Skip to main content
Erschienen in: EJNMMI Research 1/2020

Open Access 01.12.2020 | Original research

Fluorescence labeling of a NaV1.7-targeted peptide for near-infrared nerve visualization

verfasst von: Junior Gonzales, Giacomo Pirovano, Chun Yuen Chow, Paula Demetrio de Souza Franca, Lukas M. Carter, Julie K. Klint, Navjot Guru, Jason S. Lewis, Glenn F. King, Thomas Reiner

Erschienen in: EJNMMI Research | Ausgabe 1/2020

Abstract

Background

Accidental peripheral nerve injury during surgical intervention results in a broad spectrum of potentially debilitating side effects. Tissue distortion and poor visibility can significantly increase the risk of nerve injury with long-lasting consequences for the patient. We developed and characterized Hs1a-FL, a fluorescent near-infrared molecule for nerve visualization in the operating theater with the aim of helping physicians to visualize nerves during surgery. Hs1a was derived from the venom of the Chinese bird spider, Haplopelma schmidti, and conjugated to Cy7.5 dye. Hs1a-FL was injected intravenously in mice, and harvested nerves were imaged microscopically and with epifluorescence.

Results

Hs1a-FL showed specific and stable binding to the sodium channel NaV1.7, present on the surface of human and mouse nerves. Hs1a-FL allowed epifluorescence visualization of sciatic mouse nerves with favorable nerve-to-muscle contrast.

Conclusions

Fluorescent NaV1.7-targeted tracers have the potential to be adopted clinically for the intraoperative visualization of peripheral nerves during surgery, providing guidance for the surgeon and potentially improving the standard of care.
Hinweise
Junior Gonzales and Giacomo Pirovano contributed equally to this work.
Junior Gonzales and Giacomo Pirovano are co-first authors

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s13550-020-00630-4.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Background

Unintentional resection or injury of nerves during medical interventions is a significant concern during surgery [1, 2]. During surgery, there is always a degree of risk for nerves to be cut, crushed, tied off, penetrated and twisted by screws, or even injured during the removal of devices. In addition, they can be stretched by retractors, cut, or thermally damaged by electric knife, hardening bone cement, or during coagulation [3, 4]. These unintentional, iatrogenic complications typically occur because nerves are not clearly visible to the surgeon or could be mistaken for a vessel or tendon. Certain surgical procedures are considered high risk for resulting in nerve injury. These include, but are not limited to, osteosynthesis and osteotomy, arthrodesis, lymph node biopsy in the neck, parotidectomies, thyroid surgeries, carpal tunnel syndrome surgery, varicose vein surgery, excision of Baker cysts, and inguinal herniorrhaphies.
For example, in head and neck surgery, iatrogenic nerve injuries can result in facial paralysis, hoarseness or weakening of voice, respiratory distress, and other neurological complications with harsh lifestyle implications for the patient [58]. As a result, one out of four patients with neuropathic pain identifies surgical morbidity as the originating cause [9]. Oncologic surgery, in particular, poses a risk of nerve injury as anatomy is often distorted by the disease [1017]. There are existing tools for preoperative and perioperative nerve enhancement [1820], and they mostly rely on magnetic resonance imaging (MRI) and nerve ultrasound [21, 22]. However, fluorescent nerve imaging agents [23, 24], as well as multimodal optical imaging techniques, are raising interest for intraoperative applications [2535]. However, the availability of an intraoperative imaging agent that could clearly identify small nerve branches or even larger trunks below the tissue surface, especially in cases of reoperation when normal anatomy is disrupted, could significantly improve surgical precision. In an attempt to respond to this need, we developed Hs1a-FL, a near-infrared imaging agent that could be used in the described surgical settings.
Recombinant peptide Hs1a was derived from the venom of the Chinese bird spider, Haplopelma schmidti. Hs1a proved to be a potent and subtype-selective inhibitor of sodium channel NaV1.7, a key signal-transmitter located on nerve surfaces [36]. Our fluorescently labeled version of Hs1a targets NaV1.7 receptors and has the potential to be used as a vector for delivering an optical sensor to peripheral nerves in vivo. We show that the labeling of Hs1a with Cy7.5 N-hydroxy succinimide (NHS) ester could be used as a practical tool for nerve visualization during surgery in a preclinical mouse model (Fig. 1a). We anticipate that this technology will have the potential to directly impact the surgical standard of care by lending contrast to nerves, thereby decreasing iatrogenic injury and therefore surgical morbidity.

Methods

General

Unless otherwise stated, all solvents and reagents were obtained from Sigma-Aldrich or Fisher Scientific and were used without further purification. Cyanine7.5 (Cy7.5) was purchased from Lumiprobe (MD, USA). Anti-NaV1.7 antibody [N68/6] was purchased from Abcam (ab85015). Water (> 18.2 MΩ cm at 25 °C) was obtained from an Alpha-Q Ultrapure water system (Millipore). Acetonitrile (AcN) was of high-performance liquid chromatography (HPLC) grade and was purchased from Fisher Scientific. Phosphate-buffered saline (PBS) without Ca2+ or Mg2+ was obtained from the Media Preparation Facility at Memorial Sloan Kettering Cancer Center (MSKCC) and used for all in vivo injections. Reverse-phase (RP) HPLC purifications were performed on a Shimadzu HPLC system equipped with a DGU-20A degasser, SPD-M20A UV detector, LC-20AB pump system, and a CBM-20A communication bus module using RP-HPLC columns (Atlantis T3 C18, 5 μm, 4.6 × 250 mm, P/N: 186003748). Epifluorescence imaging was performed on an IVIS Spectrum imaging system (PerkinElmer). Confocal microscopy images were captured using a Leica SP8 inverted-stand confocal microscope equipped with a tunable white light laser that ranges from 470 to 670 nm. The microscope is also equipped with a 405-nm diode, argon laser (with 476 nm, 488 nm, 496 nm, and 514 nm laser line), and a 725-nm laser for near infra-red NIR imaging coupled with avalanche photo-diode detectors (APDs) which were used for detection of Hs1a-FL.

Synthesis of Hs1a

Recombinant Hs1a was produced via expression in the periplasm of E. coli using a protocol optimized for production of disulfide-rich peptides [37]. The recombinant peptide containing a non-native N-terminal glycine residue was purified by nickel affinity chromatography after liberation from the His6-MBP fusion tag via cleavage with tobacco etch virus protease. LC-ESI-MS (ES+), m/z calculated for [C164H251N49O47S6] 3850.74, [C164H251N49O47S6 + 3H]3+ 1284.58, found [M + 3H]3+ 1285.00, [C164H251N49O47S6 + 4H]4+ 963.69, found [M + 4H]4+ 964.20, [C164H251N49O47S6 + 5H]5+ 771.15, found [M + 5H]5+ 772.60, [C164H251N49O47S6 + 6H]6+ 642.79, found 643.25.

Synthesis of Hs1a-FL

Hs1a was discovered in a high-throughput fluorescent-based assay to screen spider venoms against hNaV1.7, as previously described [38]. Recombinant Hs1a peptide (0.26 mM, 200 μg in 200 μL of AcN) and Na2CO3 (1 M, 40 μL) were transferred into a 3-mL amber vial with a magnetic bar stirrer. Cy7.5-NHS (4 μL of a 24 mM solution) was dissolved in AcN and added dropwise to the reaction mixture. The final volume of the reaction mixture was 350 μL. The reaction mixture was stirred for at least 10 min before dilution with 100 μL of water. This reaction produced mono- and di-adducts of Cy7.5, which were purified and separated using RP-HPLC. Fractions containing the mono-adduct of Hs1a-FL were concentrated; then, the solvent was removed in vacuo to afford a dark greenish powder (20 μg, 14% yield from Hs1a peptide). This purified compound was then formulated in 100% Ca2+/Mg2+-free PBS or 10% dimethyl sulfoxide (DMSO) and PBS. LC-ESI-MS (ES+), m/z calculated for [C209H298N51O48S6] 4482.12, [C209H298N51O48S6 + 3H]3+ 1495.04, found [M + 3H]3+ 1495.45, [C209H298N51O48S6 + 4H]4+ 1121.53, found [M + 4H]4+ 1121.75, [C209H298N51O48S6 + 5H]5+ 897.42, found [M + 5H]5+ 897.75, [C209H298N51O48S6 + 6H]6+ 748.02, found [M + 6H]6+ 748.25

Cell lines

HEK293 cells stably expressing the human NaV channel β1 subunit (hNaVβ1) in combination with the α subunit hNaV1.1, hNaV1.2, hNaV1.3, hNaV1.4, hNaV1.5, hNaV1.6, or hNaV1.7 (Scottish Biomedical, Glasgow, UK) were cultured in DMEM/F-12 media (1:1), supplemented with 10% fetal bovine serum, 400 mg/mL geneticin, and 100 mM non-essential amino acids (all reagents from Invitrogen) at 37 °C and in 5% CO2.

Electrophysiology

Whole-cell patch-clamp experiments were performed at room temperature using a QPatch 16x automated electrophysiology platform (Sophion Bioscience, Denmark) using 16-channel planar patch-chip plates (QPlates) with a patch-hole diameter of 1 μm and resistance of 2 MΩ. Whole-cell currents were filtered at 5 kHz (8-pole Bessel) and digitized at 25 kHz. A P4 online leak-subtraction protocol was used with non-leak-subtracted currents acquired in parallel. The extracellular solution was 2 mM CaCl2, 1 mM MgCl2, 10 mM HEPES, 4 mM KCl, and 145 mM NaCl at pH 7.4, and the intracellular solution was 140 mM CsF, 1 mM/5 mM EGTA/CsOH, 10 mM HEPES, and 10 mM NaCl at pH 7.3. Hs1a-FL were dissolved in extracellular solution with 0.1% bovine serum albumin (BSA). Concentration-response data were obtained using five concentrations of peptide (2 nM to 10 μM). HEK293-hNaV cells were clamped at a holding potential of − 60 mV for NaV1.1, − 65 mV for NaV1.2, − 60 mV for NaV1.3, − 75 mV for NaV1.4, − 105 mV for NaV1.5, − 60 mV for NaV1.6, and − 75 mV for NaV1.7. For each concentration, 10 μL of peptide was added for 6 s before applying the following voltage protocol: − 80 mV for 10 ms, − 120 mV for 200 ms, 0 mV for 20 ms, then return to − 80 mV potential. This was repeated once every 60 s during liquid applications. Cells were otherwise held at the holding potential when the above voltage protocol was not executed. Upon establishment of the whole cell recording configuration, a total of five applications of the extracellular solution (1× control buffer, 3× test compound/control, 1 μM tetrodotoxin (TTX; positive control)), all containing 0.1% BSA (except for the TTX solution) were made on each cell. The voltage protocol was executed 10 times after each application. Currents were sampled at 25 kHz and filtered at 5 kHz with an 8-pole Bessel filter. The series resistance compensation level was set at 80%. All experiments were performed at room temperature (~ 22 °C). IC50 values were determined from non-linear regression of concentration-response data using GraphPad Prism.

Animal studies

Female athymic nude mice (4–8 weeks old, athymic-nude (outbred) (stock#:088; Envigo, USA) were allowed to acclimatize at the MSKCC vivarium for 1 week with ad libitum food and water prior to the experimental procedure. For imaging experiments, animals were sacrificed 30 min post-tail vein injection of Hs1a-FL, Hs1a/Hs1a-FL, or PBS. All animal experiments were performed in accordance with institutional guidelines and approved by the MSKCC Institutional Animal Care and Use Committee, following the NIH guidelines for animal welfare.

Mouse cryosectioning and image-based reconstruction

Post-euthanasia, a representative mouse was fast frozen in hexanes with dry ice. Coronal cryosectioning and white-light imaging were performed by EMIT using a Xerra imager; following each sequential removal of 50-μm-thick slices, the tissue-embedded block was imaged at 30 μm in-plane resolution. A 3D image volume of the mouse was generated through multiplanar reformation using 3D Slicer software for anatomic visualization (Fig. 1a).

Immunohistochemistry

Immunohistochemical (IHC) staining experiments were used to detect the expression and abundance of sodium channel NaV1.7 in mouse sciatic nerve tissue. Anti-NaV1.7 antibody [N68/6] (Abcam ab85015) was found to specifically bind to mouse NaV1.7 (0.5 μg/mL). Paraffin-embedded formalin-fixed 5 μm sections were deparaffinized with EZPrep buffer. For IHC detection, a 3,3′-diaminobenzidine (DAB) detection kit (Ventana Medical Systems, Tucson, AZ) was used according to the manufacturer’s instructions. These experiments were performed at the MSKCC Molecular Cytology Core Facility using the Discovery XT processor (Ventana Medical System, Tucson, AZ). Adjacent sections were stained against IgG, to control for non-specific binding to NaV1.7. Sections were counterstained with hematoxylin and eosin (H&E) and coverslipped with Permount (Fisher Scientific, Pittsburgh, PA) for morphological evaluation of tissue characteristics.

Confocal microscopy

For the confocal microscopy experiments, 5 μm cryosections of sciatic nerve tissues embedded in optimal cutting temperature compound (OCT) were used to determine the distribution and localization of Hs1a-FL from mice previously injected with the fluorescent agent Hs1a-FL (4 nmol, 45 μM of Hs1a-FL in 100 μL of PBS), the blocking solution Hs1a/Hs1a-FL (Hs1a-FL, 45 μM, 4 nmol and Hs1a 120 μM, 12 nmol in 100 μL PBS), or 100 μL of PBS. These resected nerves were incubated with Hoechst 33342 (20 μM, 1 nmol in 50 μL of PBS) to counterstain nuclei, which were subsequently embedded in Mowiol mounting medium. Fresh tissues were counterstained with Hoechst 33342 (20 μM, 1 nmol in 50 μL of PBS) and samples placed directly on a microscope slide for detection of the fluorescence signal of the fluorescent peptide.

Epifluorescence imaging

One group of animals was intravenously injected with Hs1a-FL (4 nmol, 45 μM of Hs1a-FL in 100 μL of PBS, n = 3). A second group of animals was injected with Hs1a and Hs1a-FL (Hs1a-FL, 45 μM, 4 nmol and Hs1a, 120 μM, 12 nmol in 100 μL PBS, n = 3) or PBS (n = 3). Animals were sacrificed 30 min post-injection and epifluorescence images obtained. Epifluorescence images of the right sciatic nerve (RSN) and left sciatic nerve (LSN) were obtained in situ from all the mice in the study. Epifluorescence images of the biodistribution included RSN, LSN, muscle, heart, kidney, liver, and brain and were acquired with an IVIS Spectrum imaging system (PerkinElmer) using a predetermined filter set (excitation = 710/45 nm, emission = 800–820 nm). Autofluorescence was removed through spectral unmixing. Semiquantitative analysis of the Hs1a-FL signal was conducted by measuring the average radiant efficiency (in units of [p/s/cm2/sr]/[μW/cm2]) in regions of interest (ROIs) that were placed on all resected nerves and as well in all organs from the biodistribution under white light guidance.

Statistical analyses

Statistical analyses were performed using GraphPad Prism 8. Unless otherwise stated, data points represent mean values, and error bars represent standard deviations of biological replicates. All p values were calculated using an unpaired t test. Statistical significance was considered for p values < 0.05 and as follows: ns = not significant, *p < 0.05, **p < 0.01, ***p < 0.001.

Results

Selectivity of Hs1a across Ion Channel Subtypes

Hs1a peptide was isolated from venom of the Chinese tarantula Haplopelma schmidti. To assess the selectivity across the various Nav channel subtypes, Hs1a was tested on human NaV1.1–NaV1.7 channels stably expressed in HEK293 cells using automated patch-clamp techniques. Hs1a was shown to have inhibitory affinity for neuronal Nav channels, with IC50 values in the low nanomolar range (19.4, 82, 107, 19.2, 26.9 nM for NaV1.1, NaV1.2, NaV1.3, NaV1.6, and NaV1.7, respectively), but it did not inhibit NaV1.4 and NaV1.5, which are found primarily in the muscle and heart, respectively at concentrations up to 3 μM (Fig. 1b, Table 1 in supplementary information).

Design of the fluorescence peptide, Hs1a-FL

We used recombinant Hs1a to synthesize Hs1a-FL, a NIR-labeled version of Hs1a (Fig. 1c). We chose a NIR fluorophore for its emission wavelength with favorable tissue penetration potential for intraoperative applications. We modified Hs1a via nucleophilic substitution as previously described [30]. The synthesis was performed under basic conditions in a mixture of water and acetonitrile, with 14% yield. Retention time (rt) shifted from 12 min for the unmodified Hs1a to 16 min for Hs1a-FL (Fig. 2a and Fig. S1a). The major impurities were characterized as the partially reduced peptide, 3% (rt 16.2 min), which was also present in the starting material (rt 12 min, 80% and rt 12.2 min, 20% for Hs1a and reduced Hs1a, respectively). LC/MS spectra for both Hs1a and Hs1a-FL showed clean peak families confirming the peptides’ calculated masses of 3850.74 Da and 4482.12 Da for Hs1a and Hs1a-FL, respectively (Fig. 2b, c, Table 2 in supplementary material). In addition, florescence of 0.1 μM Hs1a peptide and 0.1 μM Hs1a-FL were collected to confirm dye conjugation (Fig. 2d).

Histology and Hs1a-FL imaging of mouse sciatic nerve

To assess the possibility of using Hs1a-FL to image sciatic nerves in vivo, mice were injected intravenously with Hs1a-FL alone (4 nmol, 45 μM of Hs1a-FL in 100 μL of PBS) or in combination with an excess of unmodified peptide (120 μM, 12 nmol in 100 μL PBS, block) and sacrificed 30 min after injection. Nerves were surgically harvested and flash-frozen in OCT blocks. Blocks were then sliced on a cryotome at a 10-μm thickness and imaged. Nerves were imaged to detect fluorescent signal and H&E stained to enable the visualization of Schwann cells within the nerve structure. Anti-NaV1.7 immunohistochemistry confirmed target availability (Fig. 3a). Confocal microscopy confirmed the presence of Hs1a-FL signal in injected mice. No signal was detected in mice injected with PBS or “blocking solution” containing Hs1a-FL and a 3-fold molar excess of unlabeled Hs1a (Fig. 3b). In addition, no staining was observed when using isotype control antibodies, confirming specificity (Fig. S1b).

Ex vivo Hs1a-FL biodistribution

Mice were injected intravenously with Hs1a-FL alone (4 nmol, 45 μM of Hs1a-FL in 100 μL of PBS) or in combination with an excess of unmodified peptide (120 μM, 12 nmol in 100 μL PBS, blocking solution) and sacrificed 30 min after injection. The RSN and LSN were resected and epifluorescence imaging performed using an IVIS Spectrum imaging system (excitation = 710/45 nm, emission = 800–820 nm). In mice receiving just the imaging agent, we observed accumulation of Hs1a-FL in the resected sciatic nerves, which were clearly visible (Fig. 4a and Fig. S1c), whereas uptake was significantly reduced in the sciatic nerves of mice that received the imaging agent in combination with excess unmodified peptide (radiant efficiency: 1.6 ± 0.3 × 105 and 0.09 ± 0.03 × 105 for Hs1a-FL and co-injection (blocking solution), respectively; unpaired t test, p value < 0.001, Fig. 4b). A trend towards higher fluorescence signals in the liver, kidney, brain, and spleen was also observed (radiant efficiency: 3.0 ± 2.0 × 107 and 0.002 ± 0.001 × 107, 1.4 ± 1.1 × 107 and 0.005 ± 0.004 × 107, 0.2 ± 0.1 × 107 and 0.001 ± 0.0005 × 107, and 0.8 ± 0.5 × 107 and 0.004 ± 0.0003 × 107 for organs injected with fluorescent agent and with PBS, respectively; Fig. S2a and Fig. S2b). Nerve-to-muscle values showed a favorable accumulation in Hs1a-FL treated mice, compared to block and PBS (ratio: 4.83 ± 8.73, 0.48 ± 0.61, 2.90 ± 2.76, respectively, Supplementary Table 3).

Discussion

In the present study, we developed and characterized Hs1a-FL, a near-infrared imaging agent to target human nerves. A limitation of the present study is that, in a small rodent model, the high liver accumulation makes it difficult to discern the signal coming from NaV1.7-expressing nerve and unspecific liver signal. However, this limitation could be overcome in humans thanks to more favorable anatomy and nerves with larger diameters. Due to the preclinical model chosen, the sciatic nerves represent the largest peripheral nervous system target for our molecule in vivo. Hs1a is a peptide derived from the venom of a Chinese tarantula, with a low nanomolar affinity for neuronal sodium channels. Here, we showed that conjugation of Hs1a and Cy7.5 dye yields a fluorescent tracer for nerve imaging. The synthesis of Hs1a-FL was straightforward and efficient. We evaluated Hs1a-FL for ex vivo imaging of resected sciatic nerves via microscopy as well as imaging of exposed nerves via epifluorescence. The chosen mouse model for the in vivo investigation was athymic nude mice for their versatility and ease of use. Furthermore, nude mice do not have a furred skin and therefore allow an easier surgical exposure of the nerves without the need of hair removal. The mouse model should not affect the efficacy of Hs1a-FL in imaging nerves. Hs1a-FL was shown to specifically bind to NaV1.7 channels in vivo post-intravenous injection, and its binding could be blocked by competition with an excess dose of the unlabeled Hs1a. A more specific nerve agent could improve imaging potential and significantly limit background noise and possible toxicity. It will be interesting to investigate in the future whether blood pools or coagulated blood could represent a limitation in the signal-to-background ratio in the operation room and to identify potential changes in the binding affinity against different NaV channels after different dyes conjugation. The near-infrared emission spectrum of Cy7.5 may enable the use of this agent for visualization of nerves that are beneath the tissue surface during surgical interventions. The nerve-to-muscle ratio was calculated and showed a blockable signal of ~ 4 for Hs1a-FL-injected mice. A broader investigation is needed to identify the most ideal nerve imaging agent for intraoperative applications. This study represents the scientific basis for the development of a set of nerve-targeting agents derived from naturally available peptides.

Conclusion

Hs1a-FL proved to be a useful tool for nerve visualization. This nerve-targeting agent should be explored further with a view to developing tools that can assist surgeons to identify peripheral nerves and avoid surgical morbidity due to nerve injury.

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s13550-020-00630-4.

Acknowledgements

The authors thank the Tow Foundation and Memorial Sloan Kettering Cancer Center’s Center for Molecular Imaging & Nanotechnology (CMINT), the Imaging and Radiation Sciences Program, MSK Experimental Therapeutics Center, and the MSK Molecularly Targeted Intraoperative Imaging Fund. The authors thank the Small Animal Imaging Core, the Radiochemistry and Molecular Imaging Probes Core and the Molecular Cytology Core at Memorial Sloan Kettering Cancer Center for support. The authors thank Mohammed Farhoud for help with slicing and reconstructing the mouse (Emit Imaging, Boston, MA). L.M.C acknowledges support from the Ruth L. Kirschstein fellowship (NIH F32 EB025050). The funding sources were not involved in study design, data collection and analysis, writing of the report, or the decision to submit this article for publication.

Ethics approval

All applicable international, national, and/or institutional guidelines for the care and use of animals were followed.
All of the authors have read and approved the manuscript and possible conflict of interests are disclosed.

Competing interests

T.R. is shareholder of Summit Biomedical Imaging, LLC, and paid consultant for Theragnostics, Inc. J.G., P.D.d.S.F., G.F.K., J.S.L., and T.R. filed a patent surrounding the use of fluorophores with Hs1a and Hsp1a.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits 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/​4.​0/​.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Grinsell D, Keating CP. Peripheral nerve reconstruction after injury: a review of clinical and experimental therapies. Biomed Res Int. 2014;2014:698256.CrossRef Grinsell D, Keating CP. Peripheral nerve reconstruction after injury: a review of clinical and experimental therapies. Biomed Res Int. 2014;2014:698256.CrossRef
2.
Zurück zum Zitat Campbell WW. Evaluation and management of peripheral nerve injury. Clin Neurophysiol. 2008;119:1951–65.CrossRef Campbell WW. Evaluation and management of peripheral nerve injury. Clin Neurophysiol. 2008;119:1951–65.CrossRef
3.
Zurück zum Zitat Antoniadis G, Kretschmer T, Pedro MT, König RW, Heinen CPG, Richter H-P. Iatrogenic nerve injuries: prevalence, diagnosis and treatment. Dtsch Arztebl Int. 2014;111:273–9.PubMedPubMedCentral Antoniadis G, Kretschmer T, Pedro MT, König RW, Heinen CPG, Richter H-P. Iatrogenic nerve injuries: prevalence, diagnosis and treatment. Dtsch Arztebl Int. 2014;111:273–9.PubMedPubMedCentral
4.
Zurück zum Zitat Kretschmer T, Heinen CW, Antoniadis G, Richter HP, König RW. Iatrogenic nerve injuries. Neurosurg Clin N Am. 2009;20:73–90.CrossRef Kretschmer T, Heinen CW, Antoniadis G, Richter HP, König RW. Iatrogenic nerve injuries. Neurosurg Clin N Am. 2009;20:73–90.CrossRef
5.
Zurück zum Zitat Gordin E, Lee TS, Ducic Y, Arnaoutakis D. Facial nerve trauma: evaluation and considerations in management. Craniomaxillofac Trauma Reconstr. 2015;8:1–13.CrossRef Gordin E, Lee TS, Ducic Y, Arnaoutakis D. Facial nerve trauma: evaluation and considerations in management. Craniomaxillofac Trauma Reconstr. 2015;8:1–13.CrossRef
6.
Zurück zum Zitat Varaldo E, Ansaldo GL, Mascherini M, Cafiero F, Minuto MN. Neurological complications in thyroid surgery: a surgical point of view on laryngeal nerves. Front Endocrinol (Lausanne). 2014;5:108.CrossRef Varaldo E, Ansaldo GL, Mascherini M, Cafiero F, Minuto MN. Neurological complications in thyroid surgery: a surgical point of view on laryngeal nerves. Front Endocrinol (Lausanne). 2014;5:108.CrossRef
7.
Zurück zum Zitat Echternach M, Maurer CA, Maurer C, Mencke T, Schilling M, Verse T, et al. Laryngeal complications after thyroidectomy: is it always the surgeon. Arch Surg. 2009;144:149–53.CrossRef Echternach M, Maurer CA, Maurer C, Mencke T, Schilling M, Verse T, et al. Laryngeal complications after thyroidectomy: is it always the surgeon. Arch Surg. 2009;144:149–53.CrossRef
8.
Zurück zum Zitat Sosa JA, Bowman HM, Tielsch JM, Powe NR, Gordon TA, Udelsman R. The importance of surgeon experience for clinical and economic outcomes from thyroidectomy. Ann Surg. 1998;228:320–30.CrossRef Sosa JA, Bowman HM, Tielsch JM, Powe NR, Gordon TA, Udelsman R. The importance of surgeon experience for clinical and economic outcomes from thyroidectomy. Ann Surg. 1998;228:320–30.CrossRef
9.
Zurück zum Zitat Crombie IK, Davies HT, Macrae WA. Cut and thrust: antecedent surgery and trauma among patients attending a chronic pain clinic. Pain. 1998;76:167–71.CrossRef Crombie IK, Davies HT, Macrae WA. Cut and thrust: antecedent surgery and trauma among patients attending a chronic pain clinic. Pain. 1998;76:167–71.CrossRef
10.
Zurück zum Zitat Tasmuth T, von Smitten K, Hietanen P, Kataja M, Kalso E. Pain and other symptoms after different treatment modalities of breast cancer. Ann Oncol. 1995;6:453–9.CrossRef Tasmuth T, von Smitten K, Hietanen P, Kataja M, Kalso E. Pain and other symptoms after different treatment modalities of breast cancer. Ann Oncol. 1995;6:453–9.CrossRef
11.
Zurück zum Zitat Schneider B, Schickinger-Fischer B, Zumtobel M, Mancusi G, Bigenzahn W, Klepetko W, et al. Concept for diagnosis and therapy of unilateral recurrent laryngeal nerve paralysis following thoracic surgery. Thorac Cardiovasc Surg. 2003;51:327–31.CrossRef Schneider B, Schickinger-Fischer B, Zumtobel M, Mancusi G, Bigenzahn W, Klepetko W, et al. Concept for diagnosis and therapy of unilateral recurrent laryngeal nerve paralysis following thoracic surgery. Thorac Cardiovasc Surg. 2003;51:327–31.CrossRef
12.
Zurück zum Zitat Sihag S, Wright CD. Prevention and management of complications following tracheal resection. Thorac Surg Clin. 2015;25:499–508.CrossRef Sihag S, Wright CD. Prevention and management of complications following tracheal resection. Thorac Surg Clin. 2015;25:499–508.CrossRef
13.
Zurück zum Zitat Yumoto E, Sanuki T, Kumai Y. Immediate recurrent laryngeal nerve reconstruction and vocal outcome. Laryngoscope. 2006;116:1657–61.CrossRef Yumoto E, Sanuki T, Kumai Y. Immediate recurrent laryngeal nerve reconstruction and vocal outcome. Laryngoscope. 2006;116:1657–61.CrossRef
14.
Zurück zum Zitat Li H, Hu Y, Huang J, Yang Y, Xing K, Luo Q. Attempt of peripheral nerve reconstruction during lung cancer surgery. Thorac Cancer. 2018;9:580–3.CrossRef Li H, Hu Y, Huang J, Yang Y, Xing K, Luo Q. Attempt of peripheral nerve reconstruction during lung cancer surgery. Thorac Cancer. 2018;9:580–3.CrossRef
15.
Zurück zum Zitat Barnoiu OS, Garcia Galisteo E, Baron Lopez F, Vozmediano Chicharro R, Soler Martinez J, Del Rosal Samaniego JM, et al. Prospective urodynamic model for prediction of urinary incontinence after robot-assisted radical prostatectomy. Urol Int. 2014;92:306–9.CrossRef Barnoiu OS, Garcia Galisteo E, Baron Lopez F, Vozmediano Chicharro R, Soler Martinez J, Del Rosal Samaniego JM, et al. Prospective urodynamic model for prediction of urinary incontinence after robot-assisted radical prostatectomy. Urol Int. 2014;92:306–9.CrossRef
16.
Zurück zum Zitat Walsh PC, Donker PJ. Impotence following radical prostatectomy: insight into etiology and prevention. J Urol. 2017;197:S165–70.CrossRef Walsh PC, Donker PJ. Impotence following radical prostatectomy: insight into etiology and prevention. J Urol. 2017;197:S165–70.CrossRef
17.
Zurück zum Zitat Walz J, Burnett AL, Costello AJ, Eastham JA, Graefen M, Guillonneau B, et al. A critical analysis of the current knowledge of surgical anatomy related to optimization of cancer control and preservation of continence and erection in candidates for radical prostatectomy. Eur Urol. 2010;57:179–92.CrossRef Walz J, Burnett AL, Costello AJ, Eastham JA, Graefen M, Guillonneau B, et al. A critical analysis of the current knowledge of surgical anatomy related to optimization of cancer control and preservation of continence and erection in candidates for radical prostatectomy. Eur Urol. 2010;57:179–92.CrossRef
18.
Zurück zum Zitat Cage TA, Yuh EL, Hou SW, Birk H, Simon NG, Noss R, et al. Visualization of nerve fibers and their relationship to peripheral nerve tumors by diffusion tensor imaging. Neurosurg Focus. 2015;39:E16.CrossRef Cage TA, Yuh EL, Hou SW, Birk H, Simon NG, Noss R, et al. Visualization of nerve fibers and their relationship to peripheral nerve tumors by diffusion tensor imaging. Neurosurg Focus. 2015;39:E16.CrossRef
19.
Zurück zum Zitat Skorpil M, Karlsson M, Nordell A. Peripheral nerve diffusion tensor imaging. Magn Reson Imaging. 2004;22:743–5.CrossRef Skorpil M, Karlsson M, Nordell A. Peripheral nerve diffusion tensor imaging. Magn Reson Imaging. 2004;22:743–5.CrossRef
20.
Zurück zum Zitat Strakowski JA. Ultrasound-guided peripheral nerve procedures. Phys Med Rehabil Clin N Am. 2016;27:687–715.CrossRef Strakowski JA. Ultrasound-guided peripheral nerve procedures. Phys Med Rehabil Clin N Am. 2016;27:687–715.CrossRef
21.
Zurück zum Zitat Pitarokoili K, Kronlage M, Bäumer P, Schwarz D, Gold R, Bendszus M, et al. High-resolution nerve ultrasound and magnetic resonance neurography as complementary neuroimaging tools for chronic inflammatory demyelinating polyneuropathy. Ther Adv Neurol Disord. 2018;11:1756286418759974.CrossRef Pitarokoili K, Kronlage M, Bäumer P, Schwarz D, Gold R, Bendszus M, et al. High-resolution nerve ultrasound and magnetic resonance neurography as complementary neuroimaging tools for chronic inflammatory demyelinating polyneuropathy. Ther Adv Neurol Disord. 2018;11:1756286418759974.CrossRef
22.
Zurück zum Zitat Rangavajla G, Mokarram N, Masoodzadehgan N, Pai SB, Bellamkonda RV. Noninvasive imaging of peripheral nerves. Cells Tissues Organs. 2014;200:69–77.CrossRef Rangavajla G, Mokarram N, Masoodzadehgan N, Pai SB, Bellamkonda RV. Noninvasive imaging of peripheral nerves. Cells Tissues Organs. 2014;200:69–77.CrossRef
23.
Zurück zum Zitat Cotero VE, Siclovan T, Zhang R, Carter RL, Bajaj A, LaPlante NE, et al. Intraoperative fluorescence imaging of peripheral and central nerves through a myelin-selective contrast agent. Mol Imaging Biol. 2012;14:708–17.CrossRef Cotero VE, Siclovan T, Zhang R, Carter RL, Bajaj A, LaPlante NE, et al. Intraoperative fluorescence imaging of peripheral and central nerves through a myelin-selective contrast agent. Mol Imaging Biol. 2012;14:708–17.CrossRef
24.
Zurück zum Zitat Gibbs-Strauss SL, Nasr KA, Fish KM, Khullar O, Ashitate Y, Siclovan TM, et al. Nerve-highlighting fluorescent contrast agents for image-guided surgery. Mol Imaging. 2011;10:91–101.CrossRef Gibbs-Strauss SL, Nasr KA, Fish KM, Khullar O, Ashitate Y, Siclovan TM, et al. Nerve-highlighting fluorescent contrast agents for image-guided surgery. Mol Imaging. 2011;10:91–101.CrossRef
25.
Zurück zum Zitat Hussain T, Nguyen LT, Whitney M, Hasselmann J, Nguyen QT. Improved facial nerve identification during parotidectomy with fluorescently labeled peptide. Laryngoscope. 2016;126:2711–7.CrossRef Hussain T, Nguyen LT, Whitney M, Hasselmann J, Nguyen QT. Improved facial nerve identification during parotidectomy with fluorescently labeled peptide. Laryngoscope. 2016;126:2711–7.CrossRef
26.
Zurück zum Zitat Carolus AE, Lenz M, Hofmann M, Welp H, Schmieder K, Brenke C. High-resolution in vivo imaging of peripheral nerves using optical coherence tomography: a feasibility study. J Neurosurg. 2019:1–7. Carolus AE, Lenz M, Hofmann M, Welp H, Schmieder K, Brenke C. High-resolution in vivo imaging of peripheral nerves using optical coherence tomography: a feasibility study. J Neurosurg. 2019:1–7.
27.
Zurück zum Zitat Walsh EM, Cole D, Tipirneni KE, Bland KI, Udayakumar N, Kasten BB, et al. Fluorescence imaging of nerves during surgery. Ann Surg. 2019;270:69–76.CrossRef Walsh EM, Cole D, Tipirneni KE, Bland KI, Udayakumar N, Kasten BB, et al. Fluorescence imaging of nerves during surgery. Ann Surg. 2019;270:69–76.CrossRef
28.
Zurück zum Zitat Hingorani DV, Whitney MA, Friedman B, Kwon JK, Crisp JL, Xiong Q, et al. Nerve-targeted probes for fluorescence-guided intraoperative imaging. Theranostics. 2018;8:4226–37.CrossRef Hingorani DV, Whitney MA, Friedman B, Kwon JK, Crisp JL, Xiong Q, et al. Nerve-targeted probes for fluorescence-guided intraoperative imaging. Theranostics. 2018;8:4226–37.CrossRef
29.
Zurück zum Zitat Cha J, Broch A, Mudge S, Kim K, Namgoong JM, Oh E, et al. Real-time, label-free, intraoperative visualization of peripheral nerves and micro-vasculatures using multimodal optical imaging techniques. Biomed Opt Express. 2018;9:1097–110.CrossRef Cha J, Broch A, Mudge S, Kim K, Namgoong JM, Oh E, et al. Real-time, label-free, intraoperative visualization of peripheral nerves and micro-vasculatures using multimodal optical imaging techniques. Biomed Opt Express. 2018;9:1097–110.CrossRef
30.
Zurück zum Zitat Gonzales J, Demetrio de Souza Franca P, Jiang Y, Pirovano G, Kossatz S, Guru N, et al. Fluorescence imaging of peripheral nerves by a Nav1.7-targeted inhibitor cystine knot peptide. Bioconjugate Chem. 2019. Gonzales J, Demetrio de Souza Franca P, Jiang Y, Pirovano G, Kossatz S, Guru N, et al. Fluorescence imaging of peripheral nerves by a Nav1.7-targeted inhibitor cystine knot peptide. Bioconjugate Chem. 2019.
31.
Zurück zum Zitat Whitney MA, Crisp JL, Nguyen LT, Friedman B, Gross LA, Steinbach P, et al. Fluorescent peptides highlight peripheral nerves during surgery in mice. Nat Biotechnol. 2011;29:352–6.CrossRef Whitney MA, Crisp JL, Nguyen LT, Friedman B, Gross LA, Steinbach P, et al. Fluorescent peptides highlight peripheral nerves during surgery in mice. Nat Biotechnol. 2011;29:352–6.CrossRef
32.
Zurück zum Zitat Langhout GC, Kuhlmann KFD, Wouters MWJM, van der Hage JA, van Coevorden F, Müller M, et al. Nerve detection during surgery: optical spectroscopy for peripheral nerve localization. Lasers Med Sci. 2018;33:619–25.CrossRef Langhout GC, Kuhlmann KFD, Wouters MWJM, van der Hage JA, van Coevorden F, Müller M, et al. Nerve detection during surgery: optical spectroscopy for peripheral nerve localization. Lasers Med Sci. 2018;33:619–25.CrossRef
33.
Zurück zum Zitat He K, Zhou J, Yang F, Chi C, Li H, Mao Y, et al. Near-infrared intraoperative imaging of thoracic sympathetic nerves: from preclinical study to clinical trial. Theranostics. 2018;8:304–13.CrossRef He K, Zhou J, Yang F, Chi C, Li H, Mao Y, et al. Near-infrared intraoperative imaging of thoracic sympathetic nerves: from preclinical study to clinical trial. Theranostics. 2018;8:304–13.CrossRef
34.
Zurück zum Zitat Langhout GC, Bydlon TM, van der Voort M, Müller M, Kortsmit J, Lucassen G, et al. Nerve detection using optical spectroscopy, an evaluation in four different models: in human and swine, in-vivo, and post mortem. Lasers Surg Med. 2018;50:253–61.CrossRef Langhout GC, Bydlon TM, van der Voort M, Müller M, Kortsmit J, Lucassen G, et al. Nerve detection using optical spectroscopy, an evaluation in four different models: in human and swine, in-vivo, and post mortem. Lasers Surg Med. 2018;50:253–61.CrossRef
35.
Zurück zum Zitat Shin JG, Hwang HS, Eom TJ, Lee BH. In vivo three-dimensional imaging of human corneal nerves using Fourier-domain optical coherence tomography. J Biomed Opt. 2017;22:10501.CrossRef Shin JG, Hwang HS, Eom TJ, Lee BH. In vivo three-dimensional imaging of human corneal nerves using Fourier-domain optical coherence tomography. J Biomed Opt. 2017;22:10501.CrossRef
36.
Zurück zum Zitat King GF, Vetter I. No gain, no pain: NaV1.7 as an analgesic target. ACS Chem Neurosci. 2014;5:749–51.CrossRef King GF, Vetter I. No gain, no pain: NaV1.7 as an analgesic target. ACS Chem Neurosci. 2014;5:749–51.CrossRef
37.
Zurück zum Zitat Klint JK, Senff S, Saez NJ, Seshadri R, Lau HY, Bende NS, et al. Production of recombinant disulfide-rich venom peptides for structural and functional analysis via expression in the periplasm of E. coli. PLoS One. 2013;8:e63865.CrossRef Klint JK, Senff S, Saez NJ, Seshadri R, Lau HY, Bende NS, et al. Production of recombinant disulfide-rich venom peptides for structural and functional analysis via expression in the periplasm of E. coli. PLoS One. 2013;8:e63865.CrossRef
38.
Zurück zum Zitat Klint JK, Smith JJ, Vetter I, Rupasinghe DB, Er SY, Senff S, et al. Seven novel modulators of the analgesic target NaV 1.7 uncovered using a high-throughput venom-based discovery approach. Br J Pharmacol. 2015;172:2445–58.CrossRef Klint JK, Smith JJ, Vetter I, Rupasinghe DB, Er SY, Senff S, et al. Seven novel modulators of the analgesic target NaV 1.7 uncovered using a high-throughput venom-based discovery approach. Br J Pharmacol. 2015;172:2445–58.CrossRef
Metadaten
Titel
Fluorescence labeling of a NaV1.7-targeted peptide for near-infrared nerve visualization
verfasst von
Junior Gonzales
Giacomo Pirovano
Chun Yuen Chow
Paula Demetrio de Souza Franca
Lukas M. Carter
Julie K. Klint
Navjot Guru
Jason S. Lewis
Glenn F. King
Thomas Reiner
Publikationsdatum
01.12.2020
Verlag
Springer Berlin Heidelberg
Erschienen in
EJNMMI Research / Ausgabe 1/2020
Elektronische ISSN: 2191-219X
DOI
https://doi.org/10.1186/s13550-020-00630-4

Weitere Artikel der Ausgabe 1/2020

EJNMMI Research 1/2020 Zur Ausgabe