Skip to main content
Erschienen in: Molecular Pain 1/2010

Open Access 01.12.2010 | Research

Administration of a tropomyosin receptor kinase inhibitor attenuates sarcoma-induced nerve sprouting, neuroma formation and bone cancer pain

verfasst von: Joseph R Ghilardi, Katie T Freeman, Juan M Jimenez-Andrade, William G Mantyh, Aaron P Bloom, Michael A Kuskowski, Patrick W Mantyh

Erschienen in: Molecular Pain | Ausgabe 1/2010

Abstract

Pain often accompanies cancer and most current therapies for treating cancer pain have significant unwanted side effects. Targeting nerve growth factor (NGF) or its cognate receptor tropomyosin receptor kinase A (TrkA) has become an attractive target for attenuating chronic pain.
In the present report, we use a mouse model of bone cancer pain and examine whether oral administration of a selective small molecule Trk inhibitor (ARRY-470, which blocks TrkA, TrkB and TrkC kinase activity at low nm concentrations) has a significant effect on cancer-induced pain behaviors, tumor-induced remodeling of sensory nerve fibers, tumor growth and tumor-induced bone remodeling. Early/sustained (initiated day 6 post cancer cell injection), but not late/acute (initiated day 18 post cancer cell injection) administration of ARRY-470 markedly attenuated bone cancer pain and significantly blocked the ectopic sprouting of sensory nerve fibers and the formation of neuroma-like structures in the tumor bearing bone, but did not have a significant effect on tumor growth or bone remodeling.
These data suggest that, like therapies that target the cancer itself, the earlier that the blockade of TrkA occurs, the more effective the control of cancer pain and the tumor-induced remodeling of sensory nerve fibers. Developing targeted therapies that relieve cancer pain without the side effects of current analgesics has the potential to significantly improve the quality of life and functional status of cancer patients.
Begleitmaterial
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1744-8069-6-87) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

JRG participated in the design of the study, analysis and interpretation of the data, and drafted the manuscript. KTF participated in the analysis and interpretation of the animal behavior data. JMJA participated in the interpretation and analysis of the immunohistochemical data and contributed to the design of the study. WGM participated in the interpretation and analysis of the immunohistochemical data. APB participated in the interpretation and analysis of the μCT data. MAK provided the statistical analysis. PWM conceived of the study, provided analysis and interpretation of the data and significantly contributed to drafting the manuscript. All authors read and approved the final manuscript.

Background

Cancer pain can have a significant impact on the quality of life and functional status of the individual [1, 2]. A major reason cancer pain remains a significant health problem is the limited repertoire and negative side effects of currently available analgesics. For example, non-steroidal anti-inflammatory drugs, which are effective in reducing a variety of musculoskeletal pains, have been shown to have significant gastrointestinal side effects [3, 4]. Opiates are also frequently used to treat moderate to severe cancer pain. While opiates are highly effective at controlling ongoing cancer pain, as a class opiates have a variety of unwanted side effects including increased somnolence, agitation, constipation, dizziness, cognitive impairment and respiratory depression [5, 6].
Recently, peripherally restricted targeting of nerve growth factor (NGF) or its cognate tropomyosin receptor kinase A (TrkA) has become an attractive target for attenuating chronic pain. Four major strategies are currently being pursued (Figure 1) and each of these strategies has its potential strengths and limitations [7, 8]. For example, while monoclonal antibodies (mAbs) are extraordinarily specific in their targeting, administration of mAbs carries the risk of immune reactions such as acute anaphylaxis, serum sickness and the generation of antibodies against the therapeutic agent. In contrast, small molecule inhibitors of kinase activity do not require intravenous or intramuscular injection, are generally less expensive to make than mAbs, allow greater flexibility in dosing, but are generally less selective than mAbs [8]. Whether the kinases' lack of extraordinary specificity found with mAbs will provide greater desired efficacy or greater unwanted side effects will probably need to be examined with each mAb or kinase(s) that is being targeted.
In the present paper we use a mouse model of bone cancer pain to demonstrate that early administration of a small molecule kinase Trk inhibitor, ARRY-470, significantly reduces cancer pain in the early, middle and late time points in disease progression. Interestingly, the cancer and its associated stromal cells induced a remarkable sprouting and neuroma formation by sensory nerve fibers that innervate the tumor-bearing bone and this sprouting and neuroma formation was markedly attenuated by Trk inhibition. In contrast, Trk inhibition had no significant effect on tumor growth or tumor-induced bone remodeling in this model.

Results

Early, but not late ARRY-470 administration significantly attenuates tumor-induced pain

To assess whether inhibition of the Trks attenuates bone cancer pain, pain behaviors were analyzed in sham + vehicle and in tumor-bearing mice treated with early/acute ARRY-470, early/sustained ARRY-470, and late/acute ARRY-470. These behavioral analyses confirmed previous observations [9] that at early time points (days 8-14 post tumor cell injection), pain-related behaviors gradually increase in severity with time (Figure 2A), and correlate with tumor growth in the intramedullary space of the femur, as well as progressive tumor-induced bone destruction. Interestingly, pain behaviors escalated rapidly upon the escape of sarcoma cells from the intramedullary space (days 14-20 post tumor injection) (Figure 2A), which resulted in tumor-induced sprouting of CGRP+ and NF200+ nerve fibers in the periosteum. Behavioral analysis revealed that when ARRY-470 was given from days 6-8 post tumor injection, pain behaviors were reduced by ~40% at day 8, whereas early/sustained administration of ARRY-470 from days 6-20 reduced pain behaviors by ~60% at day 20. In contrast, late administration of ARRY-470 (initiated at day 18-20) did not significantly reduce cancer pain behaviors by day 20 (Figure 2B).

Tumor growth induces profuse sprouting of sensory nerve fibers and the formation of neuroma-like structures

Tumor-induced changes were examined in the periosteum, as this bone compartment is richly innervated by sensory nerve fibers [10], appears to be pivotally involved in detecting injury to the skeleton [11], and is the only tissue in bone that can be immunohistochemically analyzed in both decalcified frozen sections and non-decalcified whole mount preparations [10]. In naïve and sham + vehicle-treated animals CGRP+ sensory nerve fibers have a net-like organization and are typically associated with blood vessels. In sham mice, there was no difference in the organization or density of CGRP+ (Figure 3A) periosteal nerve fibers compared to naïve mice (data not shown). Twenty days following tumor cell injection, we observed significant sprouting by CGRP+ and NF200+ sensory nerve fibers. These fibers appeared to be intermingled among GFP+ tumor cells, and had a disorganized appearance that is never observed in the periosteum of naïve or sham mice.
Whereas all mice with GFP+ tumor cells growing in the periosteum showed significant sprouting of CGRP+ and NF200+ nerve fibers, approximately one half of these mice had 1-2 neuroma-like structures in the periosteum (Figure 4B, Figure 5B). These neuroma-like structures appear as a disordered mass of blind ending axons that have an interlacing or whirling morphology [12, 13] and are never observed in sham or naïve animals.

Early blockade of Trk's attenuates tumor-induced nerve sprouting and formation of neuroma-like structures and does not significantly affect disease progression

Early/sustained treatment with ARRY-470 (given twice daily from 6 to 20 days post tumor injection) largely prevented the sprouting of CGRP+ (Figure 6C, Figure 7A) and NF200+ (Figure 6F, Figure 7B) nerve fibers. Interestingly, the attenuation of this sprouting was observed only with the early/sustained ARRY-470 administration, and not with the late/acute administration (Figure 7A, B). Early/sustained administration of ARRY-470 similarly resulted in a marked decrease in the formation of neuroma-like structures of CGRP+ and NF200+ nerve fibers. Importantly, early/sustained administration of ARRY-470 did not affect the organization or density of CGRP+ or NF200+ fibers in the contralateral, non-tumor bearing bones compared to sham mice (data not shown).
In addition, the effects of ARRY-470 therapy on tumor growth and bone destruction were examined at day 14 and 20 post tumor injection, respectively. Treatment of sarcoma-injected mice with ARRY-470 at days 6-20 post tumor injection resulted in no reduction in tumor growth (100 ± 0 of intramedullary space (Additional file 1 Figure S1B) and no significant change in bone resorption (2.6 ± 0.24; as compared to sarcoma + vehicle animals (2.8 ± 0.14) (Additional file 1 Figure S1E).

Discussion

Blockade of Trks and skeletal pain

In the present study we show that early/sustained administration of a Trk inhibitor significantly inhibited sprouting and neuroma formation by sensory nerve fibers and reduced bone cancer pain-related behaviors by 50-60%. As the Trk inhibitor has a 50:1 plasma to CSF ratio, the anti-hyperalgesic actions of the inhibitor would appear to occur primarily outside the blood brain barrier. Previous reports have demonstrated that following peripheral inflammation and tissue injury, a variety of inflammatory, immune and stromal cells upregulate the expression of NGF, brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3) whose cognate receptors are TrkA, TrkB and TrkC respectively. Many studies have shown that peripheral NGF can drive pain and have suggested that NGF and perhaps peripherally released BDNF and NT-3 may play a role in modulating pain [14, 15]. As the present results show that blockade of all three Trks reduces bone cancer pain, a key question is which neurotrophins and Trks are most likely the major contributors to the generation and maintenance of bone cancer pain.
Previous results have demonstrated that in the adult NGF can directly activate and sensitize sensory neurons involved in the conduction of pain originating from the skin [16, 17], viscera and skeleton. NGF is thought to excite and sensitize sensory neurons by binding to its cognate receptor TrkA which is expressed by a subpopulation of mostly unmyelinated and thinly myelinated sensory neurons [18]. NGF binding to TrkA has been shown to directly activate TrkA-expressing nociceptors in vivo and in vitro and that binding of NGF to TrkA directly lowers the threshold for depolarization in these neurons [19, 20]. Additionally, NGF has been shown to modulate and/or sensitize a variety of neurotransmitters, receptors, ion channels and structural molecules expressed by nociceptors [17]. It has also been shown that NGF lowers the threshold and enhances the response of nociceptors to mechanical stimuli [21], suggesting that NGF activation of TrkA may play a role in activating/sensitizing mechanotransducers expressed by sensory nerve fibers. The NGF produced by target tissues or tumor cells activates TrkA receptors expressed on the terminals of C-fibers [18, 22] presumably including those innervating the skeleton. Whether the Trk inhibitor used here is exerting its effect by interfering with the retrograde signal (the internalized NGF/TrkA complex) that exerts transcriptional control in the neuronal cell body or by local modulation at the nociceptor terminal is not clear. However, in a model of bone fracture pain, with Trk inhibition at 2 days post fracture, where nerve sprouting has not yet occurred, the full analgesic effect is achieved 6-8 hours following acute administration [23]. These data suggest that local modulation of nerve fibers must be involved, as transport of the NGF/TrkA from the nerve terminals in the femoral fracture site to the cell bodies of sensory neurons that innervate the femur (which are located in the L1-L3 ganglia) would be expected to take significantly longer than 8 hours [24].
While there is strong evidence that TrkB receptors expressed by post-synaptic spinal cord neurons play a significant role in pain transmission [17] there is significantly less agreement about the role of TrkB expressed by sensory neurons in driving pain. Previous reports have suggested that TrkB receptors are expressed by a subpopulation of the DRG, nodose and trigeminal neurons and their terminals in the spinal dorsal horn and trigeminal nucleus [2527] and that peripheral inflammation in some tissues results in an increase in BDNF levels [16]. Interestingly, in pancreatitis BDNF content was reported to be correlated with pain intensity [28] and exogenous application of BDNF has been shown to excite and sensitize some cutaneous nociceptive terminals [29] (apparently via TrkB). However, the effects of BDNF on TrkB expressing sensory neurons in driving any type of chronic pain remain poorly understood [17]. Similarly, there is relatively little evidence to suggest that peripheral TrkC receptors expressed by myelinated nerve fibers play a significant role in the generation and maintenance of pain in the adult. Thus, while local injection of NT-3 has been reported to induce mild pain at the injection site [30], other reports suggest that NT-3 does not sensitize nociceptive primary afferent fibers [20] and appears to be anti-nociceptive in some pain models such as complete Freund's adjuvant-induced skin inflammation [31].
The above results, together with the present data demonstrating that the analgesic efficacy of the Trk inhibitor in blocking bone cancer pain is similar to that of anti-NGF sequestering therapy, suggest that TrkA plays the prominent role in driving bone cancer pain [32]. One unique aspect of the sensory innervation of bone, which may partially explain why Trk inhibition is effective in relieving skeletal pain, is that the majority of C-fibers that innervate the bone are CGRP-expressing fibers, and nearly all CGRP+ fibers co-express TrkA [18, 22]. Thus, most C-fibers that innervate both human [33] and rodent [34] vertebral discs and bone [35] appear to be CGRP/TrkA expressing fibers and few unmyelinated non-peptidergic IB4/RET+ nerve fibers are present in these tissues [10, 35]. Thus, since bone appears to lack the redundancy of the C-fiber non-peptidergic IB4/RET+ nerve fibers that are present in skin [10], blocking TrkA activation may be particularly efficacious in relieving bone pain vs. skin pain.

Trks and their involvement in the development of nerve sprouting and neuroma formation

Although Trks clearly play an essential role in the growth and survival of sensory neurons in the developing animal [36, 37], much less is known about the role the Trks play in the maintenance and survival of adult sensory neurons. In the adult, neurotrophins appear to be expressed in most tissues at very low levels, whereas the levels of NGF (and in some tissues BDNF and NT-3) are dramatically up-regulated by inflammation and/or injury [14, 16]. However, it has been shown that chronic NGF deprivation (by exogenous administration of an anti-NGF polyclonal antibody or autoimmunization to NGF) results in a modest hypoalgesia, where animals are less sensitive to some thermal and algogenic stimuli [38]. Whether this hypoalgesic effect observed in rats with polyclonal antibodies or autoimmunization will also be observed in humans treated with Trk inhibitors is unclear as is how much endogenous NGF, BDNF or NT-3 is required to maintain normal sensory nerve function in the adult.
The present studies suggest that similar to the tumor, sensory nerve fibers undergo a highly pathological and active reorganization as tumor cells invade the bone. Thus, as the tumor and associated stromal cells invade the bone, there is significant sprouting by CGRP+ and NF200+ sensory nerve fibers and these sensory nerve fibers are intermingled among the tumor/stromal cells that have invaded and remodeled the bone. These newly sprouted CGRP+ and NF200+ nerve fibers have a very dense and highly disorganized morphology that is never observed in the normal bone. In addition to the sprouting of nerve fibers, in approximately 1 out of 2 tumor-bearing bones we observe the appearance of neuroma-like structures that looked very similar to neuromas that have been described in both animals and humans following traumatic nerve injury. These structures appear as a disordered mass of CGRP+ and NF200+ blind ending axons that generally run parallel to each other and have an interlacing or whirling morphology [39, 40]. It should be emphasized that we have never observed these neuroma-like structures in the sham vehicle-treated or naïve bones. However, these data would agree with previous findings suggesting that NGF is involved in neuroma formation and when provided with the appropriate trophic factor, sensory nerve fibers can grow at a remarkable pace, sprouting several millimeters a day [41].
Previous studies have shown that injury to peripheral nerves associated with trauma, amputation, compression, or surgery can lead to painful neuromas [13, 42, 43], which have a morphology similar to the neuroma-like structures observed in the tumor-bearing mouse bones. In humans, these non-malignant neuromas frequently cause chronic and severe pain [13, 43] and can produce spontaneous ectopic discharges [44, 45] in part by up-regulation of sodium channels [43, 46]. Problematically, painful neuromas can be largely refractory to current medical treatment [43]. It is not currently known whether there is up-regulation of sodium channels and spontaneous discharge by these neuroma-like structures in the tumor bearing mouse bone. However, movement may not be required for these ectopic discharges to occur suggesting that this mechanism is a possible explanation for spontaneous breakthrough pain.

Conclusions

The present study shows that early/sustained administration of a small molecule Trk inhibitor attenuates sensory nerve fiber sprouting, neuroma formation, and bone cancer pain-related behaviors. Previous studies have shown that inappropriate remodeling of sensory nerve fibers, whether through sprouting or neuroma-like formation, can give rise to hyperalgesia, allodynia, and spontaneous ectopic discharges that are perceived as highly painful in humans [13, 43, 47]. The present data suggest that early/sustained administration of therapies that block the NGF/TrkA axis may be more effective than late administration in reducing the ectopic sprouting as well as cancer pain.

Methods

Animals

Experiments were performed on a total of 120 adult male C3H/HeJ mice (Jackson Laboratories, Bar Harbor, ME), initially at 8 weeks of age, weighing 20-25 g. The mice were housed in accordance with the National Institutes of Health guidelines under specific pathogen free conditions in autoclaved cages maintained at 22°C with a 12-hour alternating light and dark cycle and were given autoclaved food and water ad libitum. All procedures were approved by the Institutional Animal Care and Use Committee at the Minneapolis VA Medical Center.

Surgical procedure for implantation of cancer cells

Osteolytic murine sarcoma cells were obtained (NCTC 2472, ATCC, Rockville, MD), stably transfected with green fluorescent protein, maintained and surgically implanted into the mouse femur as previously described [32] (Additional file 1 Figure S1).

Treatment with Trk inhibitor (ARRY-470) therapy

The Trk inhibitor (ARRY-470; Array BioPharma, Boulder, CO) is a potent inhibitor of the tropomyosin kinase family of neurotrophin receptors, demonstrating nanomolar cellular inhibition of TrkA (6.5 nM), TrkB (8.1 nM), and TrkC (10.6 nM) and a high level of selectivity over a panel of kinases run at the ATP Km at 1.0 uM and non-kinase receptors [48](Additional file 1 Table S1 and S2). At doses of 10-100 mg/kg ARRY-470 reaches high concentrations in plasma and peripheral tissues, while the brain concentrations remain negligible, suggesting very limited crossing of the blood brain barrier [49]. ARRY-470 at a dose of 30 mg/kg in a mouse xenograft model derived from HEK cells constitutively expressing active human TrkA showed >90% inhibition of phosphorylated TrkA at 1 hour and >70% inhibition over a 12 hour time course [49]. Additionally, results from previous studies have demonstrated that administration of 30 mg/kg ARRY-470 significantly reduced thermal hyperalgesia and mechanical allodynia in a rat CFA model of inflammation [49].
To evaluate the effect of early vs. late dosing of a Trk inhibitor on pain-related behaviors, neurochemical changes and disease progression, treatment with ARRY-470 (30 mg/kg, p.o., bid) was initiated either when cancer-induced pain behaviors became evident (day 6-20 post-sarcoma injection) or after significant disease progression (day 18-20 post-sarcoma injection).

Behavioral measures of cancer pain

Pain behavior analysis was performed as previously described [32] (Additional file 1 Methods) and used to evaluate of the analgesic efficacy of ARRY-470 (30 mg/kg, p.o.) in attenuating bone cancer-related pain behavior. Assessment of bone cancer pain-related behaviors, including spontaneous guarding and flinching of the hind limb, was performed on days 8, 10, 12, 14, 16, 18 and 20 following tumor cell or sham inoculation. Behavioral analyses were performed within 30-60 minutes of administration of the drug or vehicle (Labrafac;polyglycolyzed glyceride).

Immunohistochemistry

Mice were sacrificed by carbon dioxide asphyxiation, delivered using a compressed gas cylinder, at day 20 post sarcoma injection and perfused intracardially with 20 ml of 0.1 M phosphate buffered saline (PBS, pH = 7.4 at 4°C) followed by 30 ml of 4% formaldehyde/12.5% picric acid solution in 0.1 M PBS (pH = 6.9 at 4°C). Ipsilateral and contralateral femurs were harvested following perfusion and post-fixed for at least 12 hours in the perfusion fixative.
To qualitatively assess the tumor-induced changes in the density and morphology of sensory nerve fibers that innervate the periosteum, whole mount preparations were processed on 20 bones according to the our previously published procedures [32]. The size of the periosteal whole mount preparation and its attached thin muscle layer used for immunohistochemistry was approximately: width = 6 mm; length = 6 mm; thickness = 0.5 mm.
For quantification, frozen sections were used as the cross sections allow visualization of the bone's landmarks (such as the growth plate), which enable the observer to locate the same anatomical area when quantifying changes in nerve fibers in different animals. The process of post-fixing, decalcification and sectioning of the femurs was performed as previously described [32].
Periosteum whole mounts and frozen sections were processed according to previously published procedures [32] using a marker of primary afferent sensory neurons (polyclonal rabbit anti-rat CGRP; 1:10,000; Sigma Chemical Co., St. Louis, MO; Catalog number C8198) and a marker of myelinated primary afferent sensory nerve fibers (chicken anti-neurofilament 200Kd; NF200, 1:1000; Chemicon, Temecula, CA; Catalog number AB5539). GFP expression levels did not require amplification to quantify tumor progression.

Quantification of nerve fiber density, sprouting and neuroma formation

The density of CGRP+ and NF200+ nerve fibers in the periosteum following treatment with vehicle or ARRY-470 was determined by capturing images of these nerve fibers in periosteal frozen sections using an Olympus Fluoview FV1000 laser scanning confocal imaging system (Olympus America Inc, Melville, NY, software v. 5.0). Approximately 30 separate, 20 μm thick frozen sections were obtained from each femur. Three images were obtained for each marker (200× magnification) and each image was acquired within 2 mm distal from the proximal femoral growth plate, with images taken from different sections at least 100 μm apart. The average area of periosteum that was analyzed was 620 μm (length), 70 μm (width), 20 μm (depth). The Z-stacked images were analyzed with Image-Pro Plus v. 6.0 (Media Cybernetics) and nerve fibers were manually traced to determine the length of nerve fibers. Nerve sprouting was reported as density of nerve fibers per volume of periosteum [32].
To quantify the extent of formation of neuroma-like structures, frozen sections were examined with a fluorescent microscope and these structures were manually counted and totaled from the entire 20 um thick section. Three different sections, each at least 100 um apart, were evaluated per animal. A neuroma-like structure was defined as i.) a disordered mass of blind ending axons (CGRP+ or NF200+) that has an interlacing and/or whirling morphology, ii.) a structure with a size of more than 10 individual axons that is at least 20 μm thick and 70 μm long, and iii.) a structure which is never observed in the periosteum of normal bone [39, 40].

Quantification of tumor growth and tumor-induced bone destruction

Images of sections from tumor-bearing femurs were acquired and the total area of intramedullary space and the percent of intramedullary space occupied by tumor cells were calculated using Image Pro Plus v6.0 software (Media Cybernetics, Silver Spring, MD) as previously described [9]. Area of intramedullary space occupied by tumor cells is presented as a percentage of total intramedullary area. Radiograph images of the medial-lateral plane of both bones were used to evaluate tumor-induced bone destruction as previously described [10] (Additional file 1 Figure S1).

Statistics

A one-way ANOVA was used to compare behavioral results and immunohistochemical measures between the experimental groups. For multiple comparisons, the Fisher's PLSD (Protected Least Significant Difference) post hoc test was used. Significance level was set at P < 0.05. In all cases, the investigator responsible for behavioral testing, plotting, measuring, and counting was blind to the experimental situation of each animal.

Acknowledgements and Funding

This work was supported by the National Institutes of Health (NS23970), the Department of Veterans Affairs, Veterans Health Administration, Rehabilitation Research and Development Service grants (O4380-I and A6707-R), Calhoun Fund for Bone Pain and a research grant from Array Biopharma. We thank Marvin Landis and the University of Arizona Information Technology Service for their expertise in generating the 3 D images of sensory nerve fibers in bone.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

JRG participated in the design of the study, analysis and interpretation of the data, and drafted the manuscript. KTF participated in the analysis and interpretation of the animal behavior data. JMJA participated in the interpretation and analysis of the immunohistochemical data and contributed to the design of the study. WGM participated in the interpretation and analysis of the immunohistochemical data. APB participated in the interpretation and analysis of the μCT data. MAK provided the statistical analysis. PWM conceived of the study, provided analysis and interpretation of the data and significantly contributed to drafting the manuscript. All authors read and approved the final manuscript.
Anhänge
Literatur
1.
Zurück zum Zitat Mercadante S: Malignant bone pain: pathophysiology and treatment. Pain 1997, 69: 1–18. 10.1016/S0304-3959(96)03267-8PubMedCrossRef Mercadante S: Malignant bone pain: pathophysiology and treatment. Pain 1997, 69: 1–18. 10.1016/S0304-3959(96)03267-8PubMedCrossRef
2.
Zurück zum Zitat Coleman RE: Skeletal complications of malignancy. Cancer 1997, 80: 1588–94. 10.1002/(SICI)1097-0142(19971015)80:8+<1588::AID-CNCR9>3.0.CO;2-GPubMedCrossRef Coleman RE: Skeletal complications of malignancy. Cancer 1997, 80: 1588–94. 10.1002/(SICI)1097-0142(19971015)80:8+<1588::AID-CNCR9>3.0.CO;2-GPubMedCrossRef
3.
Zurück zum Zitat Xian CJ, Zhou XF: Treating skeletal pain: limitations of conventional anti-inflammatory drugs, and anti-neurotrophic factor as a possible alternative. Nat Clin Pract Rheumatol 2009, 5: 92–8. 10.1038/ncprheum0982PubMedCrossRef Xian CJ, Zhou XF: Treating skeletal pain: limitations of conventional anti-inflammatory drugs, and anti-neurotrophic factor as a possible alternative. Nat Clin Pract Rheumatol 2009, 5: 92–8. 10.1038/ncprheum0982PubMedCrossRef
4.
Zurück zum Zitat Balano KB: Anti-inflammatory drugs and myorelaxants. Pharmacology and clinical use in musculoskeletal disease. Prim Care 1996, 23: 329–34.PubMedCrossRef Balano KB: Anti-inflammatory drugs and myorelaxants. Pharmacology and clinical use in musculoskeletal disease. Prim Care 1996, 23: 329–34.PubMedCrossRef
5.
Zurück zum Zitat Feinberg SD: Prescribing analgesics. How to improve function and avoid toxicity when treating chronic pain. Geriatrics 2000, 55: 44. 49–50, 53 passimPubMed Feinberg SD: Prescribing analgesics. How to improve function and avoid toxicity when treating chronic pain. Geriatrics 2000, 55: 44. 49–50, 53 passimPubMed
6.
Zurück zum Zitat Ivanhoe CB, Hartman ET: Clinical caveats on medical assessment and treatment of pain after TBI. J Head Trauma Rehabil 2004, 19: 29–39. 10.1097/00001199-200401000-00004PubMedCrossRef Ivanhoe CB, Hartman ET: Clinical caveats on medical assessment and treatment of pain after TBI. J Head Trauma Rehabil 2004, 19: 29–39. 10.1097/00001199-200401000-00004PubMedCrossRef
7.
Zurück zum Zitat Hansel TT, Kropshofer H, Singer T, Mitchell JA, George AJ: The safety and side effects of monoclonal antibodies. Nat Rev Drug Discov 2010, 9: 325–38. 10.1038/nrd3003PubMedCrossRef Hansel TT, Kropshofer H, Singer T, Mitchell JA, George AJ: The safety and side effects of monoclonal antibodies. Nat Rev Drug Discov 2010, 9: 325–38. 10.1038/nrd3003PubMedCrossRef
8.
Zurück zum Zitat Opar A: Kinase inhibitors attract attention as oral rheumatoid arthritis drugs. Nat Rev Drug Discov 2010, 9: 257–8. 10.1038/nrd3155PubMedCrossRef Opar A: Kinase inhibitors attract attention as oral rheumatoid arthritis drugs. Nat Rev Drug Discov 2010, 9: 257–8. 10.1038/nrd3155PubMedCrossRef
9.
Zurück zum Zitat Halvorson KG, Sevcik MA, Ghilardi JR, Sullivan LJ, Koewler NJ, Bauss F, Mantyh PW: Intravenous Ibandronate Rapidly Reduces Pain, Neurochemical Indices of Central Sensitization, Tumor Burden, and Skeletal Destruction in a Mouse Model of Bone Cancer. J Pain Symptom Manage 2008,36(3):289–303. 10.1016/j.jpainsymman.2007.10.005PubMedCentralPubMedCrossRef Halvorson KG, Sevcik MA, Ghilardi JR, Sullivan LJ, Koewler NJ, Bauss F, Mantyh PW: Intravenous Ibandronate Rapidly Reduces Pain, Neurochemical Indices of Central Sensitization, Tumor Burden, and Skeletal Destruction in a Mouse Model of Bone Cancer. J Pain Symptom Manage 2008,36(3):289–303. 10.1016/j.jpainsymman.2007.10.005PubMedCentralPubMedCrossRef
10.
Zurück zum Zitat Jimenez-Andrade JM, Mantyh WG, Bloom AP, Xu H, Ferng AS, Dussor G, Vanderah TW, Mantyh PW: A phenotypically restricted set of primary afferent nerve fibers innervate the bone versus skin: therapeutic opportunity for treating skeletal pain. Bone 2010, 46: 306–13. 10.1016/j.bone.2009.09.013PubMedCentralPubMedCrossRef Jimenez-Andrade JM, Mantyh WG, Bloom AP, Xu H, Ferng AS, Dussor G, Vanderah TW, Mantyh PW: A phenotypically restricted set of primary afferent nerve fibers innervate the bone versus skin: therapeutic opportunity for treating skeletal pain. Bone 2010, 46: 306–13. 10.1016/j.bone.2009.09.013PubMedCentralPubMedCrossRef
11.
Zurück zum Zitat Greenfield GQ, MV Boswell, Cole BE: Orthopaedic Pain. Journal/Weiner's pain management: a practical guide for clinicians 2006, 465–76. Greenfield GQ, MV Boswell, Cole BE: Orthopaedic Pain. Journal/Weiner's pain management: a practical guide for clinicians 2006, 465–76.
12.
Zurück zum Zitat Small JR, Scadding JW, Landon DN: A fluorescence study of changes in noradrenergic sympathetic fibres in experimental peripheral nerve neuromas. J Neurol Sci 1990, 100: 98–107. 10.1016/0022-510X(90)90019-JPubMedCrossRef Small JR, Scadding JW, Landon DN: A fluorescence study of changes in noradrenergic sympathetic fibres in experimental peripheral nerve neuromas. J Neurol Sci 1990, 100: 98–107. 10.1016/0022-510X(90)90019-JPubMedCrossRef
13.
Zurück zum Zitat Lindqvist A, Rivero-Melian C, Turan I, Fried K: Neuropeptide- and tyrosine hydroxylase-immunoreactive nerve fibers in painful Morton's neuromas. Muscle Nerve 2000, 23: 1214–8. 10.1002/1097-4598(200008)23:8<1214::AID-MUS9>3.0.CO;2-APubMedCrossRef Lindqvist A, Rivero-Melian C, Turan I, Fried K: Neuropeptide- and tyrosine hydroxylase-immunoreactive nerve fibers in painful Morton's neuromas. Muscle Nerve 2000, 23: 1214–8. 10.1002/1097-4598(200008)23:8<1214::AID-MUS9>3.0.CO;2-APubMedCrossRef
14.
15.
Zurück zum Zitat McMahon SB, Bennett DLH, Bevan S, McMahon SB, Koltzenburg M: Inflammatory mediators and modulators of pain. Journal/Wall and Melzack's textbook of pain 2006, xviii. 1239 p McMahon SB, Bennett DLH, Bevan S, McMahon SB, Koltzenburg M: Inflammatory mediators and modulators of pain. Journal/Wall and Melzack's textbook of pain 2006, xviii. 1239 p
16.
Zurück zum Zitat Paterson S, Schmelz M, McGlone F, Turner G, Rukwied R: Facilitated neurotrophin release in sensitized human skin. Eur J Pain 2009, 13: 399–405. 10.1016/j.ejpain.2008.05.005PubMedCrossRef Paterson S, Schmelz M, McGlone F, Turner G, Rukwied R: Facilitated neurotrophin release in sensitized human skin. Eur J Pain 2009, 13: 399–405. 10.1016/j.ejpain.2008.05.005PubMedCrossRef
17.
Zurück zum Zitat Pezet S, McMahon SB: Neurotrophins: mediators and modulators of pain. Annu Rev Neurosci 2006, 29: 507–38. 10.1146/annurev.neuro.29.051605.112929PubMedCrossRef Pezet S, McMahon SB: Neurotrophins: mediators and modulators of pain. Annu Rev Neurosci 2006, 29: 507–38. 10.1146/annurev.neuro.29.051605.112929PubMedCrossRef
18.
Zurück zum Zitat Averill S, McMahon SB, Clary DO, Reichardt LF, Priestley JV: Immunocytochemical localization of trkA receptors in chemically identified subgroups of adult rat sensory neurons. Eur J Neurosci 1995, 7: 1484–1494. 10.1111/j.1460-9568.1995.tb01143.xPubMedCentralPubMedCrossRef Averill S, McMahon SB, Clary DO, Reichardt LF, Priestley JV: Immunocytochemical localization of trkA receptors in chemically identified subgroups of adult rat sensory neurons. Eur J Neurosci 1995, 7: 1484–1494. 10.1111/j.1460-9568.1995.tb01143.xPubMedCentralPubMedCrossRef
19.
Zurück zum Zitat Hefti FF, Rosenthal A, Walicke PA, Wyatt S, Vergara G, Shelton DL, Davies AM: Novel class of pain drugs based on antagonism of NGF. Trends Pharmacol Sci 2006, 27: 85–91. 10.1016/j.tips.2005.12.001PubMedCrossRef Hefti FF, Rosenthal A, Walicke PA, Wyatt S, Vergara G, Shelton DL, Davies AM: Novel class of pain drugs based on antagonism of NGF. Trends Pharmacol Sci 2006, 27: 85–91. 10.1016/j.tips.2005.12.001PubMedCrossRef
21.
Zurück zum Zitat Malik-Hall M, Dina OA, Levine JD: Primary afferent nociceptor mechanisms mediating NGF-induced mechanical hyperalgesia. Eur J Neurosci 2005, 21: 3387–94. 10.1111/j.1460-9568.2005.04173.xPubMedCrossRef Malik-Hall M, Dina OA, Levine JD: Primary afferent nociceptor mechanisms mediating NGF-induced mechanical hyperalgesia. Eur J Neurosci 2005, 21: 3387–94. 10.1111/j.1460-9568.2005.04173.xPubMedCrossRef
22.
Zurück zum Zitat Walsh GS, Krol KM, Kawaja MD: Absence of the p75 neurotrophin receptor alters the pattern of sympathosensory sprouting in the trigeminal ganglia of mice overexpressing nerve growth factor. J Neurosci 1999, 19: 258–73.PubMed Walsh GS, Krol KM, Kawaja MD: Absence of the p75 neurotrophin receptor alters the pattern of sympathosensory sprouting in the trigeminal ganglia of mice overexpressing nerve growth factor. J Neurosci 1999, 19: 258–73.PubMed
23.
Zurück zum Zitat Ghilardi JR, Freeman KT, Jimenez-Andrade JM, Mantyh WG, Bloom AP, Bouhana KS, Trollinger D, Winkler J, Lee P, Andrews SW, Kuskowski MA, Mantyh PW: Sustained blockade of neurotrophin receptors TrkA, TrkB and TrkC reduces non-malignant skeletal pain but not the maintenance of sensory and sympathetic nerve fibers. Bone 2010, in press. Ghilardi JR, Freeman KT, Jimenez-Andrade JM, Mantyh WG, Bloom AP, Bouhana KS, Trollinger D, Winkler J, Lee P, Andrews SW, Kuskowski MA, Mantyh PW: Sustained blockade of neurotrophin receptors TrkA, TrkB and TrkC reduces non-malignant skeletal pain but not the maintenance of sensory and sympathetic nerve fibers. Bone 2010, in press.
24.
Zurück zum Zitat Chao MV: Neurotrophins and their receptors: a convergence point for many signalling pathways. Nat Rev Neurosci 2003, 4: 299–309. 10.1038/nrn1078PubMedCrossRef Chao MV: Neurotrophins and their receptors: a convergence point for many signalling pathways. Nat Rev Neurosci 2003, 4: 299–309. 10.1038/nrn1078PubMedCrossRef
25.
Zurück zum Zitat Behnia A, Zhang L, Charles M, Gold MS: Changes in TrkB-like immunoreactivity in rat trigeminal ganglion after tooth injury. J Endod 2003, 29: 135–40. 10.1097/00004770-200302000-00012PubMedCrossRef Behnia A, Zhang L, Charles M, Gold MS: Changes in TrkB-like immunoreactivity in rat trigeminal ganglion after tooth injury. J Endod 2003, 29: 135–40. 10.1097/00004770-200302000-00012PubMedCrossRef
26.
Zurück zum Zitat Kashiba H, Uchida Y, Senba E: Distribution and colocalization of NGF and GDNF family ligand receptor mRNAs in dorsal root and nodose ganglion neurons of adult rats. Brain Res Mol Brain Res 2003, 110: 52–62. 10.1016/S0169-328X(02)00584-3PubMedCrossRef Kashiba H, Uchida Y, Senba E: Distribution and colocalization of NGF and GDNF family ligand receptor mRNAs in dorsal root and nodose ganglion neurons of adult rats. Brain Res Mol Brain Res 2003, 110: 52–62. 10.1016/S0169-328X(02)00584-3PubMedCrossRef
27.
Zurück zum Zitat Kobayashi K, Fukuoka T, Obata K, Yamanaka H, Dai Y, Tokunaga A, Noguchi K: Distinct expression of TRPM8, TRPA1, and TRPV1 mRNAs in rat primary afferent neurons with adelta/c-fibers and colocalization with trk receptors. J Comp Neurol 2005, 493: 596–606. 10.1002/cne.20794PubMedCrossRef Kobayashi K, Fukuoka T, Obata K, Yamanaka H, Dai Y, Tokunaga A, Noguchi K: Distinct expression of TRPM8, TRPA1, and TRPV1 mRNAs in rat primary afferent neurons with adelta/c-fibers and colocalization with trk receptors. J Comp Neurol 2005, 493: 596–606. 10.1002/cne.20794PubMedCrossRef
28.
Zurück zum Zitat Zhu ZW, Friess H, Wang L, Bogardus T, Korc M, Kleeff J, Buchler MW: Nerve growth factor exerts differential effects on the growth of human pancreatic cancer cells. Clin Cancer Res 2001, 7: 105–12.PubMed Zhu ZW, Friess H, Wang L, Bogardus T, Korc M, Kleeff J, Buchler MW: Nerve growth factor exerts differential effects on the growth of human pancreatic cancer cells. Clin Cancer Res 2001, 7: 105–12.PubMed
29.
Zurück zum Zitat Kerr BJ, Bradbury EJ, Bennett DL, Trivedi PM, Dassan P, French J, Shelton DB, McMahon SB, Thompson SW: Brain-derived neurotrophic factor modulates nociceptive sensory inputs and NMDA-evoked responses in the rat spinal cord. J Neurosci 1999, 19: 5138–48.PubMed Kerr BJ, Bradbury EJ, Bennett DL, Trivedi PM, Dassan P, French J, Shelton DB, McMahon SB, Thompson SW: Brain-derived neurotrophic factor modulates nociceptive sensory inputs and NMDA-evoked responses in the rat spinal cord. J Neurosci 1999, 19: 5138–48.PubMed
30.
Zurück zum Zitat Chaudhry V, Giuliani M, Petty BG, Lee D, Seyedsadr M, Hilt D, Cornblath DR: Tolerability of recombinant-methionyl human neurotrophin-3 (r-metHuNT3) in healthy subjects. Muscle Nerve 2000, 23: 189–92. 10.1002/(SICI)1097-4598(200002)23:2<189::AID-MUS7>3.0.CO;2-8PubMedCrossRef Chaudhry V, Giuliani M, Petty BG, Lee D, Seyedsadr M, Hilt D, Cornblath DR: Tolerability of recombinant-methionyl human neurotrophin-3 (r-metHuNT3) in healthy subjects. Muscle Nerve 2000, 23: 189–92. 10.1002/(SICI)1097-4598(200002)23:2<189::AID-MUS7>3.0.CO;2-8PubMedCrossRef
31.
Zurück zum Zitat Watanabe M, Endo Y, Kimoto K, Katoh-Semba R, Arakawa Y: Inhibition of adjuvant-induced inflammatory hyperalgesia in rats by local injection of neurotrophin-3. Neurosci Lett 2000, 282: 61–4. 10.1016/S0304-3940(00)00842-9PubMedCrossRef Watanabe M, Endo Y, Kimoto K, Katoh-Semba R, Arakawa Y: Inhibition of adjuvant-induced inflammatory hyperalgesia in rats by local injection of neurotrophin-3. Neurosci Lett 2000, 282: 61–4. 10.1016/S0304-3940(00)00842-9PubMedCrossRef
32.
Zurück zum Zitat Mantyh WG, Jimenez-Andrade JM, Stake JI, Bloom AP, Kaczmarska MJ, Taylor RN, Freeman KT, Ghilardi JR, Kuskowski MA, Mantyh PW: Blockade of nerve sprouting and neuroma formation markedly attenuates the development of late stage cancer pain. Neuroscience 2010., 1: 171(2):588–98 Mantyh WG, Jimenez-Andrade JM, Stake JI, Bloom AP, Kaczmarska MJ, Taylor RN, Freeman KT, Ghilardi JR, Kuskowski MA, Mantyh PW: Blockade of nerve sprouting and neuroma formation markedly attenuates the development of late stage cancer pain. Neuroscience 2010., 1: 171(2):588–98
33.
Zurück zum Zitat Ozawa T, Ohtori S, Inoue G, Aoki Y, Moriya H, Takahashi K: The degenerated lumbar intervertebral disc is innervated primarily by peptide-containing sensory nerve fibers in humans. Spine 2006, 31: 2418–22. 10.1097/01.brs.0000239159.74211.9cPubMedCrossRef Ozawa T, Ohtori S, Inoue G, Aoki Y, Moriya H, Takahashi K: The degenerated lumbar intervertebral disc is innervated primarily by peptide-containing sensory nerve fibers in humans. Spine 2006, 31: 2418–22. 10.1097/01.brs.0000239159.74211.9cPubMedCrossRef
34.
Zurück zum Zitat Ozawa T, Aoki Y, Ohtori S, Takahashi K, Chiba T, Ino H, Moriya H: The dorsal portion of the lumbar intervertebral disc is innervated primarily by small peptide-containing dorsal root ganglion neurons in rats. Neurosci Lett 2003, 344: 65–7. 10.1016/S0304-3940(03)00416-6PubMedCrossRef Ozawa T, Aoki Y, Ohtori S, Takahashi K, Chiba T, Ino H, Moriya H: The dorsal portion of the lumbar intervertebral disc is innervated primarily by small peptide-containing dorsal root ganglion neurons in rats. Neurosci Lett 2003, 344: 65–7. 10.1016/S0304-3940(03)00416-6PubMedCrossRef
35.
Zurück zum Zitat Mach DB, Rogers SD, Sabino MC, Luger NM, Schwei MJ, Pomonis JD, Keyser CP, Clohisy DR, Adams DJ, O'Leary P, Mantyh PW: Origins of skeletal pain: sensory and sympathetic innervation of the mouse femur. Neuroscience 2002, 113: 155–66. 10.1016/S0306-4522(02)00165-3PubMedCrossRef Mach DB, Rogers SD, Sabino MC, Luger NM, Schwei MJ, Pomonis JD, Keyser CP, Clohisy DR, Adams DJ, O'Leary P, Mantyh PW: Origins of skeletal pain: sensory and sympathetic innervation of the mouse femur. Neuroscience 2002, 113: 155–66. 10.1016/S0306-4522(02)00165-3PubMedCrossRef
36.
Zurück zum Zitat Davies AM: Regulation of neuronal survival and death by extracellular signals during development. EMBO J 2003, 22: 2537–45. 10.1093/emboj/cdg254PubMedCentralPubMedCrossRef Davies AM: Regulation of neuronal survival and death by extracellular signals during development. EMBO J 2003, 22: 2537–45. 10.1093/emboj/cdg254PubMedCentralPubMedCrossRef
37.
Zurück zum Zitat Ernsberger U: Role of neurotrophin signalling in the differentiation of neurons from dorsal root ganglia and sympathetic ganglia. Cell Tissue Res 2009, 336: 349–84. 10.1007/s00441-009-0784-zPubMedCrossRef Ernsberger U: Role of neurotrophin signalling in the differentiation of neurons from dorsal root ganglia and sympathetic ganglia. Cell Tissue Res 2009, 336: 349–84. 10.1007/s00441-009-0784-zPubMedCrossRef
38.
Zurück zum Zitat McMahon SB, Bennett DLH, Bevan S, Wall PD, McMahon SB, Koltzenburg M: Inflamatory mediators and modulators of pain. Journal/Wall and Melzack's Textbook of Pain 2006, 49–72.CrossRef McMahon SB, Bennett DLH, Bevan S, Wall PD, McMahon SB, Koltzenburg M: Inflamatory mediators and modulators of pain. Journal/Wall and Melzack's Textbook of Pain 2006, 49–72.CrossRef
39.
Zurück zum Zitat Devor M, Wall PD: Type of sensory nerve fibre sprouting to form a neuroma. Nature 1976, 262: 705–8. 10.1038/262705a0PubMedCrossRef Devor M, Wall PD: Type of sensory nerve fibre sprouting to form a neuroma. Nature 1976, 262: 705–8. 10.1038/262705a0PubMedCrossRef
40.
Zurück zum Zitat Sung JH, Mastri AR: Aberrant peripheral nerves and microneuromas in otherwise normal medullas. J Neuropathol Exp Neurol 1983, 42: 522–8. 10.1097/00005072-198309000-00004PubMedCrossRef Sung JH, Mastri AR: Aberrant peripheral nerves and microneuromas in otherwise normal medullas. J Neuropathol Exp Neurol 1983, 42: 522–8. 10.1097/00005072-198309000-00004PubMedCrossRef
41.
Zurück zum Zitat Madduri S, Papaloizos M, Gander B: Synergistic effect of GDNF and NGF on axonal branching and elongation in vitro. Neurosci Res 2009, 65: 88–97. 10.1016/j.neures.2009.06.003PubMedCrossRef Madduri S, Papaloizos M, Gander B: Synergistic effect of GDNF and NGF on axonal branching and elongation in vitro. Neurosci Res 2009, 65: 88–97. 10.1016/j.neures.2009.06.003PubMedCrossRef
42.
Zurück zum Zitat Kryger GS, Kryger Z, Zhang F, Shelton DL, Lineaweaver WC, Buncke HJ: Nerve growth factor inhibition prevents traumatic neuroma formation in the rat. J Hand Surg Am 2001, 26: 635–44. 10.1053/jhsu.2001.26035PubMedCrossRef Kryger GS, Kryger Z, Zhang F, Shelton DL, Lineaweaver WC, Buncke HJ: Nerve growth factor inhibition prevents traumatic neuroma formation in the rat. J Hand Surg Am 2001, 26: 635–44. 10.1053/jhsu.2001.26035PubMedCrossRef
43.
Zurück zum Zitat Black JA, Nikolajsen L, Kroner K, Jensen TS, Waxman SG: Multiple sodium channel isoforms and mitogen-activated protein kinases are present in painful human neuromas. Ann Neurol 2008, 64: 644–53. 10.1002/ana.21527PubMedCrossRef Black JA, Nikolajsen L, Kroner K, Jensen TS, Waxman SG: Multiple sodium channel isoforms and mitogen-activated protein kinases are present in painful human neuromas. Ann Neurol 2008, 64: 644–53. 10.1002/ana.21527PubMedCrossRef
44.
Zurück zum Zitat Devor M, Keller CH, Ellisman MH: Spontaneous discharge of afferents in a neuroma reflects original receptor tuning. Brain Res 1990, 517: 245–250. 10.1016/0006-8993(90)91033-DPubMedCrossRef Devor M, Keller CH, Ellisman MH: Spontaneous discharge of afferents in a neuroma reflects original receptor tuning. Brain Res 1990, 517: 245–250. 10.1016/0006-8993(90)91033-DPubMedCrossRef
45.
Zurück zum Zitat Devor M, Govrin-Lippmann R: Axoplasmic transport block reduces ectopic impulse generation in injured peripheral nerves. Pain 1983, 16: 73–85. 10.1016/0304-3959(83)90087-8PubMedCrossRef Devor M, Govrin-Lippmann R: Axoplasmic transport block reduces ectopic impulse generation in injured peripheral nerves. Pain 1983, 16: 73–85. 10.1016/0304-3959(83)90087-8PubMedCrossRef
46.
Zurück zum Zitat Devor M, Govrin-Lippmann R, Angelides K: Na+ channel immunolocalization in peripheral mammalian axons and changes following nerve injury and neuroma formation. J Neurosci 1993, 13: 1976–92.PubMed Devor M, Govrin-Lippmann R, Angelides K: Na+ channel immunolocalization in peripheral mammalian axons and changes following nerve injury and neuroma formation. J Neurosci 1993, 13: 1976–92.PubMed
47.
Zurück zum Zitat Ceyhan GO, Bergmann F, Kadihasanoglu M, Altintas B, Demir IE, Hinz U, Muller MW, Giese T, Buchler MW, Giese NA, Friess H: Pancreatic neuropathy and neuropathic pain-a comprehensive pathomorphological study of 546 cases. Gastroenterology 2009, 136: 177–186 e1. 10.1053/j.gastro.2008.09.029PubMedCrossRef Ceyhan GO, Bergmann F, Kadihasanoglu M, Altintas B, Demir IE, Hinz U, Muller MW, Giese T, Buchler MW, Giese NA, Friess H: Pancreatic neuropathy and neuropathic pain-a comprehensive pathomorphological study of 546 cases. Gastroenterology 2009, 136: 177–186 e1. 10.1053/j.gastro.2008.09.029PubMedCrossRef
48.
Zurück zum Zitat Winkler J: Analgesic effects of a potent and selective kinase inhibitor of neurotrophin receptors TrkA, TrkB, and TrkC. Journal/8th IASP Research Symposium 2009. Winkler J: Analgesic effects of a potent and selective kinase inhibitor of neurotrophin receptors TrkA, TrkB, and TrkC. Journal/8th IASP Research Symposium 2009.
49.
Zurück zum Zitat Bouhana KS, Impastato R, Jiang Y, Wallace RD, Hartley DP, Do MG, von Carlowitz I: Analgesic effects of a potent and selective kinase inhibitor of neurotrophin receptors TrkA, TrkB, and TrkC in a model of inflammatory pain. Journal/Keystone Symposium- Neurobiology of Pain 2008, in press. Bouhana KS, Impastato R, Jiang Y, Wallace RD, Hartley DP, Do MG, von Carlowitz I: Analgesic effects of a potent and selective kinase inhibitor of neurotrophin receptors TrkA, TrkB, and TrkC in a model of inflammatory pain. Journal/Keystone Symposium- Neurobiology of Pain 2008, in press.
Metadaten
Titel
Administration of a tropomyosin receptor kinase inhibitor attenuates sarcoma-induced nerve sprouting, neuroma formation and bone cancer pain
verfasst von
Joseph R Ghilardi
Katie T Freeman
Juan M Jimenez-Andrade
William G Mantyh
Aaron P Bloom
Michael A Kuskowski
Patrick W Mantyh
Publikationsdatum
01.12.2010
Verlag
BioMed Central
Erschienen in
Molecular Pain / Ausgabe 1/2010
Elektronische ISSN: 1744-8069
DOI
https://doi.org/10.1186/1744-8069-6-87

Weitere Artikel der Ausgabe 1/2010

Molecular Pain 1/2010 Zur Ausgabe

Tennisarm: „Ein bisschen Physio würde ich mich schon trauen“

06.06.2024 Schmerzsyndrome Nachrichten

Eine Therapie mit nachgewiesenem Nutzen gibt es bei Epicondylitis lateralis derzeit nicht. Das heißt jedoch nicht, dass man die Betroffenen mit ihren Beschwerden allein lassen sollte, so der Rat eines Experten. Gute Erfahrungen habe er zum Beispiel mit der Stoßwellentherapie gemacht.

Sechs Maßnahmen gegen Regelschmerzen – von Krafttraining bis Yoga

05.06.2024 Dysmenorrhö Nachrichten

Um herauszufinden, wie gut sich körperliche Übungen dafür eignen, primäre Dysmenorrhö zu lindern, hat eine Studiengruppe sechs verschiedene Verfahren untersucht. Alle wirkten, doch eine Methode war Favorit.

Hereditäres Angioödem: Tablette könnte Akuttherapie erleichtern

05.06.2024 Hereditäres Angioödem Nachrichten

Medikamente zur Bedarfstherapie bei hereditärem Angioödem sind bisher nur als Injektionen und Infusionen verfügbar. Der Arzneistoff Sebetralstat kann oral verabreicht werden und liefert vielversprechende Daten.

Intoxikation ohne Alkoholaufnahme: An das Eigenbrauer-Syndrom denken!

05.06.2024 Internistische Diagnostik Nachrichten

Betrunken trotz Alkoholabstinenz? Der Fall einer 50-jährigen Patientin zeigt, dass dies möglich ist. Denn die Frau litt unter dem Eigenbrauer-Syndrom, bei dem durch Darmpilze eine alkoholische Gärung in Gang gesetzt wird.

Update AINS

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