongoing
[91]
Phase I/II; NCT02984085
(HOLOGENE7)
RDEB/C7
Ex vivo grafting of gene‐corrected epidermal sheets with a gamma‐retroviral vector carrying COL7A1 cDNA
12
Ongoing
[91]
Phase I; safety and wound outcomes; single center
RDEB/C7
Ex vivo genetically corrected (retroviral) autologous epidermal grafts of 35 cm2
4
Variable response of wound healing and C7; generally declined over 1 year
[30]
Phase I/IIa; single center
RDEB/C7
Ex vivo genetically corrected (retroviral) autologous epidermal grafts of 35 cm2
7
C7 expression persisted up to 2 years after treatment in two participants. Treated wounds with ≥ 50% healing demonstrated improvement in patient-reported pain, itch, and wound durability
[29]
Phase I; single center
RDEB/C7
Three intradermal injections (~ 1 × 106 cells/cm2 of intact skin) of COL7A1-modified (lentiviral) autologous fibroblasts
4
C7 restoration in vivo in treated skin at 1 year after gene therapy
[92]
Phase I/II, phase III; NCT04213261 (Castle Creek Biosciences, Inc.)
RDEB/C7
COL7A1-corrected autologous fibroblasts injected in the wounds
20
Ongoing
Phase III; NCT04227106 (Abeona Therapeutics Inc.)
RDEB/C7
Transplantation of ex vivo COL7A1-corrected autologous keratinocyte sheets
15
Ongoing
Phase I/II; European (GENEGRAFT)
RDEB/C7
Skin-equivalent grafts ex vivo genetically corrected with a COL7A1-encoding SIN retroviral vector
4
Ongoing
[33]
Phase I/II; NCT03536143 (Krystall Biotech, Inc.)
RDEB/C7
Topically administered, replication-deficient HSV-1 vector containing two functional COL7A1 genes applied directly to wounds
6
Ongoing
Phase I (Amryt Pharma, PLC)
RDEB/C7
Topically administered synthetic polymer polyplexes containing COL7A1, applied directly to wounds
NA
NA
cDNA complementary DNA, C7 type VII collagen, C17 type XVII collagen, EB epidermolysis bullosa, HSV-1 herpes simplex virus type 1, JEB junctional EB, NA not available, RDEB recessive dystrophic EB, SIN self-inactivating
Table 2
Overview of recently published CRISPR/Cas9- and RNA-based molecular therapies in preclinical development
Molecular therapy approach
EB type/gene
Correction type, targeted mutation and cells
References
Genomic editing using the CRISPR/Cas9 nuclease system
RDEB/COL7A1
COL7A1 overexpression
[93]
Correction of a mutation in exon 2
[41]
Correction of the mutation c.4317delC and generation of iPSC
[94]
Correction of mutations in exon 19 (c.2470insG) and exon 32 (c.3948insT) through homology-directed repair in iPSC
[42]
Gene reframing therapy to a recurrent frameshift mutation, c.5819delC
[43]
Correction of the mutation c.8068_8084delinsGA
[95]
Cas9/sgRNA ribonucleoproteins to excise exon 80 in skin stem cells of recessive dystrophic EB mice
[96]
Targeted deletion of mutation-bearing COL7A1 exon 80 in RDEB patient keratinocytes
[39]
Correction of a frequent inherited mutation in exon 80
[40]
JEB/LAMB3
In situ correction of LAMB3 gene in keratinocytes
[97]
EBS/KRT14
Correction of the hotspot missense mutation c.1231G > A in keratinocytes
[26]
RNA-based therapies
RDEB/COL7A1
Trans-splicing to correct mutations in the 3′ region
[98]
SIN lentiviral vector at 3′ RNA trans-splicing molecule, capable of replacing exons 65–118
[99]
2′-O-methyl ASO for skipping exons 73 and 80
[58]
ASO for in-frame exon 105 skipping
[59]
ASO for in-frame exon 13 and 105 skipping
[60]
EBS/KRT14
Therapeutic RNA trans-splicing molecule containing wild-type exons 1–7
[25]
ASO antisense oligonucleotides, EB epidermolysis bullosa, EBS EB simplex, iPSC induced pluripotent stem cells, JEB junctional EB, RDEB recessive dystrophic EB, SIN self-inactivating

2 Molecular Pathology of Epidermolysis Bullosa (EB)

Pathogenic variants in 16 genes cause the four main subtypes of classical EB: EB simplex (EBS), junctional EB (JEB), dystrophic EB (DEB) and Kindler EB; over 30 EB subtypes are further defined based on clinical and molecular criteria [2]. EBS and JEB are genetically heterogeneous, whereas DEB and Kindler EB are caused by mutations in single genes, COL7A1 and FERMT1, respectively. In addition to the classical forms of EB, five additional genes have been associated with skin fragility disorders in differential diagnosis of EB. Thus, a total of 21 genes are known to harbor mutations in skin fragility disorders in the spectrum of EB.
The determinants of the EB phenotype include the identity of the affected gene/protein system and the specific nature of the disease-causing genetic variants. Specifically, residual amounts or functions of the affected protein versus its complete absence or loss-of-function determine whether the disease will be relatively mild, intermediate or severe. Examples of genotype–phenotype correlations in patients with JEB and DEB have shown that small amounts (even less than 10%) of proteins with partial function may result in a mild/intermediate phenotype [47]. Genetic and epigenetic disease modifiers may also play a modulating role but have only been experimentally demonstrated in a few cases [812], and such findings have to be extrapolated to larger numbers of patients to allow general conclusions. Socio-economic environment and access to medical care are also critical elements in determining the natural history of the disease and the development of complications in individual cases.
Although multiple different proteins are affected, it is widely accepted that all EB types have life-long skin fragility in common and this pathology (disruption of the barrier function of the skin and mucous membranes) leads to chronic tissue damage and associated inflammation. Loss of epidermal integrity is accompanied by bacterial colonization and activation of mechanisms of innate and adaptive immunity. The cytokines engaged in the tissue damage/repair processes depend on the extent of the mucocutaneous defects and on the level of blister formation (implying either cytolysis or basement membrane zone disruption), and include interleukin (IL)-1, IL-6 and transforming growth factor (TGF)-β [1315]. With time, the ongoing regeneration processes affect the stem cells and the underlying connective tissue, leading to chronic, non-healing wounds. If these events affect a significant percentage of the body surface (such as more than 20–30% at any given time) over a long period, the “inflammation” becomes systemic, as shown by leukocytosis, increased C reactive protein and increased levels of immunoglobulins (IgG, IgM and IgA) [16]. In recessive DEB (RDEB), involvement of the oral and esophageal mucosa impairs feeding, and—together with the high energetic requirements in the context of permanent wound healing—will lead to impaired growth and low body weight. The EB-associated symptoms—pain and pruritus—are mainly related to mucocutaneous blistering and wounding. A long-term complication of chronic tissue damage is carcinogenesis, which is reflected by high risk for squamous cell carcinomas (SCCs) at a young age in patients with RDEB [17] and Kindler EB [18, 19]. Based on these considerations, the main rationale for therapy should be the correction of the skin adhesion defect as this is traditionally accepted as the main origin of pathogenic events in EB.

3 Precision Medicine for EB: From Genetic Defect to Therapeutic Approach

In EB, knowledge of the precise underlying genetic defect is the prerequisite for any molecular therapy approach. The affected gene or protein and the type of pathogenic variants and their consequences dictate which gene or protein should be replaced or edited (e.g., gene or protein therapy). Patients with premature termination codon (PTC) mutations in any EB gene may benefit from read-through therapies [2023]. Mutations in the type VII (COL7A1) and XVII (COL17A1) collagen genes, in which the majority of the exons are in frame, may be approached by skipping of the exons containing the mutations [24]. In the autosomal-dominant forms of EBS and in dominant DEB, the vast majority of mutations lead to amino acid substitutions that disturb the formation and the stability of the keratin intermediate filaments or anchoring fibrils, respectively, through dominant-negative interference of the wild-type allele. For such mutations, potential strategies include knock-down/out of the mutated allele (e.g., by small interfering RNA, RNA trans-splicing or CRISPR/Cas9) [25, 26] or prevention of misfolding (e.g., with chaperones). More palliative (and less precise) approaches include the so-called symptom-relieving therapies, which are directed against inflammatory or other perturbed pathways (e.g., IL-1β, TGFβ). Generation of dedicated databases of EB-associated pathogenic variants comprising their biological characterization, such as consequences at splicing, protein and cellular levels as well as allele frequencies, would be of benefit for researchers and physicians dealing with EB and would assist in setting priorities for specific strategies and in stratification of patients for therapies.

4 Overview of Molecular Therapies in Development for EB

Various molecular therapies are currently in preclinical or clinical development, and most of these focus on RDEB. An overview of the recent advances in those currently under development follows.

4.1 Gene-Replacement Therapies

Many laboratories, in both academia and the pharmaceutical sector, have focused on gene-replacement approaches, particularly delivery of COL7A1 to the skin of patients with RDEB (Table 1). For details on methods, hurdles and risks, we refer to recent review articles [27, 28]. Some studies have tested the efficacy of topical application of an expression vector harboring full-length COL7A1 complementary DNA (cDNA), which would then allow expression of the proα1(VII) polypeptides in the skin, followed by their incorporation into trimeric type VII collagen molecules and supramolecular assembly into functional anchoring fibrils.
In one such approach (Krystal Biotech, Inc.), the cDNA is packaged into a herpes simplex virus (HSV) delivery construct with epidermotropism. In this case, the HSV virus, which in its natural form is highly antigenic, has been modified to manifest with reduced immunogenicity, which would prevent development of immunological complications, such as formation of antibodies, which would eliminate the viral vector and preclude multiple applications (NCT03536143). Another topical application (Amryt Pharma, PLC) utilizes a recently developed non-viral carrier, a highly branched poly(β-ester) polymer, that allows delivery of the COL7A1 cDNA into the skin. In both cases, the transgene does not integrate into the recipient’s genome, and continuous—perhaps life-long—application is required to achieve sustained benefits from the treatment. Current early clinical trials are exploring the frequency of application required for maintenance of efficacy and are examining the levels of expression and turnover time of type VII collagen and its potential assembly into anchoring fibrils.
Another approach for delivery of COL7A1 into patients with RDEB entails introduction of the transgene into the patient’s own cells in cultures, with subsequent delivery of the corrected cells back into the skin. One such approach (Castle Creek Biosciences, Inc.) corrects autologous fibroblasts in culture with a vector that leads to integration of the transgene into the genome, followed by direct injection of the corrected cells to the edges of the wounds. This approach necessitates multiple injections, which can be painful, to the eroded areas of skin, and how long the corrected resident fibroblasts are present and remain active in situ is unclear. Another company (Abeona Therapeutics, Inc.) cultures autologous keratinocytes from the skin of patients with RDEB, followed by genetic correction ex vivo and development of epidermal sheaths, which can then be grafted to the denuded areas of the patient’s skin [29, 30]. These studies have utilized a lentiviral vector that allows incorporation of the COL7A1 cDNA into the genome of the recipient cells. Preliminary studies have revealed that the gene-corrected keratinocytes in the graft are capable of expressing type VII collagen, and there is evidence of assembly of anchoring fibrils. Of some concern is the durability of this approach since the early published data suggested that the expression of the collagen gene may fade over time, possibly attesting to the fact that the transgene is driven by a viral promoter or that the number of stem cells initially isolated and targeted were too low to sustain expression or possibly the graft [31]. Furthermore, in some cases, the graft fails after a certain time, potentially necessitating regrafting of the area. In this context, a recent study demonstrated that correction of both keratinocytes and fibroblasts (cell types that in normal skin both synthesize type VII collagen) is required for optimal assembly of the anchoring fibrils [32]. In this study, skin grafts were made by combining gene-corrected keratinocytes with type VII collagen-deficient fibroblasts, or, conversely, type VII collagen-deficient keratinocytes in combination with gene-corrected fibroblasts, or both cell types being gene corrected. All these three combinations of cells in skin-equivalent explant culture ex vivo expressed type VII collagen, but only the grafts in which both keratinocytes and fibroblasts were gene corrected showed assembly of functional anchoring fibrils. In keeping with these data, there is an ongoing effort to develop skin grafts (GENEGRAFT) that combine keratinocytes and fibroblasts in which both cell types have been corrected with a self-inactivating viral vector expressing type VII collagen [33].
Epidermal grafts have been shown to be highly successful in correcting the underlying defect in patients with intermediate or localized JEB with defects in one of the laminin 332 genes, LAMB3 [3335]. The difference with these studies and those published on grafting in patients with RDEB is that the grafts for patients with JEB were made of a holoclone stem cell population, thus ensuring the longevity of the cells after transplantation. In fact, in one patient with JEB, the functionality of the skin grafts was retained several years after the transplantation [36]. The difference in the outcome between the epidermal cells corrected for transplantation into patients with RDEB and JEB may also reside in the observation that, in normal skin, the laminin 332 polypeptide subunit genes, LAMA3, LAMB3 and LAMC2, provide a selective advantage for retaining stem cells in vivo [37, 38].

4.2 Gene Editing

In addition to gene-replacement approaches, a number of studies have attempted gene repair, such as correction of the mutation by CRISPR/Cas9 editing technology [3943] (Table 2). For details on gene-editing strategies and their appropriate selection depending on mutation, gene and disease type, we refer to a recent overview article [44]. Improvements in this technology have resulted in a high yield of gene correction in keratinocytes or fibroblasts derived from patients with RDEB, with subsequent development of skin grafts that have been transplanted into immunocompromised mice, demonstrating their capability of functional type VII collagen synthesis. One of the potential limitations of this approach is the requirement for a large number of cultured cells for graft production, and an innovative way to circumvent this limitation would be the development of iPSCs that, following the gene correction, can be differentiated into either keratinocytes or fibroblasts [42, 45].

4.3 Natural Gene Therapy

An intriguing possibility for cell-based therapy in EB is provided by revertant mosaicism, a phenomenon described as “natural gene therapy,” in which a number of skin cells undergo spontaneous reversal of the mutation to wild-type genotype, resulting in areas of normal skin [46, 47]. The mechanisms of the mutation reversion are multiple, including mitotic recombinations, back mutations and second site mutations [4850].
Revertant mosaicism in patients with RDEB has been primarily documented in keratinocytes, but evidence of revertant mosaic fibroblasts has also recently been reported [51]. Attempts to establish long-term cultures of keratinocytes from areas of skin with revertant mosaicism have been mostly unsuccessful, primarily due to depletion of stem cells in the affected skin. However, punch grafting of revertant skin to isolated lesions in patients with laminin 332-deficient JEB has been reported [52]. Transplantation of the biopsy specimens resulted in re-epithelialization of the wounds, but punch grafting allowed only a limited expansion of the revertant skin [53]. However, expansion of the treated area can be achieved by generation of iPSCs from revertant keratinocytes, and these cells can then be differentiated into genetically corrected keratinocytes [45]. These revertant iPSCs can be used to create three-dimensional skin equivalents ex vivo and reconstitute human skin in vivo, developing cell-based therapeutic approaches for EB.

4.4 Exon Skipping

Exon skipping makes use of antisense oligonucleotides (ASOs) to modify the splicing of pre-messenger RNA (mRNA) and consequently eliminate the mutation responsible for a disease. Antisense-mediated exon skipping was initially developed for the treatment of Duchenne muscular dystrophy (DMD) and evolved into clinical trials to target DMD (dystrophin gene) exons with recurrent mutations, such as exons 45, 53 and 51. Eteplirsen (Exondys 51) is the first approved antisense therapy for DMD in the USA and provides a treatment option for the ~ 14% of patients with DMD who are amenable to exon 51 skipping [54].
The rationale for using exon skipping in EB comes from studies of genotype–phenotype correlations showing that exon skipping can ameliorate the severity of JEB and DEB [4, 5557] and from the fact that most COL17A1 and COL7A1 exons are in frame. Several preclinical studies have demonstrated that skipping of exons 13, 70, 73, 80 or 105 in COL7A1 results in slightly shortened, partially functional protein, which is deposited at the DEJZ [5861] (Table 2). A clinical trial testing topical administration of QR-313, a water-based gel (hydrogel) containing the ASO targeting exon 73 that will be applied directly onto DEB wounds for COL7A1 correction is currently ongoing (Wings Therapeutics; NCT03605069).

4.5 Protein Therapy

Preclinical studies have shown that recombinant type VII collagen, injected locally or intravenously, homes to the DEJZ and promotes wound healing [62, 63]. Recombinant type VII collagen has obtained fast drug designation from the US FDA, and a phase I/II clinical trial evaluating its safety and tolerability, as well as clinical proof of concept in adults with RDEB, is ongoing in the USA (Phoenix Tissue Repair, Inc.; NCT03752905). The advantages of systemically administered recombinant type VII collagen include the possibility that the protein, in addition to homing into skin, will reach extracutaneous tissues affected by RDEB, such as the gastrointestinal track and the cornea of the eye, with subsequent repair.
Beside this protein-replacement approach, a therapy with a high mobility group box 1 (HMGB1) fragment is under development (redasemtide trifluoroacetate phase II, Shinogi & Co., Ltd.). HMGB1 can mobilize the Lin(−)/PDGFRα(+) cells from bone marrow to damaged tissue and facilitate tissue repair [64], probably by suppressing the inflammation of injured skin [65]. In animal models, systemic administration of the HMGB1 fragment has shown benefits, preventing deterioration of cardiac performance in the delta-sarcoglycan-deficient hamster [66] and ameliorating cutaneous and non-cutaneous manifestations in a dystrophic EB model mouse [67].

4.6 Read-Through Therapies

The read-through approach involves the use of small-molecular-weight compounds that allow the translational machinery to suppress nonsense mutations by incorporating an amino acid in place of a stop codon and results in synthesis of full-length protein.
The original prototype of such read-through molecules, PTC124, was shown to read-through only pathogenic PTCs but not through naturally occurring endogenous stop codons of translation because the nucleic acid context and the intron–exon organization of the gene provide a very robust and strong stop signal. This read-through molecule has been tested on a number of genes, principally the dystrophin gene in DMD [68], but some studies have failed to identify PTC124 as being able to efficiently read-through COL7A1 PTCs as compared with the prototypic PTC read-through drug, the aminoglycoside gentamicin [20, 23, 69]. Work has shown that gentamicin can effectively read-through COL7A1 and LAMB3 PTCs [20, 21], and clinical studies in RDEB have shown promising results [22]. Long-term systemic treatment with this antibiotic has a number of potential issues, including the risk of renal toxicity [70], and topical application might be a favorable delivery route in this context. Nevertheless, clinical studies testing the effect of intravenous injections of gentamicin are being conducted for RDEB (NCT03392909) and JEB (NCT03526159).
Another compound recently shown to induce read-through of COL7A1 is amlexanox [71], a drug with a number of different activities and targets currently in trial for a wide range of indications. This drug could potentially be used long term because of its very favorable toxicity profile and clinical history. However, the current manufacturer of amlexanox, Takeda Pharmaceutical, has removed it from the market, and an alternative supply will be needed to pursue clinical application. One interesting observation from the amlexanox study in EB was that it enhanced read-through only in cells with a low level of endogenous full-length protein, whereas cells with undetectable protein did not respond [23]. These data suggest that certain cellular conditions required for read-through need to be identified and exploited for more efficient and effective approaches.

4.7 Small Molecules Repurposed to Relieve Symptoms

Fibrosis is a major pathological complication of RDEB, and work with patient cells in culture, animal models and patients have all identified TGFβ signaling as a major driver of fibrosis and disease severity [9, 7275]. TGFβ is a primary mediator of fibrosis driving extracellular matrix (ECM) deposition in numerous pathological situations, and considerable effort has focused on understanding and inhibiting TGFβ in a number of contexts [76]. However, as TGFβ receptors participate in a signaling pathways that control many aspects of mammalian development and tissue homeostasis, global inhibition has proven problematic [77]. Indirect targeting of molecules that activate or inhibit the pathway in specific contexts will likely have improved efficacy compared with global inhibition. The first example of such an approach in RDEB is the use of losartan, a drug approved for the treatment of high blood pressure. Work in animal models identified a significant reduction of fibrosis in the paws of hypomorphic mice with losartan treatment, and this drug is now in a clinical trial (EudraCT number: 2015-003670-32) [78]. The mechanism here is thought to be a reduction in blood pressure, which reduces the bioavailability or release of TGFβ ligand from the fibrotic ECM. Other preclinical efforts have used viral delivery of decorin, an inhibitor of TGFβ, to show reduced paw fibrosis in mouse models [79], whereas in vitro work with patient cells has suggested that inhibition of thrombospondin-1, a potent activator of TGFβ, may have clinical efficacy [72]. In addition to these academic-led initiatives, a number of anti-TGFβ antibodies are in development in the commercial space that have potential to reach clinical trial in the coming years.

5 Therapies for EB-Associated Squamous Cell Carcinoma

Historically, only a few reports have documented any durable therapeutic response of SCCs in patients with RDEB, and the observations that reported short-term responses were not followed-up in the literature [80]. However, additional case studies and reviews of prior literature are beginning to emerge, and the main focus has been on cetuximab, the epidermal growth factor receptor inhibitor approved for head and neck SCC (HNSCC) [81]. A number of patients with RDEB have been treated, and progression-free survival has been reported to range between 3 and 9 months, but wound-healing deficits are often noted, and eventually the patients may succumb to their disease [8286]. With the recent approval of the immune checkpoint inhibitors nivolumab, pembrolizumab and cemiplimab, the treatment landscape of HNSCC and cutaneous SCC is changing, and long-term progression-free survival in a small proportion of patients with spontaneous SCC is becoming evident [87, 88]. For two patients with RDEB SCC treated with nivolumab, the results were negative, with no evidence of durable response [82, 86]. However, two patients are too few to rule out potential benefits, considering that only ~ 20% of patients with HNSCC show a response to this therapeutic approach. One phase I clinical trial testing the toxicity of isotretinoin in RDEB SCC has been reported [89]. Although the drug was well-tolerated by 20 patients, no further studies were conducted. Preclinical work identified the potential of the multikinase inhibitor rigosertib, an experimental therapeutic in phase III trials for treatment of myelodysplastic syndrome and RDEB SCC [90]. In cell culture and animal studies, rigosertib induced apoptosis in RDEB SCC without adversely affecting non-SCC RDEB epidermal keratinocytes. As a result, two trials, one in the USA (NCT04177498) and one in the UK and Austria (NCT03786237), have been initiated.

6 Conclusions and Future Perspectives

Tremendous progress has been made in understanding the molecular genetics and underlying pathomechanisms of different forms of EB, and the increase in knowledge about these disorders has formed the basis for treatment development. Several laboratories in academia, a number of companies and biotechnology and the “big pharma” have devoted a considerable amount of effort and resources to develop treatments and cures for EB. Such approaches include gene replacement and repair, protein replacement and cell-based therapies, and some are allele specific, requiring knowledge of mutant genes and specific mutations. A number of these strategies are in early clinical trials, and at least three are entering phase III for clinical testing in 2020. Considering the multiplicity of approaches, there is cautious optimism that some drugs may emerge in the near future as potential candidates for treatment of EB. It has also been suggested that, because of the complementary nature of these approaches, application of different drugs in combination may result in optimal treatment outcomes for an individual with EB [100].
Despite significant progress and relatively numerous ongoing clinical trials, the number of patients with EB who currently have access to molecular therapies is limited (Table 1). Current development is also primarily focused on DEB and JEB, whereas EBS, the most common EB type, has been largely neglected even though affected individuals are severely impaired in everyday life and professional activities. Furthermore, there are several questions regarding the cost of development and delivery of such pharmaceuticals and global access to these drugs once they are available in the market.
Regenerative medicine and replacement of the deficient genes and proteins are the only way to “cure” EB. Nevertheless, “adjuvant” therapies with less expensive repurposed drugs may assist in reducing disease severity and improving the quality of life of the affected individuals. In fact, such therapies may be required to complement strategies aimed at alleviating the fragility of the skin and mucous membranes as the primary pathologic process. Also, proving the efficacy of such measures seems challenging, and designing clinical trials with better endpoints and sufficient numbers of participants requires long-term expertise and international collaboration. Nevertheless, patients with EB and their families are looking forward to effective treatment, hopefully before too long.

Acknowledgements

Carol Kelly assisted in manuscript preparation.

Compliance with Ethical Standards

Conflicts of interest

C.H. was on the advisory board of Castle Creek Biosciences Inc. and A.P.S. owns stock in Krystal Biotech, Inc. J.U. has no conflicts of interest. That are Is Castle Creek Pharmaceuticals the same as the ‚Castle Creek Biosciences‘ mentioned in Table 1 and Sect. 4.1? If so, which name should be used for consistency? directly relevant to the content of this article.
Open AccessThis article is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License, which permits any non-commercial use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by-nc/​4.​0/​.
Literatur
1.
Zurück zum Zitat Fine J-D, Bruckner-Tuderman L, Eady RAJ, Bauer EA, Bauer JW, Has C, et al. Inherited epidermolysis bullosa: updated recommendations on diagnosis and classification. J Am Acad Dermatol. 2014;70:1103–26.PubMedCrossRef Fine J-D, Bruckner-Tuderman L, Eady RAJ, Bauer EA, Bauer JW, Has C, et al. Inherited epidermolysis bullosa: updated recommendations on diagnosis and classification. J Am Acad Dermatol. 2014;70:1103–26.PubMedCrossRef
2.
Zurück zum Zitat Has C, Bauer JW, Bodemer C, Bolling M, Bruckner-Tuderman L, Diem A, et al. Consensus re-classification of inherited epidermolysis bullosa and other disorders with skin fragility. Br J Dermatol. 2020. Has C, Bauer JW, Bodemer C, Bolling M, Bruckner-Tuderman L, Diem A, et al. Consensus re-classification of inherited epidermolysis bullosa and other disorders with skin fragility. Br J Dermatol. 2020.
3.
Zurück zum Zitat Uitto J, Bruckner-Tuderman L, McGrath JA, Riedl R, Robinson C. EB2017-progress in epidermolysis bullosa research toward treatment and cure. J Invest Dermatol. 2018;138:1010–6.PubMedCrossRef Uitto J, Bruckner-Tuderman L, McGrath JA, Riedl R, Robinson C. EB2017-progress in epidermolysis bullosa research toward treatment and cure. J Invest Dermatol. 2018;138:1010–6.PubMedCrossRef
4.
Zurück zum Zitat Schwieger-Briel A, Weibel L, Chmel N, Leppert J, Kernland-Lang K, Grüninger G, et al. A COL7A1 variant leading to in-frame skipping of exon 15 attenuates disease severity in recessive dystrophic epidermolysis bullosa. Br J Dermatol. 2015;173:1308–11.PubMedCrossRef Schwieger-Briel A, Weibel L, Chmel N, Leppert J, Kernland-Lang K, Grüninger G, et al. A COL7A1 variant leading to in-frame skipping of exon 15 attenuates disease severity in recessive dystrophic epidermolysis bullosa. Br J Dermatol. 2015;173:1308–11.PubMedCrossRef
5.
Zurück zum Zitat Kiritsi D, Kern JS, Schumann H, Kohlhase J, Has C, Bruckner-Tuderman L. Molecular mechanisms of phenotypic variability in junctional epidermolysis bullosa. J Med Genet. 2011;48:450–7.PubMedCrossRef Kiritsi D, Kern JS, Schumann H, Kohlhase J, Has C, Bruckner-Tuderman L. Molecular mechanisms of phenotypic variability in junctional epidermolysis bullosa. J Med Genet. 2011;48:450–7.PubMedCrossRef
6.
Zurück zum Zitat Mellerio JE, Pulkkinen L, McMillan JR, Lake BD, Horn HM, Tidman MJ, et al. Pyloric atresia-junctional epidermolysis bullosa syndrome: mutations in the integrin beta4 gene (ITGB4) in two unrelated patients with mild disease. Br J Dermatol. 1998;139:862–71.PubMedCrossRef Mellerio JE, Pulkkinen L, McMillan JR, Lake BD, Horn HM, Tidman MJ, et al. Pyloric atresia-junctional epidermolysis bullosa syndrome: mutations in the integrin beta4 gene (ITGB4) in two unrelated patients with mild disease. Br J Dermatol. 1998;139:862–71.PubMedCrossRef
7.
Zurück zum Zitat Hoffmann J, Casetti F, Reimer A, Leppert J, Grüninger G, Has C. A silent COL17A1 variant alters splicing and causes junctional epidermolysis bullosa. Acta Derm Venereol. 2019;99:460–1.PubMedCrossRef Hoffmann J, Casetti F, Reimer A, Leppert J, Grüninger G, Has C. A silent COL17A1 variant alters splicing and causes junctional epidermolysis bullosa. Acta Derm Venereol. 2019;99:460–1.PubMedCrossRef
8.
Zurück zum Zitat Titeux M, Pendaries V, Tonasso L, Decha A, Bodemer C, Hovnanian A. A frequent functional SNP in the MMP1 promoter is associated with higher disease severity in recessive dystrophic epidermolysis bullosa. Hum Mutat. 2008;29:267–76.PubMedCrossRef Titeux M, Pendaries V, Tonasso L, Decha A, Bodemer C, Hovnanian A. A frequent functional SNP in the MMP1 promoter is associated with higher disease severity in recessive dystrophic epidermolysis bullosa. Hum Mutat. 2008;29:267–76.PubMedCrossRef
9.
Zurück zum Zitat Odorisio T, Di Salvio M, Orecchia A, Di Zenzo G, Piccinni E, Cianfarani F, et al. Monozygotic twins discordant for recessive dystrophic epidermolysis bullosa phenotype highlight the role of TGF-β signalling in modifying disease severity. Hum Mol Genet. 2014;23:3907–22.PubMedCrossRef Odorisio T, Di Salvio M, Orecchia A, Di Zenzo G, Piccinni E, Cianfarani F, et al. Monozygotic twins discordant for recessive dystrophic epidermolysis bullosa phenotype highlight the role of TGF-β signalling in modifying disease severity. Hum Mol Genet. 2014;23:3907–22.PubMedCrossRef
10.
Zurück zum Zitat Vahidnezhad H, Youssefian L, Saeidian AH, Touati A, Sotoudeh S, Jazayeri A, et al. Next generation sequencing identifies double homozygous mutations in two distinct genes (EXPH5 and COL17A1) in a patient with concomitant simplex and junctional epidermolysis bullosa. Hum Mutat. 2018;39:1349–54.PubMedCrossRef Vahidnezhad H, Youssefian L, Saeidian AH, Touati A, Sotoudeh S, Jazayeri A, et al. Next generation sequencing identifies double homozygous mutations in two distinct genes (EXPH5 and COL17A1) in a patient with concomitant simplex and junctional epidermolysis bullosa. Hum Mutat. 2018;39:1349–54.PubMedCrossRef
11.
Zurück zum Zitat Vahidnezhad H, Youssefian L, Sotoudeh S, Liu L, Guy A, Lovell PA, et al. Genomics-based treatment in a patient with two overlapping heritable skin disorders: Epidermolysis bullosa and acrodermatitis enteropathica. Hum Mutat. 2020;41:906–12.PubMedCrossRef Vahidnezhad H, Youssefian L, Sotoudeh S, Liu L, Guy A, Lovell PA, et al. Genomics-based treatment in a patient with two overlapping heritable skin disorders: Epidermolysis bullosa and acrodermatitis enteropathica. Hum Mutat. 2020;41:906–12.PubMedCrossRef
12.
Zurück zum Zitat Maccari ME, Speckmann C, Heeg M, Reimer A, Casetti F, Has C, et al. Profound immunodeficiency with severe skin disease explained by concomitant novel CARMIL2 and PLEC1 loss-of-function mutations. Clin Immunol Orlando Fla. 2019;208:108228.CrossRef Maccari ME, Speckmann C, Heeg M, Reimer A, Casetti F, Has C, et al. Profound immunodeficiency with severe skin disease explained by concomitant novel CARMIL2 and PLEC1 loss-of-function mutations. Clin Immunol Orlando Fla. 2019;208:108228.CrossRef
13.
Zurück zum Zitat Wally V, Lettner T, Peking P, Peckl-Schmid D, Murauer EM, Hainzl S, et al. The pathogenetic role of IL-1β in severe epidermolysis bullosa simplex. J Invest Dermatol. 2013;133:1901–3.PubMedCrossRef Wally V, Lettner T, Peking P, Peckl-Schmid D, Murauer EM, Hainzl S, et al. The pathogenetic role of IL-1β in severe epidermolysis bullosa simplex. J Invest Dermatol. 2013;133:1901–3.PubMedCrossRef
14.
Zurück zum Zitat Chacón-Solano E, León C, Díaz F, García-García F, García M, Escámez MJ, et al. Fibroblasts activation and abnormal extracellular matrix remodelling as common hallmarks in three cancer-prone genodermatoses. Br J Dermatol. 2019;181:512–22.PubMedPubMedCentralCrossRef Chacón-Solano E, León C, Díaz F, García-García F, García M, Escámez MJ, et al. Fibroblasts activation and abnormal extracellular matrix remodelling as common hallmarks in three cancer-prone genodermatoses. Br J Dermatol. 2019;181:512–22.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Annicchiarico G, Morgese MG, Esposito S, Lopalco G, Lattarulo M, Tampoia M, et al. Proinflammatory cytokines and antiskin autoantibodies in patients with inherited epidermolysis bullosa. Medicine (Baltimore). 2015;94:e1528.PubMedPubMedCentralCrossRef Annicchiarico G, Morgese MG, Esposito S, Lopalco G, Lattarulo M, Tampoia M, et al. Proinflammatory cytokines and antiskin autoantibodies in patients with inherited epidermolysis bullosa. Medicine (Baltimore). 2015;94:e1528.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Reimer A, Hess M, Schwieger-Briel A, Kiritsi D, Schauer F, Schumann H, et al. Natural history of growth and anaemia in children with epidermolysis bullosa: a retrospective cohort study. Br J Dermatol. 2019. Reimer A, Hess M, Schwieger-Briel A, Kiritsi D, Schauer F, Schumann H, et al. Natural history of growth and anaemia in children with epidermolysis bullosa: a retrospective cohort study. Br J Dermatol. 2019.
17.
Zurück zum Zitat Fine JD, Johnson LB, Weiner M, Li KP, Suchindran C. Epidermolysis bullosa and the risk of life-threatening cancers: the National EB Registry experience, 1986-2006. J Am Acad Dermatol. 2009;60:203–11.PubMedCrossRef Fine JD, Johnson LB, Weiner M, Li KP, Suchindran C. Epidermolysis bullosa and the risk of life-threatening cancers: the National EB Registry experience, 1986-2006. J Am Acad Dermatol. 2009;60:203–11.PubMedCrossRef
18.
Zurück zum Zitat Has C, Castiglia D, del Rio M, Diez MG, Piccinni E, Kiritsi D, et al. Kindler syndrome: extension of FERMT1 mutational spectrum and natural history. Hum Mutat. 2011;32:1204–12.PubMedCrossRef Has C, Castiglia D, del Rio M, Diez MG, Piccinni E, Kiritsi D, et al. Kindler syndrome: extension of FERMT1 mutational spectrum and natural history. Hum Mutat. 2011;32:1204–12.PubMedCrossRef
19.
Zurück zum Zitat Saleva M, Has C, He Y, Vassileva S, Balabanova M, Miteva L. Natural history of Kindler syndrome and propensity for skin cancer—case report and literature review. J Dtsch Dermatol Ges J Ger Soc Dermatol JDDG. 2018;16:338–41. Saleva M, Has C, He Y, Vassileva S, Balabanova M, Miteva L. Natural history of Kindler syndrome and propensity for skin cancer—case report and literature review. J Dtsch Dermatol Ges J Ger Soc Dermatol JDDG. 2018;16:338–41.
20.
Zurück zum Zitat Cogan J, Weinstein J, Wang X, Hou Y, Martin S, South AP, et al. Aminoglycosides restore full-length type VII collagen by overcoming premature termination codons: therapeutic implications for dystrophic epidermolysis bullosa. Mol Ther J Am Soc Gene Ther. 2014;22:1741–52.CrossRef Cogan J, Weinstein J, Wang X, Hou Y, Martin S, South AP, et al. Aminoglycosides restore full-length type VII collagen by overcoming premature termination codons: therapeutic implications for dystrophic epidermolysis bullosa. Mol Ther J Am Soc Gene Ther. 2014;22:1741–52.CrossRef
21.
Zurück zum Zitat Lincoln V, Cogan J, Hou Y, Hirsch M, Hao M, Alexeev V, et al. Gentamicin induces LAMB3 nonsense mutation readthrough and restores functional laminin 332 in junctional epidermolysis bullosa. Proc Natl Acad Sci USA. 2018;115:E6536–45.PubMedCrossRefPubMedCentral Lincoln V, Cogan J, Hou Y, Hirsch M, Hao M, Alexeev V, et al. Gentamicin induces LAMB3 nonsense mutation readthrough and restores functional laminin 332 in junctional epidermolysis bullosa. Proc Natl Acad Sci USA. 2018;115:E6536–45.PubMedCrossRefPubMedCentral
22.
Zurück zum Zitat Woodley DT, Cogan J, Hou Y, Lyu C, Marinkovich MP, Keene D, et al. Gentamicin induces functional type VII collagen in recessive dystrophic epidermolysis bullosa patients. J Clin Invest. 2017;127:3028–38.PubMedPubMedCentralCrossRef Woodley DT, Cogan J, Hou Y, Lyu C, Marinkovich MP, Keene D, et al. Gentamicin induces functional type VII collagen in recessive dystrophic epidermolysis bullosa patients. J Clin Invest. 2017;127:3028–38.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Atanasova VS, Jiang Q, Prisco M, Gruber C, Piñón Hofbauer J, Chen M, et al. Amlexanox enhances premature termination codon read-through in COL7A1 and expression of full length type VII Collagen: potential therapy for recessive dystrophic epidermolysis bullosa. J Invest Dermatol. 2017;137:1842–9.PubMedPubMedCentralCrossRef Atanasova VS, Jiang Q, Prisco M, Gruber C, Piñón Hofbauer J, Chen M, et al. Amlexanox enhances premature termination codon read-through in COL7A1 and expression of full length type VII Collagen: potential therapy for recessive dystrophic epidermolysis bullosa. J Invest Dermatol. 2017;137:1842–9.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Bornert O, Peking P, Bremer J, Koller U, van den Akker PC, Aartsma-Rus A, et al. RNA-based therapies for genodermatoses. Exp Dermatol. 2017;26:3–10.PubMedPubMedCentralCrossRef Bornert O, Peking P, Bremer J, Koller U, van den Akker PC, Aartsma-Rus A, et al. RNA-based therapies for genodermatoses. Exp Dermatol. 2017;26:3–10.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Peking P, Breitenbach JS, Ablinger M, Muss WH, Poetschke FJ, Kocher T, et al. An ex vivo RNA trans-splicing strategy to correct human generalized severe epidermolysis bullosa simplex. Br J Dermatol. 2019;180:141–8.PubMedCrossRef Peking P, Breitenbach JS, Ablinger M, Muss WH, Poetschke FJ, Kocher T, et al. An ex vivo RNA trans-splicing strategy to correct human generalized severe epidermolysis bullosa simplex. Br J Dermatol. 2019;180:141–8.PubMedCrossRef
26.
Zurück zum Zitat Kocher T, Peking P, Klausegger A, Murauer EM, Hofbauer JP, Wally V, et al. Cut and paste: efficient homology-directed repair of a dominant negative KRT14 mutation via CRISPR/Cas9 nickases. Mol Ther J Am Soc Gene Ther. 2017;25:2585–98.CrossRef Kocher T, Peking P, Klausegger A, Murauer EM, Hofbauer JP, Wally V, et al. Cut and paste: efficient homology-directed repair of a dominant negative KRT14 mutation via CRISPR/Cas9 nickases. Mol Ther J Am Soc Gene Ther. 2017;25:2585–98.CrossRef
27.
Zurück zum Zitat Marinkovich MP, Tang JY. Gene therapy for epidermolysis bullosa. J Invest Dermatol. 2019;139:1221–6.PubMedCrossRef Marinkovich MP, Tang JY. Gene therapy for epidermolysis bullosa. J Invest Dermatol. 2019;139:1221–6.PubMedCrossRef
28.
Zurück zum Zitat Rashidghamat E, McGrath JA. Novel and emerging therapies in the treatment of recessive dystrophic epidermolysis bullosa. Intractable Rare Dis Res. 2017;6:6–20.PubMedPubMedCentralCrossRef Rashidghamat E, McGrath JA. Novel and emerging therapies in the treatment of recessive dystrophic epidermolysis bullosa. Intractable Rare Dis Res. 2017;6:6–20.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Eichstadt S, Barriga M, Ponakala A, Teng C, Nguyen NT, Siprashvili Z, et al. Phase 1/2a clinical trial of gene-corrected autologous cell therapy for recessive dystrophic epidermolysis bullosa. JCI Insight. 2019;4:e130554.PubMedCentralCrossRef Eichstadt S, Barriga M, Ponakala A, Teng C, Nguyen NT, Siprashvili Z, et al. Phase 1/2a clinical trial of gene-corrected autologous cell therapy for recessive dystrophic epidermolysis bullosa. JCI Insight. 2019;4:e130554.PubMedCentralCrossRef
30.
Zurück zum Zitat Siprashvili Z, Nguyen NT, Gorell ES, Loutit K, Khuu P, Furukawa LK, et al. Safety and wound outcomes following genetically corrected autologous epidermal grafts in patients with recessive dystrophic epidermolysis bullosa. JAMA. 2016;316:1808–17.PubMedCrossRef Siprashvili Z, Nguyen NT, Gorell ES, Loutit K, Khuu P, Furukawa LK, et al. Safety and wound outcomes following genetically corrected autologous epidermal grafts in patients with recessive dystrophic epidermolysis bullosa. JAMA. 2016;316:1808–17.PubMedCrossRef
31.
Zurück zum Zitat Kolodka TM, Garlick JA, Taichman LB. Evidence for keratinocyte stem cells in vitro: long term engraftment and persistence of transgene expression from retrovirus-transduced keratinocytes. Proc Natl Acad Sci USA. 1998;95:4356–61.PubMedCrossRefPubMedCentral Kolodka TM, Garlick JA, Taichman LB. Evidence for keratinocyte stem cells in vitro: long term engraftment and persistence of transgene expression from retrovirus-transduced keratinocytes. Proc Natl Acad Sci USA. 1998;95:4356–61.PubMedCrossRefPubMedCentral
32.
Zurück zum Zitat Supp DM, Hahn JM, Combs KA, McFarland KL, Schwentker A, Boissy RE, et al. Collagen VII expression is required in both keratinocytes and fibroblasts for anchoring fibril formation in bilayer engineered skin substitutes. Cell Transpl. 2019;28:1242–56.CrossRef Supp DM, Hahn JM, Combs KA, McFarland KL, Schwentker A, Boissy RE, et al. Collagen VII expression is required in both keratinocytes and fibroblasts for anchoring fibril formation in bilayer engineered skin substitutes. Cell Transpl. 2019;28:1242–56.CrossRef
33.
Zurück zum Zitat Gaucher S, Lwin SM, Titeux M, Abdul-Wahab A, Pironon N, Izmiryan A, et al. EBGene trial: patient preselection outcomes for the European GENEGRAFT ex vivo phase I/II gene therapy trial for recessive dystrophic epidermolysis bullosa. Br J Dermatol. 2020;182:794–7.PubMedCrossRef Gaucher S, Lwin SM, Titeux M, Abdul-Wahab A, Pironon N, Izmiryan A, et al. EBGene trial: patient preselection outcomes for the European GENEGRAFT ex vivo phase I/II gene therapy trial for recessive dystrophic epidermolysis bullosa. Br J Dermatol. 2020;182:794–7.PubMedCrossRef
34.
Zurück zum Zitat 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;12:1397–402.PubMedCrossRef 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;12:1397–402.PubMedCrossRef
35.
Zurück zum Zitat Bauer JW, Koller J, Murauer EM, De Rosa L, Enzo E, Carulli S, et al. Closure of a large chronic wound through transplantation of gene-corrected epidermal stem cells. J Invest Dermatol. 2017;137:778–81.PubMedCrossRef Bauer JW, Koller J, Murauer EM, De Rosa L, Enzo E, Carulli S, et al. Closure of a large chronic wound through transplantation of gene-corrected epidermal stem cells. J Invest Dermatol. 2017;137:778–81.PubMedCrossRef
36.
Zurück zum Zitat 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;551:327–32.PubMedPubMedCentralCrossRef 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;551:327–32.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Liu N, Matsumura H, Kato T, Ichinose S, Takada A, Namiki T, et al. Stem cell competition orchestrates skin homeostasis and ageing. Nature. 2019;568:344–50.PubMedCrossRef Liu N, Matsumura H, Kato T, Ichinose S, Takada A, Namiki T, et al. Stem cell competition orchestrates skin homeostasis and ageing. Nature. 2019;568:344–50.PubMedCrossRef
38.
Zurück zum Zitat De Rosa L, Secone Seconetti A, De Santis G, Pellacani G, Hirsch T, Rothoeft T, et al. Laminin 332-dependent YAP dysregulation depletes epidermal stem cells in junctional epidermolysis bullosa. Cell Rep. 2019;27(2036–2049):e6. De Rosa L, Secone Seconetti A, De Santis G, Pellacani G, Hirsch T, Rothoeft T, et al. Laminin 332-dependent YAP dysregulation depletes epidermal stem cells in junctional epidermolysis bullosa. Cell Rep. 2019;27(2036–2049):e6.
39.
Zurück zum Zitat Bonafont J, Mencía Á, García M, Torres R, Rodríguez S, Carretero M, et al. Clinically relevant correction of recessive dystrophic epidermolysis bullosa by dual sgRNA CRISPR/Cas9-mediated gene editing. Mol Ther J Am Soc Gene Ther. 2019;27:986–98.CrossRef Bonafont J, Mencía Á, García M, Torres R, Rodríguez S, Carretero M, et al. Clinically relevant correction of recessive dystrophic epidermolysis bullosa by dual sgRNA CRISPR/Cas9-mediated gene editing. Mol Ther J Am Soc Gene Ther. 2019;27:986–98.CrossRef
40.
Zurück zum Zitat Hainzl S, Peking P, Kocher T, Murauer EM, Larcher F, Del Rio M, et al. COL7A1 editing via CRISPR/Cas9 in recessive dystrophic epidermolysis bullosa. Mol Ther J Am Soc Gene Ther. 2017;25:2573–84.CrossRef Hainzl S, Peking P, Kocher T, Murauer EM, Larcher F, Del Rio M, et al. COL7A1 editing via CRISPR/Cas9 in recessive dystrophic epidermolysis bullosa. Mol Ther J Am Soc Gene Ther. 2017;25:2573–84.CrossRef
41.
Zurück zum Zitat Izmiryan A, Ganier C, Bovolenta M, Schmitt A, Mavilio F, Hovnanian A. Ex vivo COL7A1 correction for recessive dystrophic epidermolysis bullosa using CRISPR/Cas9 and homology-directed repair. Mol Ther Nucleic Acids. 2018;12:554–67.PubMedPubMedCentralCrossRef Izmiryan A, Ganier C, Bovolenta M, Schmitt A, Mavilio F, Hovnanian A. Ex vivo COL7A1 correction for recessive dystrophic epidermolysis bullosa using CRISPR/Cas9 and homology-directed repair. Mol Ther Nucleic Acids. 2018;12:554–67.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Jacków J, Guo Z, Hansen C, Abaci HE, Doucet YS, Shin JU, et al. CRISPR/Cas9-based targeted genome editing for correction of recessive dystrophic epidermolysis bullosa using iPS cells. Proc Natl Acad Sci USA. 2019;116:26846–52.CrossRefPubMedCentral Jacków J, Guo Z, Hansen C, Abaci HE, Doucet YS, Shin JU, et al. CRISPR/Cas9-based targeted genome editing for correction of recessive dystrophic epidermolysis bullosa using iPS cells. Proc Natl Acad Sci USA. 2019;116:26846–52.CrossRefPubMedCentral
43.
Zurück zum Zitat Takashima S, Shinkuma S, Fujita Y, Nomura T, Ujiie H, Natsuga K, et al. Efficient gene reframing therapy for recessive dystrophic epidermolysis bullosa with CRISPR/Cas9. J Invest Dermatol. 2019;139(1711–1721):e4. Takashima S, Shinkuma S, Fujita Y, Nomura T, Ujiie H, Natsuga K, et al. Efficient gene reframing therapy for recessive dystrophic epidermolysis bullosa with CRISPR/Cas9. J Invest Dermatol. 2019;139(1711–1721):e4.
44.
Zurück zum Zitat March OP, Kocher T, Koller U. Context-dependent strategies for enhanced genome editing of genodermatoses. Cells. 2020;9:112.PubMedCentralCrossRef March OP, Kocher T, Koller U. Context-dependent strategies for enhanced genome editing of genodermatoses. Cells. 2020;9:112.PubMedCentralCrossRef
45.
Zurück zum Zitat Umegaki-Arao N, Pasmooij AMG, Itoh M, Cerise JE, Guo Z, Levy B, et al. Induced pluripotent stem cells from human revertant keratinocytes for the treatment of epidermolysis bullosa. Sci Transl Med. 2014;6:264ra164.PubMedCrossRef Umegaki-Arao N, Pasmooij AMG, Itoh M, Cerise JE, Guo Z, Levy B, et al. Induced pluripotent stem cells from human revertant keratinocytes for the treatment of epidermolysis bullosa. Sci Transl Med. 2014;6:264ra164.PubMedCrossRef
46.
Zurück zum Zitat Jonkman MF, Scheffer H, Stulp R, Pas HH, Nijenhuis M, Heeres K, et al. Revertant mosaicism in epidermolysis bullosa caused by mitotic gene conversion. Cell. 1997;88:543–51.PubMedCrossRef Jonkman MF, Scheffer H, Stulp R, Pas HH, Nijenhuis M, Heeres K, et al. Revertant mosaicism in epidermolysis bullosa caused by mitotic gene conversion. Cell. 1997;88:543–51.PubMedCrossRef
47.
Zurück zum Zitat Pasmooij AM, Jonkman MF, Uitto J. Revertant mosaicism in heritable skin diseases: mechanisms of natural gene therapy. Discov Med. 2012;14:167–79.PubMed Pasmooij AM, Jonkman MF, Uitto J. Revertant mosaicism in heritable skin diseases: mechanisms of natural gene therapy. Discov Med. 2012;14:167–79.PubMed
48.
Zurück zum Zitat Kiritsi D, Garcia M, Brander R, Has C, Meijer R, Jose Escamez M, et al. Mechanisms of natural gene therapy in dystrophic epidermolysis bullosa. J Invest Dermatol. 2014;134:2097–104.PubMedCrossRef Kiritsi D, Garcia M, Brander R, Has C, Meijer R, Jose Escamez M, et al. Mechanisms of natural gene therapy in dystrophic epidermolysis bullosa. J Invest Dermatol. 2014;134:2097–104.PubMedCrossRef
49.
Zurück zum Zitat Pasmooij AM, Pas HH, Bolling MC, Jonkman MF. Revertant mosaicism in junctional epidermolysis bullosa due to multiple correcting second-site mutations in LAMB3. J Clin Invest. 2007;117:1240–8.PubMedPubMedCentralCrossRef Pasmooij AM, Pas HH, Bolling MC, Jonkman MF. Revertant mosaicism in junctional epidermolysis bullosa due to multiple correcting second-site mutations in LAMB3. J Clin Invest. 2007;117:1240–8.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Pasmooij AM, Nijenhuis M, Brander R, Jonkman MF. Natural gene therapy may occur in all patients with generalized non-Herlitz junctional epidermolysis bullosa with COL17A1 mutations. J Invest Dermatol. 2012;132:1374–83.PubMedCrossRef Pasmooij AM, Nijenhuis M, Brander R, Jonkman MF. Natural gene therapy may occur in all patients with generalized non-Herlitz junctional epidermolysis bullosa with COL17A1 mutations. J Invest Dermatol. 2012;132:1374–83.PubMedCrossRef
51.
Zurück zum Zitat Twaroski K, Eide C, Riddle MJ, Xia L, Lees CJ, Chen W, et al. Revertant mosaic fibroblasts in recessive dystrophic epidermolysis bullosa. Br J Dermatol. 2019;181:1247–53.PubMedCrossRefPubMedCentral Twaroski K, Eide C, Riddle MJ, Xia L, Lees CJ, Chen W, et al. Revertant mosaic fibroblasts in recessive dystrophic epidermolysis bullosa. Br J Dermatol. 2019;181:1247–53.PubMedCrossRefPubMedCentral
52.
Zurück zum Zitat Gostynski A, Pasmooij AM, Jonkman MF. Successful therapeutic transplantation of revertant skin in epidermolysis bullosa. J Am Acad Dermatol. 2014;70:98–101.PubMedCrossRef Gostynski A, Pasmooij AM, Jonkman MF. Successful therapeutic transplantation of revertant skin in epidermolysis bullosa. J Am Acad Dermatol. 2014;70:98–101.PubMedCrossRef
53.
Zurück zum Zitat Yuen WY, Huizinga J, Jonkman MF. Punch grafting of chronic ulcers in patients with laminin-332-deficient, non-Herlitz junctional epidermolysis bullosa. J Am Acad Dermatol. 2013;68:93–7 (97 e1-2).PubMedCrossRef Yuen WY, Huizinga J, Jonkman MF. Punch grafting of chronic ulcers in patients with laminin-332-deficient, non-Herlitz junctional epidermolysis bullosa. J Am Acad Dermatol. 2013;68:93–7 (97 e1-2).PubMedCrossRef
54.
Zurück zum Zitat Rodrigues M, Yokota T. An overview of recent advances and clinical applications of exon skipping and splice modulation for muscular dystrophy and various genetic diseases. Methods Mol Biol Clifton NJ. 2018;1828:31–55.CrossRef Rodrigues M, Yokota T. An overview of recent advances and clinical applications of exon skipping and splice modulation for muscular dystrophy and various genetic diseases. Methods Mol Biol Clifton NJ. 2018;1828:31–55.CrossRef
55.
Zurück zum Zitat McGrath JA, Ashton GH, Mellerio JE, Salas-Alanis JC, Swensson O, McMillan JR, et al. Moderation of phenotypic severity in dystrophic and junctional forms of epidermolysis bullosa through in-frame skipping of exons containing non-sense or frameshift mutations. J Invest Dermatol. 1999;113:314–21.PubMedCrossRef McGrath JA, Ashton GH, Mellerio JE, Salas-Alanis JC, Swensson O, McMillan JR, et al. Moderation of phenotypic severity in dystrophic and junctional forms of epidermolysis bullosa through in-frame skipping of exons containing non-sense or frameshift mutations. J Invest Dermatol. 1999;113:314–21.PubMedCrossRef
56.
Zurück zum Zitat Kowalewski C, Bremer J, Gostynski A, Wertheim-Tysarowska K, Wozniak K, Bal J, et al. Amelioration of junctional epidermolysis bullosa due to exon skipping. Br J Dermatol. 2016;174:1375–9.PubMedCrossRef Kowalewski C, Bremer J, Gostynski A, Wertheim-Tysarowska K, Wozniak K, Bal J, et al. Amelioration of junctional epidermolysis bullosa due to exon skipping. Br J Dermatol. 2016;174:1375–9.PubMedCrossRef
57.
Zurück zum Zitat Bremer J, van der Heijden EH, Eichhorn DS, Meijer R, Lemmink HH, Scheffer H, et al. Natural exon skipping sets the stage for exon skipping as therapy for dystrophic epidermolysis bullosa. Mol Ther Nucleic Acids. 2019;18:465–75.PubMedPubMedCentralCrossRef Bremer J, van der Heijden EH, Eichhorn DS, Meijer R, Lemmink HH, Scheffer H, et al. Natural exon skipping sets the stage for exon skipping as therapy for dystrophic epidermolysis bullosa. Mol Ther Nucleic Acids. 2019;18:465–75.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Turczynski S, Titeux M, Tonasso L, Décha A, Ishida-Yamamoto A, Hovnanian A. Targeted exon skipping restores type VII collagen expression and anchoring fibril formation in an in vivo RDEB model. J Invest Dermatol. 2016;136:2387–95.PubMedCrossRef Turczynski S, Titeux M, Tonasso L, Décha A, Ishida-Yamamoto A, Hovnanian A. Targeted exon skipping restores type VII collagen expression and anchoring fibril formation in an in vivo RDEB model. J Invest Dermatol. 2016;136:2387–95.PubMedCrossRef
59.
Zurück zum Zitat Bremer J, Bornert O, Nyström A, Gostynski A, Jonkman MF, Aartsma-Rus A, et al. Antisense oligonucleotide-mediated exon skipping as a systemic therapeutic approach for recessive dystrophic epidermolysis bullosa. Mol Ther Nucleic Acids. 2016;5:e379.PubMedCrossRef Bremer J, Bornert O, Nyström A, Gostynski A, Jonkman MF, Aartsma-Rus A, et al. Antisense oligonucleotide-mediated exon skipping as a systemic therapeutic approach for recessive dystrophic epidermolysis bullosa. Mol Ther Nucleic Acids. 2016;5:e379.PubMedCrossRef
60.
Zurück zum Zitat Bornert O, Kühl T, Bremer J, van den Akker PC, Pasmooij AM, Nyström A. Analysis of the functional consequences of targeted exon deletion in COL7A1 reveals prospects for dystrophic epidermolysis bullosa therapy. Mol Ther J Am Soc Gene Ther. 2016;24:1302–11.CrossRef Bornert O, Kühl T, Bremer J, van den Akker PC, Pasmooij AM, Nyström A. Analysis of the functional consequences of targeted exon deletion in COL7A1 reveals prospects for dystrophic epidermolysis bullosa therapy. Mol Ther J Am Soc Gene Ther. 2016;24:1302–11.CrossRef
61.
Zurück zum Zitat Goto M, Sawamura D, Nishie W, Sakai K, McMillan JR, Akiyama M, et al. Targeted skipping of a single exon harboring a premature termination codon mutation: implications and potential for gene correction therapy for selective dystrophic epidermolysis bullosa patients. J Invest Dermatol. 2006;126:2614–20.PubMedCrossRef Goto M, Sawamura D, Nishie W, Sakai K, McMillan JR, Akiyama M, et al. Targeted skipping of a single exon harboring a premature termination codon mutation: implications and potential for gene correction therapy for selective dystrophic epidermolysis bullosa patients. J Invest Dermatol. 2006;126:2614–20.PubMedCrossRef
62.
Zurück zum Zitat Woodley DT, Keene DR, Atha T, Huang Y, Lipman K, Li W, et al. Injection of recombinant human type VII collagen restores collagen function in dystrophic epidermolysis bullosa. Nat Med. 2004;10:693–5.PubMedCrossRef Woodley DT, Keene DR, Atha T, Huang Y, Lipman K, Li W, et al. Injection of recombinant human type VII collagen restores collagen function in dystrophic epidermolysis bullosa. Nat Med. 2004;10:693–5.PubMedCrossRef
63.
Zurück zum Zitat 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;133:1910–3.PubMedPubMedCentralCrossRef 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;133:1910–3.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat 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;108:6609–14.PubMedCrossRefPubMedCentral 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;108:6609–14.PubMedCrossRefPubMedCentral
65.
Zurück zum Zitat Aikawa E, Fujita R, Kikuchi Y, Kaneda Y, Tamai K. Systemic high-mobility group box 1 administration suppresses skin inflammation by inducing an accumulation of PDGFRα(+) mesenchymal cells from bone marrow. Sci Rep. 2015;5:11008.PubMedPubMedCentralCrossRef Aikawa E, Fujita R, Kikuchi Y, Kaneda Y, Tamai K. Systemic high-mobility group box 1 administration suppresses skin inflammation by inducing an accumulation of PDGFRα(+) mesenchymal cells from bone marrow. Sci Rep. 2015;5:11008.PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Kido T, Miyagawa S, Goto T, Tamai K, Ueno T, Toda K, et al. The administration of high-mobility group box 1 fragment prevents deterioration of cardiac performance by enhancement of bone marrow mesenchymal stem cell homing in the delta-sarcoglycan-deficient hamster. PLoS One. 2018;13:e0202838.PubMedPubMedCentralCrossRef Kido T, Miyagawa S, Goto T, Tamai K, Ueno T, Toda K, et al. The administration of high-mobility group box 1 fragment prevents deterioration of cardiac performance by enhancement of bone marrow mesenchymal stem cell homing in the delta-sarcoglycan-deficient hamster. PLoS One. 2018;13:e0202838.PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Shimbo T, Yamazaki S, Wang X, Kikuchi Y, Bruckner-Tuderman L, Kaneda Y, et al. 906 Systemic HMGB1 administration ameliorates cutaneous and non-cutaneous manifestations in a dystrophic epidermolysis bullosa model mouse. J Invest Dermatol. 2017;137:S156.CrossRef Shimbo T, Yamazaki S, Wang X, Kikuchi Y, Bruckner-Tuderman L, Kaneda Y, et al. 906 Systemic HMGB1 administration ameliorates cutaneous and non-cutaneous manifestations in a dystrophic epidermolysis bullosa model mouse. J Invest Dermatol. 2017;137:S156.CrossRef
68.
Zurück zum Zitat Welch EM, Barton ER, Zhuo J, Tomizawa Y, Friesen WJ, Trifillis P, et al. PTC124 targets genetic disorders caused by nonsense mutations. Nature. 2007;447:87–91.PubMedCrossRef Welch EM, Barton ER, Zhuo J, Tomizawa Y, Friesen WJ, Trifillis P, et al. PTC124 targets genetic disorders caused by nonsense mutations. Nature. 2007;447:87–91.PubMedCrossRef
69.
Zurück zum Zitat McElroy SP, Nomura T, Torrie LS, Warbrick E, Gartner U, Wood G, et al. A lack of premature termination codon read-through efficacy of PTC124 (Ataluren) in a diverse array of reporter assays. PLoS Biol. 2013;11:e1001593.PubMedPubMedCentralCrossRef McElroy SP, Nomura T, Torrie LS, Warbrick E, Gartner U, Wood G, et al. A lack of premature termination codon read-through efficacy of PTC124 (Ataluren) in a diverse array of reporter assays. PLoS Biol. 2013;11:e1001593.PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Walker PD, Shah SV. Evidence suggesting a role for hydroxyl radical in gentamicin-induced acute renal failure in rats. J Clin Invest. 1988;81:334–41.PubMedPubMedCentralCrossRef Walker PD, Shah SV. Evidence suggesting a role for hydroxyl radical in gentamicin-induced acute renal failure in rats. J Clin Invest. 1988;81:334–41.PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Gonzalez-Hilarion S, Beghyn T, Jia J, Debreuck N, Berte G, Mamchaoui K, et al. Rescue of nonsense mutations by amlexanox in human cells. Orphanet J Rare Dis. 2012;7:58.PubMedPubMedCentralCrossRef Gonzalez-Hilarion S, Beghyn T, Jia J, Debreuck N, Berte G, Mamchaoui K, et al. Rescue of nonsense mutations by amlexanox in human cells. Orphanet J Rare Dis. 2012;7:58.PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Atanasova VS, Russell RJ, Webster TG, Cao Q, Agarwal P, Lim YZ, et al. Thrombospondin-1 is a major activator of TGF-β signaling in recessive dystrophic epidermolysis bullosa fibroblasts. J Invest Dermatol. 2019;139(1497–1505):e5. Atanasova VS, Russell RJ, Webster TG, Cao Q, Agarwal P, Lim YZ, et al. Thrombospondin-1 is a major activator of TGF-β signaling in recessive dystrophic epidermolysis bullosa fibroblasts. J Invest Dermatol. 2019;139(1497–1505):e5.
73.
Zurück zum Zitat Nyström A, Velati D, Mittapalli VR, Fritsch A, Kern JS, Bruckner-Tuderman L. Collagen VII plays a dual role in wound healing. J Clin Invest. 2013;123:3498–509.PubMedPubMedCentralCrossRef Nyström A, Velati D, Mittapalli VR, Fritsch A, Kern JS, Bruckner-Tuderman L. Collagen VII plays a dual role in wound healing. J Clin Invest. 2013;123:3498–509.PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat Nyström A, Bruckner-Tuderman L. Injury- and inflammation-driven skin fibrosis: the paradigm of epidermolysis bullosa. Matrix Biol J Int Soc Matrix Biol. 2018;68–69:547–60.CrossRef Nyström A, Bruckner-Tuderman L. Injury- and inflammation-driven skin fibrosis: the paradigm of epidermolysis bullosa. Matrix Biol J Int Soc Matrix Biol. 2018;68–69:547–60.CrossRef
75.
Zurück zum Zitat Ng YZ, Pourreyron C, Salas-Alanis JC, Dayal JH, Cepeda-Valdes R, Yan W, et al. Fibroblast-derived dermal matrix drives development of aggressive cutaneous squamous cell carcinoma in patients with recessive dystrophic epidermolysis bullosa. Cancer Res. 2012;72:3522–34.PubMedCrossRef Ng YZ, Pourreyron C, Salas-Alanis JC, Dayal JH, Cepeda-Valdes R, Yan W, et al. Fibroblast-derived dermal matrix drives development of aggressive cutaneous squamous cell carcinoma in patients with recessive dystrophic epidermolysis bullosa. Cancer Res. 2012;72:3522–34.PubMedCrossRef
76.
Zurück zum Zitat Shi Y, Massagué J. Mechanisms of TGF-beta signaling from cell membrane to the nucleus. Cell. 2003;113:685–700.PubMedCrossRef Shi Y, Massagué J. Mechanisms of TGF-beta signaling from cell membrane to the nucleus. Cell. 2003;113:685–700.PubMedCrossRef
78.
Zurück zum Zitat Nyström A, Thriene K, Mittapalli V, Kern JS, Kiritsi D, Dengjel J, et al. Losartan ameliorates dystrophic epidermolysis bullosa and uncovers new disease mechanisms. EMBO Mol Med. 2015;7:1211–28.PubMedPubMedCentralCrossRef Nyström A, Thriene K, Mittapalli V, Kern JS, Kiritsi D, Dengjel J, et al. Losartan ameliorates dystrophic epidermolysis bullosa and uncovers new disease mechanisms. EMBO Mol Med. 2015;7:1211–28.PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Cianfarani F, De Domenico E, Nyström A, Mastroeni S, Abeni D, Baldini E, et al. Decorin counteracts disease progression in mice with recessive dystrophic epidermolysis bullosa. Matrix Biol J Int Soc Matrix Biol. 2019;81:3–16.CrossRef Cianfarani F, De Domenico E, Nyström A, Mastroeni S, Abeni D, Baldini E, et al. Decorin counteracts disease progression in mice with recessive dystrophic epidermolysis bullosa. Matrix Biol J Int Soc Matrix Biol. 2019;81:3–16.CrossRef
80.
Zurück zum Zitat Mellerio JE, Robertson SJ, Bernardis C, Diem A, Fine JD, George R, et al. Management of cutaneous squamous cell carcinoma in patients with epidermolysis bullosa: best clinical practice guidelines. Br J Dermatol. 2016;174:56–67.PubMedCrossRef Mellerio JE, Robertson SJ, Bernardis C, Diem A, Fine JD, George R, et al. Management of cutaneous squamous cell carcinoma in patients with epidermolysis bullosa: best clinical practice guidelines. Br J Dermatol. 2016;174:56–67.PubMedCrossRef
81.
Zurück zum Zitat Ciardiello F, Tortora G. EGFR antagonists in cancer treatment. N Engl J Med. 2008;358:1160–74.PubMedCrossRef Ciardiello F, Tortora G. EGFR antagonists in cancer treatment. N Engl J Med. 2008;358:1160–74.PubMedCrossRef
82.
Zurück zum Zitat Medek K, Koelblinger P, Koller J, Diem A, Ude-Schoder K, Bauer JW, et al. Wundheilungsstörungen während der antitumorösen Therapie mit Cetuximab bei schwerer generalisierter dystropher Epidermolysis bullosa. J Dtsch Dermatol Ges J Ger Soc Dermatol JDDG. 2019;17:448–50. Medek K, Koelblinger P, Koller J, Diem A, Ude-Schoder K, Bauer JW, et al. Wundheilungsstörungen während der antitumorösen Therapie mit Cetuximab bei schwerer generalisierter dystropher Epidermolysis bullosa. J Dtsch Dermatol Ges J Ger Soc Dermatol JDDG. 2019;17:448–50.
83.
Zurück zum Zitat Arnold AW, Bruckner-Tuderman L, Zuger C, Itin PH. Cetuximab therapy of metastasizing cutaneous squamous cell carcinoma in a patient with severe recessive dystrophic epidermolysis bullosa. Dermatol Basel Switz. 2009;219:80–3.CrossRef Arnold AW, Bruckner-Tuderman L, Zuger C, Itin PH. Cetuximab therapy of metastasizing cutaneous squamous cell carcinoma in a patient with severe recessive dystrophic epidermolysis bullosa. Dermatol Basel Switz. 2009;219:80–3.CrossRef
84.
Zurück zum Zitat Kim M, Li M, Intong LRA, Tran K, Melbourne W, Marucci D, et al. Use of cetuximab as an adjuvant agent to radiotherapy and surgery in recessive dystrophic epidermolysis bullosa with squamous cell carcinoma. Br J Dermatol. 2013;169:208–10.PubMedCrossRef Kim M, Li M, Intong LRA, Tran K, Melbourne W, Marucci D, et al. Use of cetuximab as an adjuvant agent to radiotherapy and surgery in recessive dystrophic epidermolysis bullosa with squamous cell carcinoma. Br J Dermatol. 2013;169:208–10.PubMedCrossRef
85.
Zurück zum Zitat Reimer A, Lu S, He Y, Bruckner-Tuderman L, Technau-Hafsi K, Meiss F, et al. Combined anti-inflammatory and low-dose antiproliferative therapy for squamous cell carcinomas in recessive dystrophic epidermolysis bullosa. J Eur Acad Dermatol Venereol JEADV. 2020;34:e1–3.PubMedCrossRef Reimer A, Lu S, He Y, Bruckner-Tuderman L, Technau-Hafsi K, Meiss F, et al. Combined anti-inflammatory and low-dose antiproliferative therapy for squamous cell carcinomas in recessive dystrophic epidermolysis bullosa. J Eur Acad Dermatol Venereol JEADV. 2020;34:e1–3.PubMedCrossRef
86.
Zurück zum Zitat Diociaiuti A, Steinke H, Nyström A, Schwieger-Briel A, Meiss F, Pfannenberg C, et al. EGFR inhibition for metastasized cutaneous squamous cell carcinoma in dystrophic epidermolysis bullosa. Orphanet J Rare Dis. 2019;14:278.PubMedPubMedCentralCrossRef Diociaiuti A, Steinke H, Nyström A, Schwieger-Briel A, Meiss F, Pfannenberg C, et al. EGFR inhibition for metastasized cutaneous squamous cell carcinoma in dystrophic epidermolysis bullosa. Orphanet J Rare Dis. 2019;14:278.PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat Ferris RL, Blumenschein G, Fayette J, Guigay J, Colevas AD, Licitra L, et al. Nivolumab for recurrent squamous-cell carcinoma of the head and neck. N Engl J Med. 2016;375:1856–67.PubMedPubMedCentralCrossRef Ferris RL, Blumenschein G, Fayette J, Guigay J, Colevas AD, Licitra L, et al. Nivolumab for recurrent squamous-cell carcinoma of the head and neck. N Engl J Med. 2016;375:1856–67.PubMedPubMedCentralCrossRef
88.
Zurück zum Zitat Migden MR, Rischin D, Schmults CD, Guminski A, Hauschild A, Lewis KD, et al. PD-1 blockade with cemiplimab in advanced cutaneous squamous-cell carcinoma. N Engl J Med. 2018;379:341–51.PubMedCrossRef Migden MR, Rischin D, Schmults CD, Guminski A, Hauschild A, Lewis KD, et al. PD-1 blockade with cemiplimab in advanced cutaneous squamous-cell carcinoma. N Engl J Med. 2018;379:341–51.PubMedCrossRef
89.
Zurück zum Zitat Fine JD, Johnson LB, Weiner M, Stein A, Suchindran C. Chemoprevention of squamous cell carcinoma in recessive dystrophic epidermolysis bullosa: results of a phase 1 trial of systemic isotretinoin. J Am Acad Dermatol. 2004;50:563–71.PubMedCrossRef Fine JD, Johnson LB, Weiner M, Stein A, Suchindran C. Chemoprevention of squamous cell carcinoma in recessive dystrophic epidermolysis bullosa: results of a phase 1 trial of systemic isotretinoin. J Am Acad Dermatol. 2004;50:563–71.PubMedCrossRef
90.
Zurück zum Zitat Atanasova VS, Pourreyron C, Farshchian M, Lawler M, Brown CA, Watt SA, et al. Identification of rigosertib for the treatment of recessive dystrophic epidermolysis bullosa-associated squamous cell carcinoma. Clin Cancer Res Off J Am Assoc Cancer Res. 2019;25:3384–91.CrossRef Atanasova VS, Pourreyron C, Farshchian M, Lawler M, Brown CA, Watt SA, et al. Identification of rigosertib for the treatment of recessive dystrophic epidermolysis bullosa-associated squamous cell carcinoma. Clin Cancer Res Off J Am Assoc Cancer Res. 2019;25:3384–91.CrossRef
91.
92.
Zurück zum Zitat Lwin SM, Syed F, Di W-L, Kadiyirire T, Liu L, Guy A, et al. Safety and early efficacy outcomes for lentiviral fibroblast gene therapy in recessive dystrophic epidermolysis bullosa. JCI Insight. 2019;4:e126243.PubMedCentralCrossRef Lwin SM, Syed F, Di W-L, Kadiyirire T, Liu L, Guy A, et al. Safety and early efficacy outcomes for lentiviral fibroblast gene therapy in recessive dystrophic epidermolysis bullosa. JCI Insight. 2019;4:e126243.PubMedCentralCrossRef
93.
Zurück zum Zitat Osborn MJ, Lees CJ, McElroy AN, Merkel SC, Eide CR, Mathews W, et al. CRISPR/Cas9-based cellular engineering for targeted gene overexpression. Int J Mol Sci. 2018;19:946.PubMedCentralCrossRef Osborn MJ, Lees CJ, McElroy AN, Merkel SC, Eide CR, Mathews W, et al. CRISPR/Cas9-based cellular engineering for targeted gene overexpression. Int J Mol Sci. 2018;19:946.PubMedCentralCrossRef
94.
Zurück zum Zitat Webber BR, Osborn MJ, McElroy AN, Twaroski K, Lonetree C-L, DeFeo AP, et al. CRISPR/Cas9-based genetic correction for recessive dystrophic epidermolysis bullosa. NPJ Regen Med. 2016;1:16014.PubMedPubMedCentralCrossRef Webber BR, Osborn MJ, McElroy AN, Twaroski K, Lonetree C-L, DeFeo AP, et al. CRISPR/Cas9-based genetic correction for recessive dystrophic epidermolysis bullosa. NPJ Regen Med. 2016;1:16014.PubMedPubMedCentralCrossRef
95.
Zurück zum Zitat Shinkuma S, Guo Z, Christiano AM. Site-specific genome editing for correction of induced pluripotent stem cells derived from dominant dystrophic epidermolysis bullosa. Proc Natl Acad Sci USA. 2016;113:5676–81.PubMedCrossRefPubMedCentral Shinkuma S, Guo Z, Christiano AM. Site-specific genome editing for correction of induced pluripotent stem cells derived from dominant dystrophic epidermolysis bullosa. Proc Natl Acad Sci USA. 2016;113:5676–81.PubMedCrossRefPubMedCentral
96.
Zurück zum Zitat Wu W, Lu Z, Li F, Wang W, Qian N, Duan J, et al. Efficient in vivo gene editing using ribonucleoproteins in skin stem cells of recessive dystrophic epidermolysis bullosa mouse model. Proc Natl Acad Sci USA. 2017;114:1660–5.PubMedCrossRefPubMedCentral Wu W, Lu Z, Li F, Wang W, Qian N, Duan J, et al. Efficient in vivo gene editing using ribonucleoproteins in skin stem cells of recessive dystrophic epidermolysis bullosa mouse model. Proc Natl Acad Sci USA. 2017;114:1660–5.PubMedCrossRefPubMedCentral
97.
Zurück zum Zitat Benati D, Miselli F, Cocchiarella F, Patrizi C, Carretero M, Baldassarri S, et al. CRISPR/Cas9-mediated in situ correction of LAMB3 gene in keratinocytes derived from a junctional epidermolysis bullosa patient. Mol Ther J Am Soc Gene Ther. 2018;26:2592–603.CrossRef Benati D, Miselli F, Cocchiarella F, Patrizi C, Carretero M, Baldassarri S, et al. CRISPR/Cas9-mediated in situ correction of LAMB3 gene in keratinocytes derived from a junctional epidermolysis bullosa patient. Mol Ther J Am Soc Gene Ther. 2018;26:2592–603.CrossRef
98.
Zurück zum Zitat Tockner B, Kocher T, Hainzl S, Reichelt J, Bauer JW, Koller U, et al. Construction and validation of an RNA trans-splicing molecule suitable to repair a large number of COL7A1 mutations. Gene Ther. 2016;23:775–84.PubMedPubMedCentralCrossRef Tockner B, Kocher T, Hainzl S, Reichelt J, Bauer JW, Koller U, et al. Construction and validation of an RNA trans-splicing molecule suitable to repair a large number of COL7A1 mutations. Gene Ther. 2016;23:775–84.PubMedPubMedCentralCrossRef
99.
Zurück zum Zitat Peking P, Koller U, Duarte B, Murillas R, Wolf S, Maetzig T, et al. An RNA-targeted therapy for dystrophic epidermolysis bullosa. Nucleic Acids Res. 2017;45:10259–69.PubMedPubMedCentralCrossRef Peking P, Koller U, Duarte B, Murillas R, Wolf S, Maetzig T, et al. An RNA-targeted therapy for dystrophic epidermolysis bullosa. Nucleic Acids Res. 2017;45:10259–69.PubMedPubMedCentralCrossRef
100.
Zurück zum Zitat Mellerio J, Uitto J. Meeting report: The first global congress on epidermolysis bullosa, EB2020 -Toward treatment and cure. J Invest Dermatol (in press). Mellerio J, Uitto J. Meeting report: The first global congress on epidermolysis bullosa, EB2020 -Toward treatment and cure. J Invest Dermatol (in press).
Metadaten
Titel
Molecular Therapeutics in Development for Epidermolysis Bullosa: Update 2020
verfasst von
Cristina Has
Andrew South
Jouni Uitto
Publikationsdatum
01.06.2020
Verlag
Springer International Publishing
Erschienen in
Molecular Diagnosis & Therapy / Ausgabe 3/2020
Print ISSN: 1177-1062
Elektronische ISSN: 1179-2000
DOI
https://doi.org/10.1007/s40291-020-00466-7

Weitere Artikel der Ausgabe 3/2020

Molecular Diagnosis & Therapy 3/2020 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.