Skip to main content
Erschienen in: Wiener Medizinische Wochenschrift 9-10/2019

Open Access 13.12.2018 | review

Nerve grafting for peripheral nerve injuries with extended defect sizes

verfasst von: Tim Kornfeld, Peter M. Vogt, Christine Radtke, MD, PhD

Erschienen in: Wiener Medizinische Wochenschrift | Ausgabe 9-10/2019

Summary

Artificial and non-artificial nerve grafts are the gold standard in peripheral nerve reconstruction in cases with extensive loss of nerve tissue, particularly where a direct end-to-end suture or an autologous nerve graft is inauspicious. Different materials are marketed and approved by the US Food and Drug Administration (FDA) for peripheral nerve graft reconstruction. The most frequently used materials are collagen and poly(DL-lactide-ε-caprolactone). Only one human nerve allograft is listed for peripheral nerve reconstruction by the FDA. All marketed nerve grafts are able to demonstrate sufficient nerve regeneration over small distances not exceeding 3.0 cm. A key question in the field is whether nerve reconstruction on large defect lengths extending 4.0 cm or more is possible. This review gives a summary of current clinical and experimental approaches in peripheral nerve surgery using artificial and non-artificial nerve grafts in short and long distance nerve defects. Strategies to extend nerve graft lengths for long nerve defects, such as enhancing axonal regeneration, include the additional application of Schwann cells, mesenchymal stem cells or supporting co-factors like growth factors on defect sizes between 4.0 and 8.0 cm.
Hinweise
Tim Kornfeld, Peter M. Vogt and Christine Radtke contributed equally to this work.
Abkürzungen
CMAP
Compound action potentials
CSPGs
Chondroitin sulfate proteoglycans
EM
Electron microscopy
ENT
Ear, nose and throat
FDA
US Food and Drug Administration
FGF
Fibroblast growth factor
MHC
Major histocompatibility complex
MSCs
Mesenchymal stem cells
PGA
Polyglycolic acid

Introduction

Peripheral nerve damage is a severe and critical problem in all disciplines of surgery. With an overall incidence of 17.4% of iatrogenic acquired nerve lesions, this appears to be an underestimated major problem in patient treatment [1]. Several cases have been reported where a peripheral nerve was mistakenly used as a muscle tendon graft [2, 3] or nerve fibers have been damaged during anesthetic interventions [46]. The vast majority of peripheral nerve defects are caused by severe trauma or tumor infiltration. Noble et al. revealed that severe trauma leads to peripheral nerve injury with a prevalence of 2.8% in level 1 trauma patients [7].
Whether the nerve defect is caused iatrogenically or traumatically there is no difference in the surgical approach for nerve reconstruction. The current gold standard in nerve repair surgery is the tension free end-to-end suture. If this is not achievable an autologous nerve graft is indicated where a donor nerve is harvested and sutured to bridge the defect [8, 9]. The major disadvantage of this technique is the remarkable loss of sensitivity in the area of distribution and the limited availability of autologous donor tissue [10].
Artificial nerve grafts can be used as an alternative in cases of multiple nerve lesions where subsequent treatment with autografts is not possible due to limitation of donor tissue [11]. A variety of artificial nerve grafts is commercially available and approved by the FDA (US Food and Drug Administration) [12]. The data of the FDA revealed that collagen, chitosan and poly (DL-lactide-ε-caprolactone) are the most frequently used and approved materials (Table 1). Table 1 summarize all marketed and FDA-approved artificial nerve grafts for the surgical reconstruction of peripheral nerve tissue.
Table 1
FDA-approved nerve tubes for peripheral nerve repair. (Modified and supplemented from FDA Medical Device Database [104])
Name
Product name
Company
Date of approval
K510
Available Length (cm)
Material
Neuragen 3D
NeuraGen®
Integra Lifescience Corporation, Plainsboro, NJ, USA
Apr 24, 2014
K130557
3.0
Collagen
Neurogen Nerve Guide
NeuroGen®
Integra Lifescience Corporation, Plainsboro, NJ, USA
Jun 22, 2001
K011168
3.0
Collagen
Flexible collagen nerve
NeuroFlex®
Collagen Matrix Inc., Oakland, NJ, USA
Apr 03, 2014
K131541
2.5
Collagen
Collagen nerve cuff
NeuroMatrix®
Collagen Matrix Inc., Oakland, NJ, USA
Sep 21, 2001
K012814
n.a
Collagen
Reaxon Plus®
Reaxon Plus®
Medovent GmbH, Mainz, Germany
Dec 02, 2015
K143711
1.0
Chitosan
Neurotube®
Neurotube®
Neuroregen L.C.C, Bel Air, MD, USA
Mar 22, 1999
K983007
3.0
Poly(DL-lactide-ε-caprolactone)
Neurolac® Nerve Guide
Neurolac®
Polyganics BV, Rozenburglaan, Netherlands
Oct 20, 2011
K112267
2.0
Poly(DL-lactide-ε-caprolactone)
Neurolac® Nerve Guide
Neurolac®
Polyganics BV, Rozenburglaan, Netherlands
May 04, 2005
K050573
2.0
Poly(DL-lactide-ε-caprolactone)
Neurolac® Nerve Guide Models NG01-15/03, NG01-020/03, NG01-025/03, NG01-030/03
Neurolac®
Polyganics BV, Rozenburglaan, Netherlands
Oct 10, 2003
K032115
2.0
Poly(DL-lactide-ε-caprolactone)
AxoGen Avance®
AxoGen Avance®
AxoGen, Alachua, FL, USA
5.0
Human nerve allograft
The first collagen based peripheral nerve implants were approved for surgical intervention by the FDA in the early 2000s. Nearly simultaneously Neurogen® (Integra Lifescience Corporation, Plainsboro, NJ, USA) and Neuroflex® (Collagen Matrix Inc, Oakland, NJ, USA) have been introduced to the market. Neurogen® is a collagen based nerve tube demonstrating a satisfying recovery rate of 43% of level 1 trauma treated patients with peripheral nerve defects pending between 2.5–20.0 mm in length [13]. Neuoflex® is a collagen based nerve tube distributed by Stryker Corporation (Stryker Corporation, Kalamazoo, MI, USA). No clinical data is available after intensive literature research. Importantly, the collagen based materials have a significant low antigenicity and immunogenicity in vivo what makes them to a favorable material for in vivo applications [14].
Reaxon plus® (Medovent GmbH, Mainz, Germany) represents an approved conduit constructed out of the natural biomaterial chitosan. Chitosan was successfully used in rodent animal experiments for nerve regeneration over distances of up to 15.0 mm [15]. In this study nerve regeneration using chitosan nerve grafts was as effective as autologous nerve transplantation. Subsequent studies revealed that chitosan nerve grafts are able to reduce post-traumatic formation of neuroma and epineural fibrosis [16]. Unfortunately, chitosan was also reported to induce a foreign body reaction during degradation in vivo [17].
Neurotube® (Neuroregen L.C.C, Bel Air, MD, USA) and Neurolac® (Polyganics BV, Rozenburglaan, Netherlands) are nerve cuffs manufactured out of poly(DL-lactide-ε-caprolactone) and are approved for surgical reconstruction of nerve defects up to 3.0 cm in length. Neurotube® demonstrated successful facial nerve regeneration in several cases on defect sizes between 1.0 and 3.0 cm [18]. No signs of tissue rejection or inflammation processes occurred during application in human facial nerve repair. However, Duncan et al. report a case concerning the extrusion of a Neurotube® implant associated with inflammation processes [19]. Neurolac® is a competitive product to Neurotube® and one report suggests improved regeneration compared to autologous nerve grafts on a 1.0 cm peripheral nerve defect in rodents [20]. Supporting the results from den Dunnen et al. [19] a case series from Brazil indicates that the use of Neurolac® is a safe and successful procedure for peripheral nerve surgery [21].
AxoGen Avance® (AxoGen, Alachua, FL, USA) is the only FDA approved human nerve allograft. In several studies in animal models the efficiency of nerve regeneration through decellularized allografts was demonstrated, but was still inferior to isografts [22]. Clinical trials revealed that 87% of 132 nerve injuries treated with Axogen Avance® (AxoGen, Alachua, FL, USA) regained sensory and/or motor functions for treated peripheral nerve defect sizes between 5.0 and 50.0 mm [23]. Just recently Rinker et al. analyzed the results of the Ranger I study in regard to peripheral nerve defects on small diameter nerve defects on the hand [24]. Accordingly to the results of Brooks et al. [23] nerve regeneration with regained sensory function (S3+ or higher) was observable in 86% of the included cases.
All marketed and FDA approved nerve grafts demonstrate satisfying recovery on defect length from up to 3.0 cm with a minimal amount of side effects or regeneration failure. Despite these high number of FDA approved and commercially available artificial nerve grafts for reconstruction of peripheral nerve defects, no implant is approved and available for defect sizes extending 3.0 cm or longer in length. Nerve defects extending these 3.0 cm are usually considered to be critical [25]. Good clinical and non-clinical data is available for short nerve defects not extending 3.0 cm [1321]. Recently Kaplan et al. criticized that only little clinical data is available for long gaped peripheral nerve defects and that rodent animals might not be a suitable animal model for translational research in peripheral nerve surgery [25]. Sufficient data concerning larger gap sizes is rare and the vast majority of scientific approaches does not lead to the expected results. Nevertheless surgical approaches for long gap nerve defects are needed during daily medical practice. Especially extended and multiple injuries of e. g. plexus nerves by trauma or long length tumor infiltration of peripheral nerve tissue are challenging in reconstruction These review gives a brief overview about current approaches in peripheral nerve surgery for critical gap sizes between 4.0 and 8.0 cm in small and large animal models using acellular nerve grafts or pre-seeded conduits.

Material and methods

Including criteria

Only scientific work in regard to peripheral nerve regeneration/reconstruction was included. Main criteria was a surgical nerve reconstruction in small/large animal models with artificial/non-artificial nerve grafts on nerve defect sizes ≥4.0 cm.

Matches

In all, 30 original articles met the inclusion criteria of nerve reconstruction on nerve defects ≥4.0 cm. Four records were excluded after identified as duplicates. Two article were removed due to incomparable methods.
A literature search was performed via PubMed and Google Scholar. A key word search was performed using the following: “long gap nerve defects”, “extended nerve defects”, “reconstruction of extended nerve defects”, “critical sized nerve defects”, “nerve defects in large animal models”.

Current experimental approaches in peripheral nerve reconstruction

Small animal models

Small animal models are widely used in experimental in vivo investigation in the field of peripheral nerve surgery [22, 2628]. High availability with moderate holding costs makes them a competitive and indispensable model for efficient and high throughput surgical testing experiments [29]. The sciatic nerves are easily accessible via a dorsal operation route. Postsurgical nerve regeneration can be evaluated in vivo either by electrophysiology, nerve pinch test or walking track analysis [30, 31]. Literature shows that the small animal models are also suitable for nerve surgery in settings with nerve defects extending 4.0 cm in length [3240]. A disadvantage of the rodent model is that the dynamics of peripheral nerve regeneration may be different than in large animal models including humans, due to issues of scale [25, 41]. Table 2 gives a brief overview of current approaches in peripheral nerve surgery in small rodent animal models with or without cell transplantation and the addition of co-factors (Table 2).
Table 2
Small animal models in peripheral nerve surgery
 
Defect size (cm)
Nerve
Animal
Implant
Time
Outcome
Quotation
Small animal models in nerve surgery without cell transplantation
1
4.0
Tibial nerve
Rat
Chondroitinase ABC processed nerve
3 M
Higher numbers of regenerated axons compared to control
Neubauer et al. [32]
2
6.0
Sciaitic nerve
Rat
Allograft vs. autograft
20 W
Superior regeneration in allograft group compared to control
Saheb-al-Zamani et al. [33]
3
3.0, 5.0, 7.0
Saphenous nerve
Rabbit
Autograft
15 M
Decreasing regeneration with increasing defects size
Koller et al. [34]
4
5.0
Sciatic nerve
Rabbit
Muscle grafts
4 M
Light regeneration in muscle grafts
Mligiliche et al. [35]
5
10.0
Sciatic nerve
Rabbit
Muscle grafts
2 M
No regeneration
Hems et al. [36]
Small animal models in nerve surgery with cell transplantation
6
4.0
Tibial nerve
Rabbit
Autologous vein with and without SC, Autograft as a control
2 M
Axonal regeneration in Isograft group and Vessel filled with Schwann cells
Zhang et al. [37]
7
1.0–6.0
Peroneal nerve
Rabbit
Vein
Regeneration on 3.0 cm. Poor outcome on long distances
Strauch et al. [39]
8
6.0
Peroneal nerve
Rabbit
Vein
4 M
Regeneration on full distance
Strauch et al. [38]
9
4.0
Ulnar nerve
Rat
ε-caprolactone-co-trimethylene carbonate filled with Schwann cells
12 M
No regeneration, extended formation of neuroma
Sinis et al. [40]
M month, W week

Acellular nerve grafting

In cases were an alternative to the standard nerve end-to-end suture is desperately needed it can be defaulted to autologous nerve grafts or peripheral allografts as previously mentioned [11]. An acellular allograft or an artificial nerve graft appears as a widely investigated and suitable alternative. Especially isografts and acellular allograft have been extensively investigated in recent decades [4244].
A recent study concerning nerve grafting with the previously mentioned acellular allograft was carried out on a 4.0 cm sciatic nerve defect in Fischer F344 rats with thermally decellularized allografts pretreated with chondroitinase ABC to breakdown axon inhibitory chondroitin sulfate proteoglycans (CSPGs) [32]. The authors report that nerve grafts pretreated with chondroitinase ABC shows a higher number of regenerated myelinated axons distal of the grafts suture compared to controls. Functionally they observed a higher number of positive nerve pinch tests and a reduction of retrograde axon growth in pretreated allografts indicating that chondroitinase ABC can support nerve regeneration over long nerve defect distances.
Whether the limited regeneration in long nerve grafts is caused by high doses of chondroitin sulfate proteoglycan and other scar related axon inhibitory elements or increasing number of senescent Schwann cells is not known. Saheb-Al-Zamani et al. investigated nerve regeneration through 2.0, 4.0 and 6.0 cm acellular nerve allografts [33]. Results demonstrate high expression of senescence markers (p16INK4A and β‑galactosidase) in all grafts. Electron microscopy (EM) revealed a rising number of chromatin clumped cell nuclei with central involution in Schwann cells on acellular allografts >4.0 cm which is typical for cell aging. However, reorganization of chromatin in cell nuclei was not verifiable in short nerve grafts of 2.0 cm allograft and isografts respectively. Independent of this, nerve regeneration in autografts was superior to acellular allografts due to significantly higher numbers of myelinated fibers in distal parts of the nerve segments.
Another study investigated nerve regeneration using an autologous nerve graft on defect distances from 3.0, 5.0 and 7.0 cm in length in rabbit [34]. Maximum tetanic tension and number of myelinated axons deteriorate with increasing autograft length over a maximum 15-month observation time. Koller et al. reported that this result may indicate that nerve regeneration over a distance up to 7.0 cm is possible and the poor outcome regarding tetanic tension and remyelinization is explained by inferior vascularization in long length peripheral nerve grafts.
As an alternative to allo- and autografts, acellular muscle autografts have been investigated. The approach was first used in early the 1980s and 1990s [4547]. Mligiliche et al. carried out a study using acellular muscle autografts on a 5.0 cm sciatic nerve defect in Japanese white rabbits [35]. After 4 months following implantation, compound action potentials (CMAP) and EM recordings were performed. Results indicate regeneration within the acellular muscle graft throughout the entire defect length with decreasing number of myelinated axons dependent upon graft length. CMAP shows light reinnervation on target muscle in each graft.
Hems et al. [36] performed a similar experiment in white New Zealand rabbits. Coapting a 5.0 and 10.0 cm sciatic nerve defect using cold freeze-dried muscle grafts compared to an autologous nerve graft. Light microscopy revealed inferior recovery in muscle grafts compared to controls. Nerve regeneration deteriorates after 2.0 cm and graft tissue was replaced by fat and connective tissue.
Data on long gaped peripheral nerve defects in small animals are limited possibly due to the issue of scale. However, small animal studies revealed that peripheral nerve regeneration through autologous donor nerves is nearly possible without side effects and that regeneration through artificial nerve grafts constructed from various materials is possible. Regeneration in allografts was reported to be less satisfactory than expected [32, 33]. Given these caveats in small animal models they have certainly provided valuable information that has advanced the field.

Grafting with autologous donor cells

Schwann cells play a crucial role not only for myelination of peripheral nerve fibers, but for providing trophic support and structural guidance for axonal regeneration [48]. After nerve damage and during Wallerian degeneration when axons are degenerating, the Schwann cells disassociate from the axons and they begin to divide [49]. These changes are within the endoneural tubes or bands of Bügner. It is well established that Schwann cells not only form myelin for the electric insolation of peripheral nerves, but play a major role during the regeneration process of the peripheral nervous system by producing trophic factors and structural guidance for axonal growth [49, 50]. Several studies have investigated whether autologous Schwann cells can support peripheral nerve regeneration in artificial and non-artificial nerve grafts [5155].
Experiments have been carried out to test Schwann cells on defect sizes <3 cm [5356]. A 4.0 cm autologous monochanneled vein graft pre-seeded with 1 × 106 autologous Schwann cells in rabbit was investigated by Zhang et al. [37]. Two months post-surgery electrophysiology showed evoked muscle potentials in both autologous controls and vein grafts-seeded with Schwann cells. Histological analysis revealed that the vein graft without cell suspension collapsed and the nerve formed a neuroma, whereas autologous controls showed almost normal full regenerated nerve tissue. The vein-Schwann cell grafts revealed adequate nerve fascicle formation and high numbers of myelinated axons in the distal part of the implant. Importantly, the autologous nerve graft seeded with Schwann cells demonstrated successful nerve regeneration over a distance of 4.0 cm. However, the results may be inferior to autologous nerve grafts.
Strauch et al. [38] performed a comparable experiment with some small variations in experimental settings on 6.0 cm nerve defects in rabbits. Previously, they were able to show that regeneration through 3.0 cm autologous vein grafts without cell transplantation is possible, but poor in longer grafts [39]. For this reason the experiments were repeated to investigate the effect of long autologous vein grafts seeded with 1 × 106 Schwann cells in a peroneal nerve defect model. In contrast to the study by Zhang et al. [37] autologous vein grafts were harvested and filled with a mixture of matrigel and Schwann cells in suspension. Autologous veins filled only with matrigel were used as the control. As expected the controls filled with matrigel showed poor to no regeneration and a considerable amount of fibrosis. The autologous vein conduit filled with Schwann cells, however, showed myelinated axons in distal parts of the conduit.
In contrast to the previously described studies from Zhang et al. [37] and Strauch et al. [38] where a monochanneled vein autograft seeded with Schwann cells was successfully used on large nerve defect up to 6.0 cm, Sinis et al. [40] demonstrated negative results after treating a 4.0 cm nerve defect in rat, with a monochanneled ε‑caprolactone-co-trimethylene carbonate graft which was seeded with autologous Schwann cells. Autologous nerve graft controls demonstrate effective nerve regeneration as demonstrated by electrophysiology and immunohistology. Unfortunately, the newly developed ε‑caprolactone-co-trimethylene artificial nerve graft showed extended formation of neuroma in 13 out of 16 animals.

Large animal models

Allograft and autografts in several nerve defect injury models in small animals have indicated that regeneration is possible on critical nerve defects. As previously mentioned Koller et al., Neubauer et al. and Saheb-Al-Zamani et al. were able to demonstrate that nerve regeneration in nerve defects up to a maximal length of 7.0 cm in small animals (rabbit, rat) is realistic [3234].
In order to achieve results that are applicable to humans, a translational large animal model for peripheral nerve surgery is highly important. There is no standard large animal model for nerve repair studies, but a number of species are used in other disciplines. The ovine animal model is established as a standard surgery model regarding orthopedic questions to investigate treatment options in arthrosis, osteoarthritis or damage of cruciate ligaments [5759]. Porcine animals are predominantly used in trauma and intensive care research [6062] and feline models are used as ear, nose and throat (ENT) surgical standard models [63]. Artificial nerve conduits have been used successfully in different ovine animal models [6466], yet there is no standard animal model on large peripheral nerve surgery. Table 3 gives an overview of study designs in peripheral nerve surgery using large animal models.
Table 3
Large animal models in peripheral nerve surgery
 
Defect size (cm)
Nerve
Model
Implant
Time
Result
Author
Large animal models in nerve surgery without cell transplantation or immunosuppression
1
8.0
Mesian nerve
Sheep
Autolog vs. allograft
6 and 10 M
No regeneration in allografts. Good results in autografts
Strasberg et al. [65]
2
8.0
Ulnar nerve
Swine
Autograft vs. allograft
6 and 10 M
Autograft significant superior to allograft
Atchabahian et al. [67]
3
7.0
Median nerve
Sheep
Autograft
6 and 9 M
Slightly decreased results in electrophysiology compared to untreated controls
Forden et al. [66]
4
5.0
Sciatic nerve
Cat
Freeze-dried alginate gel covered by polyglycolic acid mesh
7 M
Recovery through alginate gel is possible
Suzuki et al. [68]
5
5.0
Sciatic nerve
Cat
Freeze-dried alginate gel covered by polyglycolic acid mesh
10 M
Good regeneration without tubular structure
Sufan et al. [71]
6
8.0
Peroneal nerve
Dog
PGA-collagen-laminin
12 M
Regeneration throughout the conduit with some differences in histological appearance
Matsumoto et al. [72]
7
2.0, 5.0
Ulnar nerve
Primate
Maxon® collagen graft
14 M
Maxon® is superior compared to controls on defects sizes <2.0
Mackinson et al. [81]
8
6.0
Tibial nerve
Sheep
Vein filled with spider silk vs. autograft
10 M
Full functional recovery
Radtke et al. [64]
Large animal models in nerve surgery with immunosuppression or cell transplantation/co-factors
9
4.0
Ulnar nerve
Primate
Allografts with MSC
6 M
Good recovery but inferior to isografts and Schwann cell seeded nerve grafts
Hu et al. [74]
10
8.0
Peroneal nerve
Sheep
Autograft, allograft + cyclosporine A
35 and 47 d
Major side effects due immunosuppression
Matsuyama et al. [99]
11
4.0
Ulnar nerve
Primate
Autograft, allograft + FK506
8 M
Comparable results of autograft and allograft + Fk506
Auba et al. [75]
12
5.0
Ulnar nerve
Primate
Autograft, allograft + anti-CD40 ligand
4 and 6 M
Anti-CD40 ligand monoclonal antibody can improve regeneration
Brenner et al. [73]
13
5.0
Ulnar nerve
Swine
Allografts + MHC Schwann cells + controls without cells
20 W
Good results regarding cold preservation of allografts
Brenner et al. [101]
14
5.0
Peroneal nerve
Dog
Allograft, autograft + bFGF
1 and 3 M
FGF can improve regeneration, 5.0 are possible without immunosuppression
Ide et al. [103]
15
8.0
Ulnar nerve
Swine
Autograft, allograft + FK506
24 W
FK506 improve regeneration, major side effects due to immunosuppression
Jensen et al. [102]
M month, W week, d day
In 1996 Strasberg et al. [65] used an ovine animal model to investigate an 8.0 cm peripheral nerve defect on median nerves surgically treated with either autologous nerve graft, nerve allograft, cold-preserved autograft or cold preserved allografts, thus introducing the ovine animal as a large animal model for peripheral nerve surgery. Analyses were performed after 6 and 10 months post-surgery. They concluded that both fresh autograft and cold-preserved autograft demonstrated nerve regeneration across the 8.0 cm nerve gap. Unfortunately, fresh allograft and cold preserved allografts demonstrated insufficient axonal regeneration.
Subsequently a study in the porcine animal model was performed with slightly modified parameters. In 12 adult swine an 8.0 cm bilateral ulnar nerve defect was induced and treated with an autologous nerve graft or a nerve allograft [67]. After 6 and 10 months nerves were harvested and histomorphometry revealed nerve regeneration and remyelinization in the autologous nerve transplant group. As previously shown in the ovine animal model [65] the regeneration of the allograft treated group remained unsatisfactory.
About 15 years later experiments in sheep were performed and reintroduced the sheep as a large animal model for nerve repair surgery [66]. Results from a median nerve defect in the sheep with a defect length of 5.0 cm coapted with a 7.0 cm autologous radial nerve graft was explored. After 6 and 9 months of surgery animals were euthanized and nerve grafts were analyzed regarding axonal regeneration. Electrophysiology revealed an insignificant difference between autograft and control. Results demonstrated regeneration throughout the entire graft length. Morphometric analyses showed axons with significantly smaller diameter in autologous nerve graft after 6 and 9 months compared to control, which is characteristic of regenerated axons. Extrafascicular and endoneural tissue were distinct. Forden et al. were able to show that the median nerve in sheep was an adequate model for peripheral nerve surgery. Thus, they were able to reproduce the results of Strasberg et al. [65] with an improved surgical and methodological approach.
Another study investigating freeze dried alginate gel on 5.0 cm sciatic nerve defects in cat [68]. Alginate was previously used in chronic wound healing and wound dressing [69, 70]. After 3 and 13 weeks compound muscle action potentials showed ongoing regeneration in the implant group with increasing latency and amplitudes after 13 weeks. Histomorphological analysis using electron microscopy (EM) demonstrated axon regeneration throughout the conduit 7 months following surgery. As already shown in the studies by Forden et al. [66] myelinated axons appeared with a significantly smaller diameter compared to controls.
In order to improve alginate as a material in peripheral nerve surgery, the experiments were repeated by Sufan et al. on a 5.0 cm sciatic defect model in feline animals two years later with a completely modified implant design [71]. A tubulated implant constructed from freeze dried alginate and polyglycolic acid (PGA) was compared to a non-tubulated alginate only graft. EM revealed regeneration in both graft groups independent of the tubulated and non-tubulated graft 10 months after surgery. As expected from prior studies from Suzuki et al. [68] and Forden et al. [66], axons appeared myelinated and smaller in diameter than in normal nerve tissue. Suzuki et al. were able to demonstrate that regeneration within the two different nerve grafts is comparable.
Following an entirely different approach Matsumoto et al. performed an experiment in 16 adult canine animals on an 8.0 cm peroneal nerve defect repaired with a polyglycolic based conduit filled with laminin coated collagen fibers [72]. The peroneal nerve was harvested 12 months post-surgery. Monthly recorded compound muscle action potentials (CMAPs) demonstrated incomplete regeneration throughout the 8.0 cm artificial nerve graft. EM showed that regeneration is still ongoing though decreasing axon diameter and number of myelinated axons. Immunohistochemistry could demonstrate growing neurofilament positive nerve fibers in the distal nerve stump. Matsumoto et al. were able to develop a suitable PGA-collagen implant for surgical intervention on large peripheral nerve defects.
Several experiments in nerve surgery have been carried out in non-human primates [7375]. Primates are regarded as an important translational animal model in medicine for some situations [76, 77]. Nonetheless the cost and ethical concerns limit their use [7880]. Mackinson et al. compared a synthetic glycolide trimethylene carbonate nerve graft with a collagen based conduit in a 2.0 cm and 5.0 cm ulnar/radial nerve defect in non-human primates [81]. The 14 month post-surgery results demonstrate a complete and excellent regeneration through the 2.0 cm grafts. Regeneration over a 5.0 cm nerve gap was significantly better in glycolide trimethylene carbonate conduits than in collagen based grafts but still poor in comparison to the 2.0 cm grafts.
Radtke et al. used a nerve implant constructed out of spider silk gained from the species Nephilla inside of a decellularized vessel for reconstruction of a 6.0 cm tibial nerve defect in ovine animals [64]. The spider silk nerve implant shows good results in electrophysiology and histological examination 10 months post-surgery. Results indicates that nerve regeneration through the spider silk nerve implant is at least as effective as autologous nerve grafts. The spider silk conduit was designed and prior to application in large animal model extensively tested in vitro and in small animal models in vivo [8286]. Especially results of the current in vitro investigation of the spider silk based nerve implant emphasize that the implant might be suitable for nerve reconstruction on defect sides extending 6.0 cm in length [82]. Outcome of previous the in vitro study are currently being reevaluated in the ovine animal model [82]. Results of these in vivo investigation are pending. Although spider silk is a natural material it appears to be suitable for different surgical approaches due to its mechanical and thermal properties [8793]. Radtke et al. briefly reviewed the use of silk in relation to peripheral nerve reconstruction and regeneration and discussed future material optimization and translation to daily medical practice [94].

Grafting with cell transplantation on long gaped nerve defects

Mesenchymal stem cells (MSCs) play a major role in proliferation and differentiation during regenerating processes in tissue e. g. fat, bone and cartilage. An interesting property of MSCs with regard to nerve regeneration is the interaction they have with the innate and adaptive immune system as well as the down regulation of proinflammatory cytokines in damaged tissue [95].
Jun Hu et al. used this approach to investigate an allograft on a 4.0 cm ulnar nerve defect in non-human primates [74]. Allograft pre-seeded with MSCs were compared to Schwann cell seeded allograft, acellular autografts and acellular controls. After 6 months, the results indicated that regeneration in allografts with MSCs are better than in acellular controls, but still inferior to grafts pre-seeded with Schwann cells and isografts.

Grafting with immunosuppression

Immunosuppression is needed to prevent foreign body reactions and rejections of implants [96]. Allografts have been reported to precipitate immune reactions resulting in implant rejection [97]. Cyclosporine A usually used after organ transplantation [98] was used to prevent allograft rejection in ovine animals on a 5.0 cm median nerve defect grafted with an 8.0 cm allograft [99]. Immunosuppression led to severe bacterial infections resulting in controlled experimental truncation. However, the results indicated that immunosuppression can prevent tissue rejection of allografts.
On the other hand, FK506 (tacrolimus) was able to demonstrate enhanced nerve regeneration in allografts in small rodents [100]. Auba et al. used a nonhuman primate model with immunosuppression [75]. A 4.0 cm nerve defect in nonhuman primate ulnar nerve was inducted and coapted with either allograft under immunosuppression (2 months only) with FK506 or isograft. Apart from slightly elevated nerve conduction velocities in the autologous control no significant difference was observed in the histological analysis 8 months after surgery. The investigators reported that this could indicate tissue rejection within the allograft.
Instead of using a systemic immunosuppression to prevent allograft rejection Brenner et al. used anti-CD 40 ligand monoclonal antibody on a 5.0 cm ulnar nerve defect in non-human primates [73]. To investigate the full influence of anti-CD 40 ligand monoclonal antibody a skin allograft was transplanted additionally to the nerve allograft in one group. Results demonstrated that anti-CD 40 ligand monoclonal antibody is able to avoid foreign body reactions that lead to tissue rejection ensuring equal regeneration compared to autografts. Skin grafts were rejected and indicate partial rejection of nerve allografts supposed to be caused by overwhelming foreign body reactions. A subsequent approach by Brenner et al. was to use pre-seeded allograft with MHC matched Schwann cells to downregulate foreign body reactions in porcine animals with and without pre-operative ultraviolet-B donor alloantigen injections [101]. Therefore a 5.0 cm ulnar nerve allograft model was designed. Allografts + MHC-Schwann cells resulted in excellent nerve regeneration compared to controls. Influence of UV-B donor alloantigen injections is not fully understood.
In 2005 Jensen et al. performed an experiment in 8 outbreed swine [102]. An 8.0 cm ulnar nerve defects was reconstructed with an autologous nerve graft and allografts. Six operated animals received an FK506 injection every 14 days to maintain immunosuppression. Three FK506 treated animals deceased before experimental endpoints suffering pulmonary abscess, infarcted bowel or cardiac arrest. Results demonstrate a comparable regeneration in FK506 allografts compared to untreated autografts. Autografts from animals treated with FK506 showed a slight improved regeneration compared to autologous controls.

Grafting without immunosuppression

Brenner et al. [73, 101], Matsuyama et al. [99] and several other investigators demonstrated that allografting without immunosuppression leads to tissue rejection. Ide et al. tested an acellular allograft in combination with fibroblast growth factor (FGF) on a 5.0 cm peroneal nerve defect in a canine animal model [103]. They report that nerve regeneration throughout the allograft is possible without immunosuppression. However, the regeneration is less than that obtained from autografts. Clearly the use of immunosuppression is an important issue for use of autografts and research in this area is critical for non-autologous biosynthetic graft construction and use in clinical studies for nerve repair.

Conclusion

Translation to clinical practice

Ongoing developments in peripheral nerve surgery are auspicious. Allografts are nearly a perfect alternative to the current gold standard technique, especially because immunosuppression is not indicated anymore to avoid a foreign body reaction. Nerve grafting with cell suspensions e. g. with Schwann cells shows promising results on small defect sizes in animals but is limited on translation to human organism by highly regulated local laws for transplantation of human stem cells. Muscle grafts and mono channeled nerve grafts are not appropriate for reconstruction of long gaped peripheral nerve defects. Nerve reconstruction with biomaterials like spider silk are as good as the autologous gold standard. Research is still ongoing and transfer to the human organism is pending.

Research approaches

A literature search demonstrated that scientific approaches and animal models in peripheral nerve surgery are extremely versatile with the result of limited and impeded possibilities of comparison. Scientists in the field of peripheral nerve surgery should focus on comparable approaches regarding animal models and methods to produce comparable and competitive results. Nevertheless most reviewed approaches are promising regarding future clinical translation. Especially the transplantation of autologous donor cells and factors within artificial and non-artificial nerve grafts might be advantageous in future settings.

Effect on daily medical practice

The autologous nerve graft is still and with good reason the gold standard for reconstruction of peripheral nerve tissue. Nevertheless autologous donor material is highly limited in number and often mismatch the nerve defects of the reconstruction side. Especially multiple long gaped plexus injuries are challenging during reconstruction. Furthermore the donor side morbidity after autograft harvest can lead to post-operative complications e. g. with neuroma formation, permanent sensitivity loss and adherent scars. Obviously autologous donor harvest is limited. In those surgical settings with limited availability of donor tissue the use of artificial nerve grafts should be recommended for peripheral nerve defects <3.0 cm. Both artificial nerve graft e. g. NeuroGen®, Neurotube®, Neurolac® and nerve allografts e. g. AxoGen® Avance can be recommended as alternative approaches for nerve reconstruction after literature analysis. Translational and alternative approaches for the reconstruction of long gaped defect sides are desperately needed. Especially the cell free approaches including allografts or spider silk nerve implants could be a contemporary and sufficient transfer technology beside neurotization until cell based procedures are safe and a statutory basis is set.

Funding

This work was funded by the Volkswagen Foundation.

Conflict of interest

T. Kornfeld, P.M. Vogt, and C. Radtke declare that they have no competing interests.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

Abo für kostenpflichtige Inhalte

Literatur
1.
Zurück zum Zitat Kretschmer T, Antoniadis G, Braun V, Rath SA, Richter HP. Evaluation of iatrogenic lesions in 722 surgically treated cases of peripheral nerve trauma. J Neurosurg. 2001;94(6):905–12.PubMed Kretschmer T, Antoniadis G, Braun V, Rath SA, Richter HP. Evaluation of iatrogenic lesions in 722 surgically treated cases of peripheral nerve trauma. J Neurosurg. 2001;94(6):905–12.PubMed
2.
Zurück zum Zitat McGeorge D, Sturzenegger M, Buchler U. Tibial nerve mistakenly used as a tendon graft. Reports of three cases. Bone Joint J. 1992;74-B(3):365–6. McGeorge D, Sturzenegger M, Buchler U. Tibial nerve mistakenly used as a tendon graft. Reports of three cases. Bone Joint J. 1992;74-B(3):365–6.
3.
Zurück zum Zitat Weber RV, Mackinnon SE. Median nerve mistaken for palmaris longus tendon: restoration of function with sensory nerve transfers. Hand (N Y). 2007;2(1):1–4. Weber RV, Mackinnon SE. Median nerve mistaken for palmaris longus tendon: restoration of function with sensory nerve transfers. Hand (N Y). 2007;2(1):1–4.
4.
Zurück zum Zitat Renton T, Adey-Viscuso D, Meechan JG, Yilmaz Z. Trigeminal nerve injuries in relation to the local anaesthesia in mandibular injections. Br Dent J. 2010;209(9):E15–E15.PubMed Renton T, Adey-Viscuso D, Meechan JG, Yilmaz Z. Trigeminal nerve injuries in relation to the local anaesthesia in mandibular injections. Br Dent J. 2010;209(9):E15–E15.PubMed
5.
Zurück zum Zitat Sawyer RJ, Richmond MN, Hickey JD, Jarrratt JA. Peripheral nerve injuries associated with anaesthesia. Anaesthesia. 2000;55(10):980–9991.PubMed Sawyer RJ, Richmond MN, Hickey JD, Jarrratt JA. Peripheral nerve injuries associated with anaesthesia. Anaesthesia. 2000;55(10):980–9991.PubMed
6.
Zurück zum Zitat Jung Kim H, Hyun Park S. Sciatic nerve injection injury. J Int Med Res. 2014;42(4):887–97.PubMed Jung Kim H, Hyun Park S. Sciatic nerve injection injury. J Int Med Res. 2014;42(4):887–97.PubMed
7.
Zurück zum Zitat Noble J, Munro CA, Prasad VS, Midha R. Analysis of upper and lower extremity peripheral nerve injuries in a population of patients with multiple injuries. J Trauma. 1998;45(1):116–22.PubMed Noble J, Munro CA, Prasad VS, Midha R. Analysis of upper and lower extremity peripheral nerve injuries in a population of patients with multiple injuries. J Trauma. 1998;45(1):116–22.PubMed
9.
Zurück zum Zitat Siemionow M, Brzezicki G. Chapter 8: current techniques and concepts in peripheral nerve repair. Int Rev Neurobiol. 2009;87:141–72.PubMed Siemionow M, Brzezicki G. Chapter 8: current techniques and concepts in peripheral nerve repair. Int Rev Neurobiol. 2009;87:141–72.PubMed
10.
Zurück zum Zitat Ray WZ, Mackinnon SE. Management of nerve gaps: autografts, allografts, nerve transfers, and end-to-side neurorrhaphy. Exp Neurol. 2010;223(1):77–85.PubMed Ray WZ, Mackinnon SE. Management of nerve gaps: autografts, allografts, nerve transfers, and end-to-side neurorrhaphy. Exp Neurol. 2010;223(1):77–85.PubMed
11.
Zurück zum Zitat Arslantunali D, Dursun T, Yucel D, Hasirci N, Hasirci V. Peripheral nerve conduits: technology update. Med Devices (Auckl). 2014;7:405–24. Arslantunali D, Dursun T, Yucel D, Hasirci N, Hasirci V. Peripheral nerve conduits: technology update. Med Devices (Auckl). 2014;7:405–24.
12.
Zurück zum Zitat Kehoe S, Zhang XF, Boyd D. FDA approved guidance conduits and wraps for peripheral nerve injury: a review of materials and efficacy. Injury. 2012;43(5):553–72.PubMed Kehoe S, Zhang XF, Boyd D. FDA approved guidance conduits and wraps for peripheral nerve injury: a review of materials and efficacy. Injury. 2012;43(5):553–72.PubMed
13.
Zurück zum Zitat Wangensteen KJ, Kalliainen LK. Collagen tube conduits in peripheral nerve repair: a retrospective analysis. Hand (N Y). 2010;5(3):273–7. Wangensteen KJ, Kalliainen LK. Collagen tube conduits in peripheral nerve repair: a retrospective analysis. Hand (N Y). 2010;5(3):273–7.
14.
Zurück zum Zitat Lynn AK, Yannas IV, Bonfield W. Antigenicity and immunogenicity of collagen. J Biomed Mater Res Part B Appl Biomater. 2004;71(2):343–54. Lynn AK, Yannas IV, Bonfield W. Antigenicity and immunogenicity of collagen. J Biomed Mater Res Part B Appl Biomater. 2004;71(2):343–54.
15.
Zurück zum Zitat Haastert-Talini K, Geuna S, Dahlin LB, Meyer C, Stenberg L, Freier T, et al. Chitosan tubes of varying degrees of acetylation for bridging peripheral nerve defects. Biomaterials. 2013;34(38):9886–904.PubMed Haastert-Talini K, Geuna S, Dahlin LB, Meyer C, Stenberg L, Freier T, et al. Chitosan tubes of varying degrees of acetylation for bridging peripheral nerve defects. Biomaterials. 2013;34(38):9886–904.PubMed
16.
Zurück zum Zitat Marcol W, Larysz-Brysz M, Kucharska M, Niekraszewicz A, Slusarczyk W, Kotulska K, et al. Reduction of post-traumatic neuroma and epineural scar formation in rat sciatic nerve by application of microcrystallic chitosan. Microsurgery. 2011;31(8):642–9.PubMed Marcol W, Larysz-Brysz M, Kucharska M, Niekraszewicz A, Slusarczyk W, Kotulska K, et al. Reduction of post-traumatic neuroma and epineural scar formation in rat sciatic nerve by application of microcrystallic chitosan. Microsurgery. 2011;31(8):642–9.PubMed
17.
Zurück zum Zitat Duda S, Dreyer L, Behrens P, Wienecke S, Chakradeo T, Glasmacher B, et al. Outer electrospun polycaprolactone shell induces massive foreign body reaction and impairs axonal regeneration through 3D multichannel chitosan nerve guides. Biomed Res Int. 2014;2014:835269.PubMedPubMedCentral Duda S, Dreyer L, Behrens P, Wienecke S, Chakradeo T, Glasmacher B, et al. Outer electrospun polycaprolactone shell induces massive foreign body reaction and impairs axonal regeneration through 3D multichannel chitosan nerve guides. Biomed Res Int. 2014;2014:835269.PubMedPubMedCentral
18.
Zurück zum Zitat Navissano M, Malan F, Carnino R, Battiston B. Neurotube® for facial nerve repair. Microsurgery. 2005;25(4):268–2271.PubMed Navissano M, Malan F, Carnino R, Battiston B. Neurotube® for facial nerve repair. Microsurgery. 2005;25(4):268–2271.PubMed
19.
Zurück zum Zitat Duncan SF, Kakinoki R, Rizzo M, Kang W. Extrusion of a Neurotube: a case report. Ochsner J. 2015;15(2):191–2.PubMedPubMedCentral Duncan SF, Kakinoki R, Rizzo M, Kang W. Extrusion of a Neurotube: a case report. Ochsner J. 2015;15(2):191–2.PubMedPubMedCentral
20.
Zurück zum Zitat den Dunnen WF, van der Lei B, Schakenraad JM, Stokroos I, Blaauw E, Bartels H, et al. Poly(DL-lactide-epsilon-caprolactone) nerve guides perform better than autologous nerve grafts. Microsurgery. 1996;17(7):348–57. den Dunnen WF, van der Lei B, Schakenraad JM, Stokroos I, Blaauw E, Bartels H, et al. Poly(DL-lactide-epsilon-caprolactone) nerve guides perform better than autologous nerve grafts. Microsurgery. 1996;17(7):348–57.
22.
Zurück zum Zitat Whitlock EL, Tuffaha SH, Luciano JP, Yan Y, Hunter DA, Magill CK, et al. Processed allografts and type I collagen conduits for repair of peripheral nerve gaps. Muscle Nerve. 2009;39(6):787–99.PubMed Whitlock EL, Tuffaha SH, Luciano JP, Yan Y, Hunter DA, Magill CK, et al. Processed allografts and type I collagen conduits for repair of peripheral nerve gaps. Muscle Nerve. 2009;39(6):787–99.PubMed
23.
Zurück zum Zitat Brooks DN, Weber RV, Chao JD, Rinker BD, Zoldos J, Robichaux MR, et al. Processed nerve allografts for peripheral nerve reconstruction: a multicenter study of utilization and outcomes in sensory, mixed, and motor nerve reconstructions. Microsurgery. 2012;32(1):1–14.PubMed Brooks DN, Weber RV, Chao JD, Rinker BD, Zoldos J, Robichaux MR, et al. Processed nerve allografts for peripheral nerve reconstruction: a multicenter study of utilization and outcomes in sensory, mixed, and motor nerve reconstructions. Microsurgery. 2012;32(1):1–14.PubMed
24.
Zurück zum Zitat Rinker B, Zoldos J, Weber RV, Ko J, Thayer W, Greenberg J, et al. Use of processed nerve allografts to repair nerve injuries greater than 25 mm in the hand. Ann Plast Surg. 2017;78(6S Suppl 5):S292–S5.PubMed Rinker B, Zoldos J, Weber RV, Ko J, Thayer W, Greenberg J, et al. Use of processed nerve allografts to repair nerve injuries greater than 25 mm in the hand. Ann Plast Surg. 2017;78(6S Suppl 5):S292–S5.PubMed
25.
Zurück zum Zitat Kaplan HM, Mishra P, Kohn J. The overwhelming use of rat models in nerve regeneration research may compromise designs of nerve guidance conduits for humans. J Mater Sci Mater Med. 2015;26(8):226.PubMedPubMedCentral Kaplan HM, Mishra P, Kohn J. The overwhelming use of rat models in nerve regeneration research may compromise designs of nerve guidance conduits for humans. J Mater Sci Mater Med. 2015;26(8):226.PubMedPubMedCentral
26.
Zurück zum Zitat Bozkurt A, Boecker A, Tank J, Altinova H, Deumens R, Dabhi C, et al. Efficient bridging of 20 mm rat sciatic nerve lesions with a longitudinally micro-structured collagen scaffold. Biomaterials. 2016;75:112–22.PubMed Bozkurt A, Boecker A, Tank J, Altinova H, Deumens R, Dabhi C, et al. Efficient bridging of 20 mm rat sciatic nerve lesions with a longitudinally micro-structured collagen scaffold. Biomaterials. 2016;75:112–22.PubMed
27.
Zurück zum Zitat Kusaba H, Terada-Nakaishi M, Wang W, Itoh S, Nozaki K, Nagai A, et al. Comparison of nerve regenerative efficacy between decellularized nerve graft and nonwoven chitosan conduit. Biomed Mater Eng. 2016;27(1):75–85.PubMed Kusaba H, Terada-Nakaishi M, Wang W, Itoh S, Nozaki K, Nagai A, et al. Comparison of nerve regenerative efficacy between decellularized nerve graft and nonwoven chitosan conduit. Biomed Mater Eng. 2016;27(1):75–85.PubMed
28.
Zurück zum Zitat Ozkan HS, Karatas Silistreli O, Ergur B, Irkoren S. Repairing peripheral nerve defects by vein grafts filled with adipose tissue derived stromal vascular fraction: an experimental study in rats. Ulus Travma Acil Cerrahi Derg. 2016;22(1):7–11.PubMed Ozkan HS, Karatas Silistreli O, Ergur B, Irkoren S. Repairing peripheral nerve defects by vein grafts filled with adipose tissue derived stromal vascular fraction: an experimental study in rats. Ulus Travma Acil Cerrahi Derg. 2016;22(1):7–11.PubMed
29.
Zurück zum Zitat Fitzgerald TA. Comparison of research cost: man—primate animal—other animal models. J Med Primatol. 1983;12(3):138–45.PubMed Fitzgerald TA. Comparison of research cost: man—primate animal—other animal models. J Med Primatol. 1983;12(3):138–45.PubMed
30.
Zurück zum Zitat Varejao AS, Meek MF, Ferreira AJ, Patricio JA, Cabrita AM. Functional evaluation of peripheral nerve regeneration in the rat: walking track analysis. J Neurosci Methods. 2001;108(1):1–9.PubMed Varejao AS, Meek MF, Ferreira AJ, Patricio JA, Cabrita AM. Functional evaluation of peripheral nerve regeneration in the rat: walking track analysis. J Neurosci Methods. 2001;108(1):1–9.PubMed
31.
Zurück zum Zitat Werdin F, Grussinger H, Jaminet P, Kraus A, Manoli T, Danker T, et al. An improved electrophysiological method to study peripheral nerve regeneration in rats. J Neurosci Methods. 2009;182(1):71–7.PubMed Werdin F, Grussinger H, Jaminet P, Kraus A, Manoli T, Danker T, et al. An improved electrophysiological method to study peripheral nerve regeneration in rats. J Neurosci Methods. 2009;182(1):71–7.PubMed
32.
Zurück zum Zitat Neubauer D, Graham JB, Muir D. Chondroitinase treatment increases the effective length of acellular nerve grafts. Exp Neurol. 2007;207(1):163–70.PubMedPubMedCentral Neubauer D, Graham JB, Muir D. Chondroitinase treatment increases the effective length of acellular nerve grafts. Exp Neurol. 2007;207(1):163–70.PubMedPubMedCentral
33.
Zurück zum Zitat Saheb-Al-Zamani M, Yan Y, Farber SJ, Hunter DA, Newton P, Wood MD, et al. Limited regeneration in long acellular nerve allografts is associated with increased Schwann cell senescence. Exp Neurol. 2013;247:165–77.PubMed Saheb-Al-Zamani M, Yan Y, Farber SJ, Hunter DA, Newton P, Wood MD, et al. Limited regeneration in long acellular nerve allografts is associated with increased Schwann cell senescence. Exp Neurol. 2013;247:165–77.PubMed
34.
Zurück zum Zitat Koller R, Rab M, Todoroff BP, Neumayer C, Haslik W, Stohr HG, et al. The influence of the graft length on the functional and morphological result after nerve grafting: an experimental study in rabbits. Br J Plast Surg. 1997;50(8):609–14.PubMed Koller R, Rab M, Todoroff BP, Neumayer C, Haslik W, Stohr HG, et al. The influence of the graft length on the functional and morphological result after nerve grafting: an experimental study in rabbits. Br J Plast Surg. 1997;50(8):609–14.PubMed
35.
Zurück zum Zitat Mligiliche N, Tabata Y, Endoh K, Ide C. Peripheral nerve regeneration through a long detergent-denatured muscle autografts in rabbits. Neuroreport. 2001;12(8):1719–22.PubMed Mligiliche N, Tabata Y, Endoh K, Ide C. Peripheral nerve regeneration through a long detergent-denatured muscle autografts in rabbits. Neuroreport. 2001;12(8):1719–22.PubMed
36.
Zurück zum Zitat Hems TE, Glasby MA. The limit of graft length in the experimental use of muscle grafts for nerve repair. J Hand Surg Br. 1993;18(2):165–70.PubMed Hems TE, Glasby MA. The limit of graft length in the experimental use of muscle grafts for nerve repair. J Hand Surg Br. 1993;18(2):165–70.PubMed
37.
Zurück zum Zitat Zhang F, Blain B, Beck J, Zhang J, Chen Z, Chen ZW, et al. Autogenous venous graft with one-stage prepared Schwann cells as a conduit for repair of long segmental nerve defects. J Reconstr Microsurg. 2002;18(4):295–300.PubMed Zhang F, Blain B, Beck J, Zhang J, Chen Z, Chen ZW, et al. Autogenous venous graft with one-stage prepared Schwann cells as a conduit for repair of long segmental nerve defects. J Reconstr Microsurg. 2002;18(4):295–300.PubMed
38.
Zurück zum Zitat Strauch B, Rodriguez DM, Diaz J, Yu HL, Kaplan G, Weinstein DE. Autologous Schwann cells drive regeneration through a 6-cm autogenous venous nerve conduit. J Reconstr Microsurg. 2001;17(8):589–95. discussion 596–7.PubMed Strauch B, Rodriguez DM, Diaz J, Yu HL, Kaplan G, Weinstein DE. Autologous Schwann cells drive regeneration through a 6-cm autogenous venous nerve conduit. J Reconstr Microsurg. 2001;17(8):589–95. discussion 596–7.PubMed
39.
Zurück zum Zitat Strauch B, Ferder M, Lovelle-Allen S, Moore K, Kim DJ, Llena J. Determining the maximal length of a vein conduit used as an interposition graft for nerve regeneration. J Reconstr Microsurg. 1996;12(8):521–7.PubMed Strauch B, Ferder M, Lovelle-Allen S, Moore K, Kim DJ, Llena J. Determining the maximal length of a vein conduit used as an interposition graft for nerve regeneration. J Reconstr Microsurg. 1996;12(8):521–7.PubMed
40.
Zurück zum Zitat Sinis N, Schaller HE, Becker ST, Schlosshauer B, Doser M, Roesner H, et al. Long nerve gaps limit the regenerative potential of bioartificial nerve conduits filled with Schwann cells. Restor Neurol Neurosci. 2007;25(2):131–41.PubMed Sinis N, Schaller HE, Becker ST, Schlosshauer B, Doser M, Roesner H, et al. Long nerve gaps limit the regenerative potential of bioartificial nerve conduits filled with Schwann cells. Restor Neurol Neurosci. 2007;25(2):131–41.PubMed
41.
Zurück zum Zitat Kirk AD. Crossing the bridge: large animal models in translational transplantation research. Immunol Rev. 2003;196:176–96.PubMed Kirk AD. Crossing the bridge: large animal models in translational transplantation research. Immunol Rev. 2003;196:176–96.PubMed
42.
Zurück zum Zitat Konofaos P, Ver Halen JP. Nerve repair by means of tubulization: past, present, future. J Reconstr Microsurg. 2013;29(3):149–64.PubMed Konofaos P, Ver Halen JP. Nerve repair by means of tubulization: past, present, future. J Reconstr Microsurg. 2013;29(3):149–64.PubMed
43.
Zurück zum Zitat Isaacs J, Browne T. Overcoming short gaps in peripheral nerve repair: conduits and human acellular nerve allograft. Hand (N Y). 2014;9(2):131–7. Isaacs J, Browne T. Overcoming short gaps in peripheral nerve repair: conduits and human acellular nerve allograft. Hand (N Y). 2014;9(2):131–7.
44.
Zurück zum Zitat Meek MF, Coert JH. Clinical use of nerve conduits in peripheral-nerve repair: review of the literature. J Reconstr Microsurg. 2002;18(2):97–109.PubMed Meek MF, Coert JH. Clinical use of nerve conduits in peripheral-nerve repair: review of the literature. J Reconstr Microsurg. 2002;18(2):97–109.PubMed
45.
Zurück zum Zitat Fawcett JW, Keynes RJ. Muscle basal lamina: a new graft material for peripheral nerve repair. J Neurosurg. 1986;65(3):354–63.PubMed Fawcett JW, Keynes RJ. Muscle basal lamina: a new graft material for peripheral nerve repair. J Neurosurg. 1986;65(3):354–63.PubMed
46.
Zurück zum Zitat Battiston B, Tos P, Cushway TR, Geuna S. Nerve repair by means of vein filled with muscle grafts I. Clinical results. Microsurgery. 2000;20(1):32–6.PubMed Battiston B, Tos P, Cushway TR, Geuna S. Nerve repair by means of vein filled with muscle grafts I. Clinical results. Microsurgery. 2000;20(1):32–6.PubMed
47.
Zurück zum Zitat Norris R, Glasby M, Gattuso J, Bowden R. Peripheral nerve repair in humans using muscle autografts. A new technique. Bone Joint J. 1988;70-B(4):530–3. Norris R, Glasby M, Gattuso J, Bowden R. Peripheral nerve repair in humans using muscle autografts. A new technique. Bone Joint J. 1988;70-B(4):530–3.
48.
Zurück zum Zitat Radtke C, Akiyama Y, Lankford KL, Vogt PM, Krause DS, Kocsis JD. Integration of engrafted Schwann cells into injured peripheral nerve: axonal association and nodal formation on regenerated axons. Neurosci Lett. 2005;387(2):85–9.PubMedPubMedCentral Radtke C, Akiyama Y, Lankford KL, Vogt PM, Krause DS, Kocsis JD. Integration of engrafted Schwann cells into injured peripheral nerve: axonal association and nodal formation on regenerated axons. Neurosci Lett. 2005;387(2):85–9.PubMedPubMedCentral
49.
Zurück zum Zitat Frostick SP, Yin Q, Kemp GJ. Schwann cells, neurotrophic factors, and peripheral nerve regeneration. Microsurgery. 1998;18(7):397–405.PubMed Frostick SP, Yin Q, Kemp GJ. Schwann cells, neurotrophic factors, and peripheral nerve regeneration. Microsurgery. 1998;18(7):397–405.PubMed
50.
Zurück zum Zitat Scheib J, Hoke A. Advances in peripheral nerve regeneration. Nat Rev Neurol. 2013;9(12):668–76.PubMed Scheib J, Hoke A. Advances in peripheral nerve regeneration. Nat Rev Neurol. 2013;9(12):668–76.PubMed
51.
Zurück zum Zitat Hadlock TA, Sundback CA, Hunter DA, Vacanti JP, Cheney ML. A new artificial nerve graft containing rolled Schwann cell monolayers. Microsurgery. 2001;21(3):96–101.PubMed Hadlock TA, Sundback CA, Hunter DA, Vacanti JP, Cheney ML. A new artificial nerve graft containing rolled Schwann cell monolayers. Microsurgery. 2001;21(3):96–101.PubMed
52.
Zurück zum Zitat Hood B, Levene HB, Levi AD. Transplantation of autologous Schwann cells for the repair of segmental peripheral nerve defects. Neurosurg Focus. 2009;26(2):1–9. Hood B, Levene HB, Levi AD. Transplantation of autologous Schwann cells for the repair of segmental peripheral nerve defects. Neurosurg Focus. 2009;26(2):1–9.
53.
Zurück zum Zitat Hadlock T, Sundback C, Hunter D, Cheney M, Vacanti JP. A polymer foam conduit seeded with Schwann cells promotes guided peripheral nerve regeneration. Tissue Eng. 2000;6(2):119–27.PubMed Hadlock T, Sundback C, Hunter D, Cheney M, Vacanti JP. A polymer foam conduit seeded with Schwann cells promotes guided peripheral nerve regeneration. Tissue Eng. 2000;6(2):119–27.PubMed
54.
Zurück zum Zitat Mosahebi A, Fuller P, Wiberg M, Terenghi G. Effect of allogeneic Schwann cell transplantation on peripheral nerve regeneration. Exp Neurol. 2002;173(2):213–23.PubMed Mosahebi A, Fuller P, Wiberg M, Terenghi G. Effect of allogeneic Schwann cell transplantation on peripheral nerve regeneration. Exp Neurol. 2002;173(2):213–23.PubMed
55.
Zurück zum Zitat Rodriguez FJ, Verdu E, Ceballos D, Navarro X. Nerve guides seeded with autologous schwann cells improve nerve regeneration. Exp Neurol. 2000;161(2):571–84.PubMed Rodriguez FJ, Verdu E, Ceballos D, Navarro X. Nerve guides seeded with autologous schwann cells improve nerve regeneration. Exp Neurol. 2000;161(2):571–84.PubMed
56.
Zurück zum Zitat Ansselin AD, Fink T, Davey DF. Peripheral nerve regeneration through nerve guides seeded with adult Schwann cells. Neuropathol Appl Neurobiol. 1997;23(5):387–3398.PubMed Ansselin AD, Fink T, Davey DF. Peripheral nerve regeneration through nerve guides seeded with adult Schwann cells. Neuropathol Appl Neurobiol. 1997;23(5):387–3398.PubMed
57.
Zurück zum Zitat Florida SE, VanDusen KW, Mahalingam VD, Schlientz AJ, Wojtys EM, Wellik DM, et al. In vivo structural and cellular remodeling of engineered bone-ligament-bone constructs used for anterior cruciate ligament reconstruction in sheep. Connect Tissue Res. 2016;57(6):526–38.PubMedPubMedCentral Florida SE, VanDusen KW, Mahalingam VD, Schlientz AJ, Wojtys EM, Wellik DM, et al. In vivo structural and cellular remodeling of engineered bone-ligament-bone constructs used for anterior cruciate ligament reconstruction in sheep. Connect Tissue Res. 2016;57(6):526–38.PubMedPubMedCentral
58.
Zurück zum Zitat Egermann M, Goldhahn J, Holz R, Schneider E, Lill CA. A sheep model for fracture treatment in osteoporosis: benefits of the model versus animal welfare. Lab Anim. 2008;42(4):453–64.PubMed Egermann M, Goldhahn J, Holz R, Schneider E, Lill CA. A sheep model for fracture treatment in osteoporosis: benefits of the model versus animal welfare. Lab Anim. 2008;42(4):453–64.PubMed
59.
Zurück zum Zitat Malhotra A, Pelletier MH, Yu Y, Christou C, Walsh WR. A sheep model for cancellous bone healing. Front Surg. 2014;1:37.PubMedPubMedCentral Malhotra A, Pelletier MH, Yu Y, Christou C, Walsh WR. A sheep model for cancellous bone healing. Front Surg. 2014;1:37.PubMedPubMedCentral
60.
Zurück zum Zitat Bauman RA, Ling G, Tong L, Januszkiewicz A, Agoston D, Delanerolle N, et al. An introductory characterization of a combat-casualty-care relevant swine model of closed head injury resulting from exposure to explosive blast. J Neurotrauma. 2009;26(6):841–60.PubMed Bauman RA, Ling G, Tong L, Januszkiewicz A, Agoston D, Delanerolle N, et al. An introductory characterization of a combat-casualty-care relevant swine model of closed head injury resulting from exposure to explosive blast. J Neurotrauma. 2009;26(6):841–60.PubMed
62.
Zurück zum Zitat Xanthos TT, Balkamou XA, Stroumpoulis KI, Pantazopoulos IN, Rokas GI, Agrogiannis GD, et al. A model of hemorrhagic shock and acute lung injury in landrace-large white swine. Comp Med. 2011;61(2):158–62.PubMedPubMedCentral Xanthos TT, Balkamou XA, Stroumpoulis KI, Pantazopoulos IN, Rokas GI, Agrogiannis GD, et al. A model of hemorrhagic shock and acute lung injury in landrace-large white swine. Comp Med. 2011;61(2):158–62.PubMedPubMedCentral
63.
Zurück zum Zitat Kretzmer EA, Meltzer NE, Haenggeli CA, Ryugo DK. An animal model for cochlear implants. Arch Otolaryngol Head Neck Surg. 2004;130(5):499–508.PubMed Kretzmer EA, Meltzer NE, Haenggeli CA, Ryugo DK. An animal model for cochlear implants. Arch Otolaryngol Head Neck Surg. 2004;130(5):499–508.PubMed
64.
Zurück zum Zitat Radtke C, Allmeling C, Waldmann KH, Reimers K, Thies K, Schenk HC, et al. Spider silk constructs enhance axonal regeneration and remyelination in long nerve defects in sheep. PLoS ONE. 2011;6(2):e16990.PubMedPubMedCentral Radtke C, Allmeling C, Waldmann KH, Reimers K, Thies K, Schenk HC, et al. Spider silk constructs enhance axonal regeneration and remyelination in long nerve defects in sheep. PLoS ONE. 2011;6(2):e16990.PubMedPubMedCentral
65.
Zurück zum Zitat Strasberg SR, Mackinnon SE, Genden EM, Bain JR, Purcell CM, Hunter DA, et al. Long-segment nerve allograft regeneration in the sheep model: experimental study and review of the literature. J Reconstr Microsurg. 1996;12(8):529–37.PubMed Strasberg SR, Mackinnon SE, Genden EM, Bain JR, Purcell CM, Hunter DA, et al. Long-segment nerve allograft regeneration in the sheep model: experimental study and review of the literature. J Reconstr Microsurg. 1996;12(8):529–37.PubMed
66.
Zurück zum Zitat Forden J, Xu QG, Khu KJ, Midha R. A long peripheral nerve autograft model in the sheep forelimb. Neurosurgery. 2011;68(5):1354–62. discussion 1362.PubMed Forden J, Xu QG, Khu KJ, Midha R. A long peripheral nerve autograft model in the sheep forelimb. Neurosurgery. 2011;68(5):1354–62. discussion 1362.PubMed
67.
Zurück zum Zitat Atchabahian A, Genden EM, MacKinnon SE, Doolabh VB, Hunter DA. Regeneration through long nerve grafts in the swine model. Microsurgery. 1998;18(6):379–82.PubMed Atchabahian A, Genden EM, MacKinnon SE, Doolabh VB, Hunter DA. Regeneration through long nerve grafts in the swine model. Microsurgery. 1998;18(6):379–82.PubMed
68.
Zurück zum Zitat Suzuki Y, Tanihara M, Ohnishi K, Suzuki K, Endo K, Nishimura Y. Cat peripheral nerve regeneration across 50 mm gap repaired with a novel nerve guide composed of freeze-dried alginate gel. Neurosci Lett. 1999;259(2):75–8.PubMed Suzuki Y, Tanihara M, Ohnishi K, Suzuki K, Endo K, Nishimura Y. Cat peripheral nerve regeneration across 50 mm gap repaired with a novel nerve guide composed of freeze-dried alginate gel. Neurosci Lett. 1999;259(2):75–8.PubMed
69.
Zurück zum Zitat Boateng JS, Matthews KH, Stevens HN, Eccleston GM. Wound healing dressings and drug delivery systems: a review. J Pharm Sci. 2008;97(8):2892–923.PubMed Boateng JS, Matthews KH, Stevens HN, Eccleston GM. Wound healing dressings and drug delivery systems: a review. J Pharm Sci. 2008;97(8):2892–923.PubMed
70.
71.
Zurück zum Zitat Sufan W, Suzuki Y, Tanihara M, Ohnishi K, Suzuki K, Endo K, et al. Sciatic nerve regeneration through alginate with tubulation or nontubulation repair in cat. J Neurotrauma. 2001;18(3):329–38.PubMed Sufan W, Suzuki Y, Tanihara M, Ohnishi K, Suzuki K, Endo K, et al. Sciatic nerve regeneration through alginate with tubulation or nontubulation repair in cat. J Neurotrauma. 2001;18(3):329–38.PubMed
72.
Zurück zum Zitat Matsumoto K, Ohnishi K, Kiyotani T, Sekine T, Ueda H, Nakamura T, et al. Peripheral nerve regeneration across an 80-mm gap bridged by a polyglycolic acid (PGA)-collagen tube filled with laminin-coated collagen fibers: a histological and electrophysiological evaluation of regenerated nerves. Brain Res. 2000;868(2):315–28.PubMed Matsumoto K, Ohnishi K, Kiyotani T, Sekine T, Ueda H, Nakamura T, et al. Peripheral nerve regeneration across an 80-mm gap bridged by a polyglycolic acid (PGA)-collagen tube filled with laminin-coated collagen fibers: a histological and electrophysiological evaluation of regenerated nerves. Brain Res. 2000;868(2):315–28.PubMed
73.
Zurück zum Zitat Brenner MJ, Jensen JN, Lowe JB 3rd, Myckatyn TM, Fox IK, Hunter DA, et al. Anti-CD40 ligand antibody permits regeneration through peripheral nerve allografts in a nonhuman primate model. Plast Reconstr Surg. 2004;114(7):1802–14. discussion 1815–7.PubMed Brenner MJ, Jensen JN, Lowe JB 3rd, Myckatyn TM, Fox IK, Hunter DA, et al. Anti-CD40 ligand antibody permits regeneration through peripheral nerve allografts in a nonhuman primate model. Plast Reconstr Surg. 2004;114(7):1802–14. discussion 1815–7.PubMed
74.
Zurück zum Zitat Hu J, Zhu QT, Liu XL, Xu YB, Zhu JK. Repair of extended peripheral nerve lesions in rhesus monkeys using acellular allogenic nerve grafts implanted with autologous mesenchymal stem cells. Exp Neurol. 2007;204(2):658–66.PubMed Hu J, Zhu QT, Liu XL, Xu YB, Zhu JK. Repair of extended peripheral nerve lesions in rhesus monkeys using acellular allogenic nerve grafts implanted with autologous mesenchymal stem cells. Exp Neurol. 2007;204(2):658–66.PubMed
75.
Zurück zum Zitat Auba C, Hontanilla B, Arcocha J, Gorria O. Peripheral nerve regeneration through allografts compared with autografts in FK506-treated monkeys. J Neurosurg. 2006;105(4):602–9.PubMed Auba C, Hontanilla B, Arcocha J, Gorria O. Peripheral nerve regeneration through allografts compared with autografts in FK506-treated monkeys. J Neurosurg. 2006;105(4):602–9.PubMed
76.
Zurück zum Zitat VandeBerg JL, Williams-Blangero S. Advantages and limitations of nonhuman primates as animal models in genetic research on complex diseases. J Med Primatol. 1997;26(3):113–9.PubMed VandeBerg JL, Williams-Blangero S. Advantages and limitations of nonhuman primates as animal models in genetic research on complex diseases. J Med Primatol. 1997;26(3):113–9.PubMed
77.
Zurück zum Zitat Phillips KA, Bales KL, Capitanio JP, Conley A, Czoty PW, ’t Hart BA, et al. Why primate models matter. Am J Primatol. 2014;76(9):801–27.PubMedPubMedCentral Phillips KA, Bales KL, Capitanio JP, Conley A, Czoty PW, ’t Hart BA, et al. Why primate models matter. Am J Primatol. 2014;76(9):801–27.PubMedPubMedCentral
78.
Zurück zum Zitat Gagneux P, Moore JJ, Varki A. The ethics of research on great apes. Nature. 2005;437(7055):27–9.PubMed Gagneux P, Moore JJ, Varki A. The ethics of research on great apes. Nature. 2005;437(7055):27–9.PubMed
79.
Zurück zum Zitat Roush W. Chimp retirement plan proposed. Science. 1997;277(5325):471.PubMed Roush W. Chimp retirement plan proposed. Science. 1997;277(5325):471.PubMed
80.
Zurück zum Zitat Rowan AN. The uncertain future of research chimpanzees. Science. 2007;315(5818):1493–4.PubMed Rowan AN. The uncertain future of research chimpanzees. Science. 2007;315(5818):1493–4.PubMed
81.
Zurück zum Zitat Mackinnon SE, Dellon AL. A study of nerve regeneration across synthetic (Maxon) and biologic (collagen) nerve conduits for nerve gaps up to 5 cm in the primate. J Reconstr Microsurg. 1990;6(2):117–21.PubMed Mackinnon SE, Dellon AL. A study of nerve regeneration across synthetic (Maxon) and biologic (collagen) nerve conduits for nerve gaps up to 5 cm in the primate. J Reconstr Microsurg. 1990;6(2):117–21.PubMed
82.
Zurück zum Zitat Kornfeld T, Vogt P, Bucan V, Peck C, Reimers K, Radtke C. Characterization and Schwann cell seeding of up to 15.0 cm long spider silk nerve conduits for reconstruction of peripheral nerve defects. J Funct Biomater. 2016;7(4):30.PubMedCentral Kornfeld T, Vogt P, Bucan V, Peck C, Reimers K, Radtke C. Characterization and Schwann cell seeding of up to 15.0 cm long spider silk nerve conduits for reconstruction of peripheral nerve defects. J Funct Biomater. 2016;7(4):30.PubMedCentral
83.
Zurück zum Zitat Santin M, Motta A, Freddi G, Cannas M. In vitro evaluation of the inflammatory potential of the silk fibroin. J Biomed Mater Res. 1999;46(3):382–9.PubMed Santin M, Motta A, Freddi G, Cannas M. In vitro evaluation of the inflammatory potential of the silk fibroin. J Biomed Mater Res. 1999;46(3):382–9.PubMed
84.
Zurück zum Zitat Panilaitis B, Altman GH, Chen J, Jin HJ, Karageorgiou V, Kaplan DL. Macrophage responses to silk. Biomaterials. 2003;24(18):3079–85.PubMed Panilaitis B, Altman GH, Chen J, Jin HJ, Karageorgiou V, Kaplan DL. Macrophage responses to silk. Biomaterials. 2003;24(18):3079–85.PubMed
85.
Zurück zum Zitat Allmeling C, Jokuszies A, Reimers K, Kall S, Vogt PM. Use of spider silk fibres as an innovative material in a biocompatible artificial nerve conduit. J Cell Mol Med. 2006;10(3):770–7.PubMed Allmeling C, Jokuszies A, Reimers K, Kall S, Vogt PM. Use of spider silk fibres as an innovative material in a biocompatible artificial nerve conduit. J Cell Mol Med. 2006;10(3):770–7.PubMed
86.
Zurück zum Zitat Allmeling C, Jokuszies A, Reimers K, Kall S, Choi CY, Brandes G, et al. Spider silk fibres in artificial nerve constructs promote peripheral nerve regeneration. Cell Prolif. 2008;41(3):408–20.PubMedPubMedCentral Allmeling C, Jokuszies A, Reimers K, Kall S, Choi CY, Brandes G, et al. Spider silk fibres in artificial nerve constructs promote peripheral nerve regeneration. Cell Prolif. 2008;41(3):408–20.PubMedPubMedCentral
87.
Zurück zum Zitat Altman GH, Diaz F, Jakuba C, Calabro T, Horan RL, Chen J, et al. Silk-based biomaterials. Biomaterials. 2003;24(3):401–16.PubMed Altman GH, Diaz F, Jakuba C, Calabro T, Horan RL, Chen J, et al. Silk-based biomaterials. Biomaterials. 2003;24(3):401–16.PubMed
88.
Zurück zum Zitat Fuente R, Mendioroz A, Salazar A. Revising the exceptionally high thermal diffusivity of spider silk. Mater Lett. 2014;114:1–3. Fuente R, Mendioroz A, Salazar A. Revising the exceptionally high thermal diffusivity of spider silk. Mater Lett. 2014;114:1–3.
89.
Zurück zum Zitat Schafer-Nolte F, Hennecke K, Reimers K, Schnabel R, Allmeling C, Vogt PM, et al. Biomechanics and biocompatibility of woven spider silk meshes during remodeling in a rodent fascia replacement model. Ann Surg. 2014;259(4):781–92.PubMed Schafer-Nolte F, Hennecke K, Reimers K, Schnabel R, Allmeling C, Vogt PM, et al. Biomechanics and biocompatibility of woven spider silk meshes during remodeling in a rodent fascia replacement model. Ann Surg. 2014;259(4):781–92.PubMed
91.
Zurück zum Zitat Vollrath F. Strength and structure of spiders’ silks. J Biotechnol. 2000;74(2):67–83.PubMed Vollrath F. Strength and structure of spiders’ silks. J Biotechnol. 2000;74(2):67–83.PubMed
92.
Zurück zum Zitat Vollrath F, Barth P, Basedow A, Engstrom W, List H. Local tolerance to spider silks and protein polymers in vivo. In Vivo. 2002;16(4):229–34.PubMed Vollrath F, Barth P, Basedow A, Engstrom W, List H. Local tolerance to spider silks and protein polymers in vivo. In Vivo. 2002;16(4):229–34.PubMed
93.
Zurück zum Zitat Kuhbier JW, Reimers K, Kasper C, Allmeling C, Hillmer A, Menger B, et al. First investigation of spider silk as a braided microsurgical suture. J Biomed Mater Res Part B Appl Biomater. 2011;97(2):381–7. Kuhbier JW, Reimers K, Kasper C, Allmeling C, Hillmer A, Menger B, et al. First investigation of spider silk as a braided microsurgical suture. J Biomed Mater Res Part B Appl Biomater. 2011;97(2):381–7.
95.
Zurück zum Zitat Satija NK, Singh VK, Verma YK, Gupta P, Sharma S, Afrin F, et al. Mesenchymal stem cell-based therapy: a new paradigm in regenerative medicine. J Cell Mol Med. 2009;13(11–12):4385–402.PubMedPubMedCentral Satija NK, Singh VK, Verma YK, Gupta P, Sharma S, Afrin F, et al. Mesenchymal stem cell-based therapy: a new paradigm in regenerative medicine. J Cell Mol Med. 2009;13(11–12):4385–402.PubMedPubMedCentral
97.
Zurück zum Zitat Barbour JR, Yee A, Moore AM, Trulock EP, Buchowski JM, Mackinnon SE. Cadaveric nerve allotransplantation in the treatment of persistent thoracic neuralgia. Ann Thorac Surg. 2015;99(4):1414–7.PubMed Barbour JR, Yee A, Moore AM, Trulock EP, Buchowski JM, Mackinnon SE. Cadaveric nerve allotransplantation in the treatment of persistent thoracic neuralgia. Ann Thorac Surg. 2015;99(4):1414–7.PubMed
98.
Zurück zum Zitat Cohen DJ, Loertscher R, Rubin MF, Tilney NL, Carpenter CB, Strom TB. Cyclosporine: a new immunosuppressive agent for organ transplantation. Ann Intern Med. 1984;101(5):667–82.PubMed Cohen DJ, Loertscher R, Rubin MF, Tilney NL, Carpenter CB, Strom TB. Cyclosporine: a new immunosuppressive agent for organ transplantation. Ann Intern Med. 1984;101(5):667–82.PubMed
99.
Zurück zum Zitat Matsuyama T, Midha R, Mackinnon SE, Munro CA, Wong PY, Ang LC. Long nerve allografts in sheep with Cyclosporin A immunosuppression. J Reconstr Microsurg. 2000;16(3):219–25.PubMed Matsuyama T, Midha R, Mackinnon SE, Munro CA, Wong PY, Ang LC. Long nerve allografts in sheep with Cyclosporin A immunosuppression. J Reconstr Microsurg. 2000;16(3):219–25.PubMed
101.
Zurück zum Zitat Brenner MJ, Lowe JB 3rd, Fox IK, Mackinnon SE, Hunter DA, Darcy MD, et al. Effects of Schwann cells and donor antigen on long-nerve allograft regeneration. Microsurgery. 2005;25(1):61–70.PubMed Brenner MJ, Lowe JB 3rd, Fox IK, Mackinnon SE, Hunter DA, Darcy MD, et al. Effects of Schwann cells and donor antigen on long-nerve allograft regeneration. Microsurgery. 2005;25(1):61–70.PubMed
102.
Zurück zum Zitat Jensen JN, Brenner MJ, Tung TH, Hunter DA, Mackinnon SE. Effect of FK506 on peripheral nerve regeneration through long grafts in inbred swine. Ann Plast Surg. 2005;54(4):420–7.PubMed Jensen JN, Brenner MJ, Tung TH, Hunter DA, Mackinnon SE. Effect of FK506 on peripheral nerve regeneration through long grafts in inbred swine. Ann Plast Surg. 2005;54(4):420–7.PubMed
103.
Zurück zum Zitat Ide C, Tohyama K, Tajima K, Endoh K, Sano K, Tamura M, et al. Long acellular nerve transplants for allogeneic grafting and the effects of basic fibroblast growth factor on the growth of regenerating axons in dogs: a preliminary report. Exp Neurol. 1998;154(1):99–112.PubMed Ide C, Tohyama K, Tajima K, Endoh K, Sano K, Tamura M, et al. Long acellular nerve transplants for allogeneic grafting and the effects of basic fibroblast growth factor on the growth of regenerating axons in dogs: a preliminary report. Exp Neurol. 1998;154(1):99–112.PubMed
Metadaten
Titel
Nerve grafting for peripheral nerve injuries with extended defect sizes
verfasst von
Tim Kornfeld
Peter M. Vogt
Christine Radtke, MD, PhD
Publikationsdatum
13.12.2018
Verlag
Springer Vienna
Erschienen in
Wiener Medizinische Wochenschrift / Ausgabe 9-10/2019
Print ISSN: 0043-5341
Elektronische ISSN: 1563-258X
DOI
https://doi.org/10.1007/s10354-018-0675-6

Weitere Artikel der Ausgabe 9-10/2019

Wiener Medizinische Wochenschrift 9-10/2019 Zur Ausgabe