Potential Uses of Isolated Toxin Peptides in Neuropathic Pain Relief: A Literature Review

Potential Uses of Isolated Toxin Peptides in Neuropathic Pain Relief: A Literature Review

Accepted Manuscript Potential Uses of Isolated Toxin Peptides in Neuropathic Pain Relief: A Literature Review M.K. Hamad, K. He, H.F. Abdulrazeq, A.M...

1MB Sizes 0 Downloads 21 Views

Accepted Manuscript Potential Uses of Isolated Toxin Peptides in Neuropathic Pain Relief: A Literature Review M.K. Hamad, K. He, H.F. Abdulrazeq, A.M. Mustafa, J. Nakhla, M.M. Herzallah, A. Mammis PII:

S1878-8750(18)30159-1

DOI:

10.1016/j.wneu.2018.01.116

Reference:

WNEU 7314

To appear in:

World Neurosurgery

Received Date: 22 August 2017 Revised Date:

15 January 2018

Accepted Date: 16 January 2018

Please cite this article as: Hamad M, He K, Abdulrazeq H, Mustafa A, Nakhla J, Herzallah M, Mammis A, Potential Uses of Isolated Toxin Peptides in Neuropathic Pain Relief: A Literature Review, World Neurosurgery (2018), doi: 10.1016/j.wneu.2018.01.116. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT

Potential Uses of Isolated Toxin Peptides in Neuropathic Pain Relief: A Literature Review Department of Neurological Surgery, Rutgers University- New Jersey Medical School

RI PT

Hamad MK1, He K1, Abdulrazeq HF1, Mustafa AM2, Nakhla J2, Herzallah MM3, 4, and Mammis A1

1- Department of Neurological Surgery, Rutgers University – New Jersey Medical School 2- Albert Einstein College of Medicine

M AN U

SC

3- Center for Molecular and Behavioral Neuroscience, Rutgers University 4- Palestinian Neuroscience Initiative, Al-Quds University

TE D

Disclosures: The authors of this literature review have no conflicts of interest and no disclosures.

EP

Funding: This research did not receive any specific grant from funding agencies in the public, commercial or not-for-profit sectors.

AC C

Corresponding Author: Mousa K. Hamad

[email protected]

90 Bergen Street, Suite 8100 Newark, NJ, 07101

Attn: Antonios Mammis, MD 973-626-2376

1

ACCEPTED MANUSCRIPT

Abstract: Neuropathic pain is a subset of chronic pain that is caused by neurons that are damaged or firing aberrantly in the peripheral or central nervous systems. The treatment guidelines for neuropathic pain

RI PT

include antidepressants, calcium channel α 2 delta ligands, topical therapy, and opioids as a second line option. Unfortunately, pharmacotherapy has not been effective in the treatment of neuropathic pain except in the treatment of trigeminal neuralgia with carbamazepine. The inability to properly treat neuropathic

SC

pain causes frustration in both the patients and their treating physicians. Venoms, which are classically thought to be causes of pain and death, have peptide components that have been implicated in pain relief.

M AN U

Although some venoms are efficacious and have shown benefits in patients, their side effect profile precludes their more widespread use. This review identifies and explores the use of venoms in neuropathic pain relief. This can open doors to potential therapeutic targets. We believe that further research into the mechanisms of action of these receptors as well as their functions in nature will provide

Perspective:

TE D

alternative therapies as well as a window into how they affect neuropathic pain.

Understanding the mechanism of action of these venom toxins can direct future investigation into possible

EP

therapies for patients with refractory neuropathic pain. Research should be directed at utilizing venoms in

AC C

additional beneficial ways and formulating venoms for the most appropriate pain relief techniques.

Keywords:

Neuropathic pain, venoms, chronic pain, non-opiate analgesia, opioids

2

ACCEPTED MANUSCRIPT

INTRODUCTION Neuropathic pain is a subset of chronic pain that is caused by neurons that are damaged or neurons that are firing aberrantly. It affects 15-19% of the world population, and produces pain that is

RI PT

severe enough to interfere with daily life.4, 17, 148 Classically, neuropathic pain is described as a widespread, burning sensation that initiates in seemingly unprovoked waves and is associated with

allodynia and some sensory deficit. Allodynia is described as a sensation where a normally painless

stimulus produces pain. The initial source can be localized to either the peripheral or central nervous

SC

systems and generally involves pathways connected to nociception.14 Many different disease states

M AN U

(diabetes, multiple sclerosis, viral infection, etc.) can lead to neuropathic pain and there are various mechanistic explanations for the onset of pain in each of these diseases. For example, in trigeminal neuralgia, a disease of the trigeminal nerve which causes debilitating attacks of pain in the V1, V2, and V3 dermatomes, the pain may be caused by nerve compression (most commonly by blood vessels) at the site where the peripheral nerve reaches the brainstem. Postherpetic neuralgia on the other hand is caused

TE D

by viral infiltration of nerve roots leading to increased expression of voltage gated sodium and potassium channels, as well as upregulation of receptors involved in pain pathways.56, 134 It is important to consider that various manifestations of neuropathic pain may require different treatment options than those

EP

currently available.

The search for safe and effective pain control is pertinent now more than ever. Current treatment

AC C

guidelines for neuropathic pain usually start with non-opioid analgesics (such as NSAIDs, acetaminophen, and aspirin) followed by tramadol. Severe non-remitting pain is then treated with opioids, which are classically addictive and can lead to potential abuse. Currently, α2 adrenergic agonists, antidepressants (Tricyclics and SNRIs), and antiepileptics (gabapentin, pregabalin, etc.) are treatment options for opioid-resistant pain.126 These traditional medications have shown limited effectiveness. The addictive potential of opioids and severe side-effect profiles of these drugs when used chronically have also been problematic.

3

ACCEPTED MANUSCRIPT

The rising tide of opiate addiction necessitates new research on pain relief. According to the Centers for Disease Control and Prevention, the amount of prescription opioids sold in the US as well as number of deaths from prescription opioids have quadrupled since 1999. Prescription narcotics continue

RI PT

to be widely prescribed, since no effective alternative is available to patients with chronic neuropathic pain.

Venoms, whether they are from snakes, lizards, leeches, snails, scorpions, mammals, or spiders,

SC

are traditionally sources of severe pain or death. However, analyzing the structure and functions of these venoms in preliminary studies has demonstrated various channel blockers that are more potent and

M AN U

effective than current treatment options for neuropathic pain. In this review, the authors have identified a list of toxins that have been used in animal studies or clinical trials with the potential for therapeutic use. The authors have only identified one venom-derived analgesic that is currently in practice for severe intractable neuropathic pain; however, its severe side-effect profile limits its usefulness. An isolated venom peptide, which goes by the generic name Ziconotide, is a synthetic version

TE D

of ω-conotoxin MVIIA derived from the venom of Conus magus, a marine snail. Conotoxins are small peptides derived from the genus Conus, which are all venomous predators. Three different types, α, µ, and ω, interact with a variety of receptors involved in regulating pain. The α-conotoxin interacts with nAchR,

EP

the ω-conotoxins interacts with both Nav and Cav channels, and the µ-conotoxin targets Nav channels. The ω-conotoxins, from which ziconotide is derived, paralyze prey after VGCCs are inhibited. They have

AC C

been studied extensively for their role in pain relief.65 Ziconotide is currently available as an intrathecal injection after successfully completing three double-blind, placebo-controlled trials and is in use for management of severe chronic pain.65 Although its usefulness as an analgesic is apparent, Ziconotide has an extensive side effect profile including rhabdomyolysis, sleep disturbance, and headache among many others.65, 106 Therefore, with increasing knowledge about options for non-opioid analgesics, it is essential to continue research for safer efficacious alternatives.

4

ACCEPTED MANUSCRIPT

nAChR INHIBITORS

α-conotoxins:

RI PT

In reviewing the breadth of research on animal toxins for the treatment of neuropathic pain, the family of conotoxins from which ziconotide is derived comes into sight. Conotoxins, as inhibitors of a variety of ion channels, present as strong candidates in the field of pain relief and as potential alternatives

SC

to ziconotide because of its adverse side effects and invasive route of administration.65

Sodium channels have been found to play a significant role in the mechanism of pain. After nerve injury, it was found that sodium channels began to accumulate at the site of injury and along the length of

M AN U

the axon.122 This resulted in hyperexcitability and spontaneous action potential discharges. Another group also found that nociceptors spontaneously fired after nerve injury.125 A popular but controversial route of analgesia revolves around α9 nicotinic acetylcholine receptors (α9nAChRs) and their relationship with αconotoxins. Although evidence for the involvement of α9nAChRs, and the α9α10 nAChR in particular, in

TE D

pain treatment has been found,97, 164 the types of pain these receptors may be related to as well as their exact mechanism of action have been contested.107, 114 Further research indicates that the putative α9nAChR pain-alleviation mechanism may not require the α9nAChR, but rather incorporate a GABA-

EP

dependent N-type voltage gated Ca2+ channel pathway;21, 22, 32, 77, 114, 115 recent evidence both supports and precludes the GABA-dependent calcium channel inhibition mechanism of pain relief through α-

AC C

conotoxins.67, 171 As of yet, the true mechanism remains unclear. Three α-conotoxins of interest are the peptides α-conotoxin Vc1.1, α-conotoxin RgIA, and α-conotoxin GeXIVA. 1. α-conotoxin Vc1.1 This toxin, discovered from the mRNA sequence of the Queen Victoria cone sea snail Conus

victoriae,145 has been analyzed for its nAChR selectivity and binding to α9α10.30 Intramuscular injection of α-conotoxin Vc1.1 dose-dependently attenuated mechanical hyperalgesia in chronic constriction injury (CCI) and partial nerve ligation (PNL) models of neuropathic pain.146, 163 The former model calls for exposure and multiple ligations of the sciatic nerve, usually resulting in forms of hyperalgesia and 5

ACCEPTED MANUSCRIPT

allodynia. Acute treatment of CCI rats with an intramuscular bolus of α-conotoxin Vc1.1 led to significant attenuation of hyperalgesia, defined by increased weight threshold prior to paw withdrawal. Compared to saline, rats treated with the toxin were able to tolerate up to approximately double the weight before

RI PT

withdrawing from pain. This effect lasted between 1 and 3 hours after injection. Short term effects lasted 24 hours after injection and rats who received the maximal dose were restored to pre-operative weight levels before withdrawing. Long term effects yielded significant attenuation of mechanical hyperalgesia

SC

even at the lowest dose with 50% of attenuation lasting one week after injections.146 Moreover, treatment with substance P illustrated a significantly greater vascular inflammatory response 8 weeks post-injury in

M AN U

rats treated with Vc1.1 compared to saline control, suggesting an enhanced recovery of injured neurons. Using normal rats as the control for inflammation at 100% post treatment with substance P, rats treated with Vc1.1 demonstrated up to 83% functional recovery while those treated with saline showed 47%.146 Intramuscular administrations of Vc1.1 have also reduced allodynia in PNL and other neuropathic pain models.77, 188 Intrathecal administration of Vc1.1 dose-dependently reduced mechanical allodynia without

TE D

adverse motor effects as evidenced by rotarod test. Rats treated with the conotoxin had increased paw withdrawal thresholds for up to 6 hours while those treated with saline had no changes in threshold.114 However, other investigators have suggested that the mechanism of conopeptides are dependent on

EP

pathways other than Vc1.1.171 However, other investigators have suggested that the mechanism of conopeptides are dependent on pathways other than Vc1.1.171 They found that conopeptides had variable

AC C

effects on Vc1.1. in different sensory neurons and that there was only minimal inhibition via this pathway. Further research needs to be done to determine the true pathway of analgesia for conopeptides so that improved therapies may be developed. 2. α-conotoxin RgIA Found in the crown cone sea snail Conus regius,51 this toxin also blocks the α9α10 nAChR, and has been examined for its binding mechanism and stability as a potential drug.7, 51, 52 Intramuscular administrations of RgIA in CCI neuropathic rat models attenuated hyperalgesia and allodynia in a dosedependent manner through chronic and acute treatment, without noted adverse effects. In one study, CCI 6

ACCEPTED MANUSCRIPT

rats treated chronically for 14 days with RgIA had progressive decrease in paw withdrawal threshold; 61.7 ± 0.6 g and 65.0 ± 0.9 g in 2 nmol- and 10 nmol-treated group compared to 39.2 ± 1.3 g in the saline treated group.44, 163 Of note is RgIA’s ability to decrease degeneration of both dorsal root ganglion and

RI PT

sciatic nerve morphology in the CCI model as well as reduce the glial cell-mediated inflammatory response after intramuscular injection.44 Finally, a recent study using a chemotherapeutic drug-induced (oxaliplatin) model of neuropathic pain revealed dose-dependent reductions in mechanical hyperalgesia

SC

and mechanical and cold allodynia, alongside prevention of DRG degeneration after intramuscular

injection of RgIA conducted at a set dosage. This study found resolution of pain threshold over the control group value after 21 days of treatment with high dose RgIA. No tolerance to the toxin’s effect was

M AN U

noted.129 In the spinal cord, although no significant effects were seen in microglial expression, cotreatment of RgIA and oxaliplatin reduced astrocyte number, measured by glial fibrillary acidic protein (GFAP) expression, compared to oxaliplatin alone.129 Interestingly, RgIA alone increased astrocyte number compared to oxaliplatin alone.129

TE D

3. αO-conotoxin GeXIVA

In addition to the former two toxins, another conotoxin, αO-conotoxin GeXIVA, isolated from the general cone sea snail Conus generalis, was recently identified and studied for its properties.98 Similar

EP

to Vc1.1 and RgIA, GeXIVA inhibits the α9α10 nAChR and presented significant analgesic effects in the area of neuropathic pain.86, 98 There are three isoforms of GeXIVA: [1,2] (bead form), [1,3] (globular

AC C

form), and [1] (ribbon form), with the [1,2] isomer most potently inhibiting α9α10 nAChR.98 In a CCI rat model, GeXIVa [1,2] produced dose-dependent attenuation of mechanical hyperalgesia comparable to morphine after intramuscular injection, with no significant deficits in motor control as evidenced by a rotarod test.98 An additional study of the [1] isomer also showed significant dose-dependent reduction in mechanical hyperalgesia through intramuscular administration, alongside the slightly better effect of the [1,2] isomer and similar non-significance in the rotarod test.86 The significant effects of [1] only applied to single, acute, short term injection patterns; the pain-relieving effect lost significance by one week after

7

ACCEPTED MANUSCRIPT

treatment periods.86 No motor performance effects were found, nor were any behavior disturbances noted.86 Overall, in spite of the contradicting evidence and uncertainty surrounding the relevancy of

RI PT

α9α10 nAChR and GABA-dependent VGCC pathways of pain relief, these α-conotoxins have clear potential in the treatment of neuropathic pain. Future research will likely elucidate the exact mechanism of pain alleviation and the scope of the toxins’ effects, as well as reveal other novel toxins with

SC

therapeutic implications.

M AN U

CbTx and Haditoxin

Two final toxins to note in the family of nAChR inhibitors are cobratoxin (CbTx) and haditoxin. CbTx is believed to inhibit the α7 nAChR as determined by known α7 nAChR agonists and antagonists.58 CbTx, isolated from the Southeast Asian monocled cobra Naja naja kaouthia, is an α-cobratoxin that dose dependently diminished thermal and mechanical hyperalgesia, defined as Thermal Withdrawal Latency

TE D

(TWL) and Mechanical Withdrawal Threshold (MWT) respectively, in a PNL rat model after intrathecal injection. The data show an almost two-fold increase in MWT and a 1.5sec increase in TWL 15 minutes after intrathecal injection of high dose CbTx compared to saline.58 This effect remained significant for

EP

less than an hour. No information on side effects was provided.58 Additionally, CbTx and a derived analog, receptin, inhibit thermal, arthritic, and chemically-induced inflammatory and abdominal writhing

AC C

pain in animal models.29, 94, 152 Haditoxin, on the other hand, has been shown to have an effect on human nAChRs with a strong preference for α7 nAChR, but no studies have yet been conducted on its applications to neuropathic pain.140

MULTIPLE SITE INHIBITION Bee Venom

8

ACCEPTED MANUSCRIPT

Bee venom, or apitoxin, is a mixture of biologically-active chemicals from the honey bee Apis mellifera. It includes bioamines, phospholipase A2, apamin, melittin, and mast cell degradation peptide.1, 26, 156

Bee venom is a relatively new toxin being studied for its clinical applications; however, bee venom

RI PT

therapy has prevailed in traditional Middle Eastern and Eastern medicine for many years and is still in practice today.1, 26

Diluted bee venom (DBV) has been the focus of current research surrounding its use in

acupuncture, known as apipuncture. There are data suggesting DBV’s efficacy as an anti-inflammatory as

SC

well as an analgesic for neuropathic and non-neuropathic pain.155, 156 A slew of studies have been conducted on subcutaneous DBV apipuncture’s attenuating effect on allodynia, including oxaliplatin-

M AN U

induced models of neuropathic pain,75 and spinal nerve ligation models.75, 79, 82, 90, 181, 185 In a spinal cord injury-induced pain rat model, DBV appears to reduce central neuropathic pain, described by an over two-fold increase in mechanical withdrawal threshold and two second increase in thermal withdrawal latency compared to the saline treated group.72 The spinal cord injury model involved spinal cord

TE D

hemisection-induced thermal hyperalgesia and allodynia; after repetitive acupunctural DBV treatment was administered 1-5 days post-surgery, a significantly greater effect in neuropathic pain alleviation was discovered compared to treatment 15-20 days post-surgery and controls.72 This indicates a potential early

EP

benefit of DBV. Repetitive DBV treatment also produced significantly faster motor function recovery in the spinal cord injury model. In this study, the effect of DBV was noted mostly when it was applied to the

AC C

ipsilateral dorsal root ganglion, suggesting a key role of the dorsal horn in DBV’s mechanism of action.72 Similarly, in a chronic constriction injury rat model, repetitive non-acupuntural subcutaneous DBV injections alleviated thermal hyperalgesia.71 Another study using CCI and subcutaneous apipunctural single injection of DBV reported significant effects on thermal hyperalgesia, but none on mechanical allodynia.138 The mechanism by which bee venom functions appears to involve a complex variety of pathways that is not yet fully understood.25 A proposed mechanism that involves glial cell inhibition cited lack of upregulation of GFAP and Iba-1 as evidence for DBV reduction of glial cell activation.72 Normally, after spinal cord injury, glial scar formation is induced by way of glial cell activation and 9

ACCEPTED MANUSCRIPT

recruitment, and biomarkers such as GFAP and Iba-1 are upregulated in the process. Adrenergic, serotonergic, and cholinergic systems have all been implicated.82, 138, 182 Also, in the latter chronic compression injury model, repetitive subcutaneous DBV injection may alleviate pain by way of

RI PT

adrenoceptors and increased noradrenergic activity in the locus coeruleus.71 Furthermore, bee venom was presented as a novel method of potentiating the analgesic effects of clonidine, an α2-adrenoreceptor antagonist, in neuropathic thermal hyperalgesia and mechanical

SC

allodynia.70, 184 Subcutaneous apipuncture of DBV with clonidine attenuated pain comparable to higher doses of clonidine alone and reduced the side effect profile. Rats treated with DBV and clonidine together had a 3.5-fold leftward shift in clonidine’s dose-response curve acutely and a 5-fold leftward shift during

M AN U

late phase treatment.184 A similar premise was used in a study comparing DBV with morphine, which displayed stronger and longer lasting effects of cold and mechanical allodynia in the group with combined DBV and morphine administration compared to DBV or morphine alone.75 Although bee venom does pose potential harm to humans due to inherent risks of reactions

TE D

toward bee venom (e.g. anaphylaxis),26 its use in therapy persists. There exist uncontrolled trials on bee venom therapy and prospective studies on its application as alternative medicine.183 However, there are also case reports on instances where a bee sting or bite induced immune thrombocytopenia (ITP)1, 2, 113, 153, Of note is a case report in which the patient presenting with ITP had undergone bee venom

EP

156, 160

therapy.1 This last report draws attention to a potential complication in bee venom therapy; however,

AC C

given the lack of information on the administration of the therapy in this particular case as well as lack of dosing control during other reports,1, 153, 160 the true benefits and harms are still unclear. Despite the research detailing bee venom’s potential benefit in animal models and in humans, it does cause many of the same pain symptoms that it seeks to treat.26 However, further investigation with improved study models of diluted bee venom and its potential to alleviate neuropathic pain is well warranted.

10

ACCEPTED MANUSCRIPT

Crotoxin Crotoxin, isolated from the venom of the South American rattlesnake Crotalus durissus terrificus,

RI PT

shows analgesic effects through opioid-independent pathways.190 Muscarinic, serotonergic, and adrenergic receptor antagonists all inhibited the anti-hyperalgesic effect of directly applied crotoxin on sciatic nerve transection rats.121 However, studies have provided contradicting evidence regarding the involvement of the cholinergic system with varying results dependent on the dosage of anticholinergic

SC

chemicals.121, 190 Direct application of crotoxin to transected branches of sciatic nerve in rats immediately after surgery also showed a long lasting (64 days) reduction in hyperalgesia, whereas subcutaneous

M AN U

administration only produced a short term alleviation of pain (between 2 and 24 hours).121 No adverse effects on motor activity were noted after either direct application or intraperitoneal crotoxin injection.121, 169

Further evidence of crotoxin’s potential use in analgesia comes from an early Phase I clinical trial

TE D

for cancer treatment in which the toxin was administered intramuscularly for the treatment of solid tumors.33 Although pain relief was not a primary endpoint of the experiment, a majority (18/23) of the evaluated patients experienced a decrease in cancer-related pain illustrated by a reduction in analgesic use

AC C

EP

and patient input.33

VOLTAGE-GATED CALCIUM CHANNEL INHIBITORS CVIE and VIF

CVIE and CVIF are conotoxins isolated from a cDNA library of the Conus catus. They act by

inhibiting N-type VGCCs. The peptides were found to reduce allodynic behavior in the rat PNL model.11 They inhibited the Cav2.2 channel in a reversible and voltage-dependent manner in the dorsal horn of the spinal cord. CVIE in particular was found to have the highest potency in inhibiting N-type VGCCs and was also able to partially reverse impaired weight bearing in rats when given subcutaneously.141 No major 11

ACCEPTED MANUSCRIPT

side effects when administering the conotoxins systemically were observed. Due to the potency and lack of side effects seen thus far in these studies, CVIE seems a promising candidate for the treatment of neuropathic pain. However, it is important to note the limitation of the data as studies have only been

RI PT

conducted in rat models and CVIE and CVIF have not been studied pharmacologically in humans.

Phα1β

Phα1β is a toxin isolated from the venom of the Brazilian wandering spider Phoneutria

SC

nigriventer. It reversibly inhibits N-type VGCCs.162 It has only recently been studied for its potent effects

M AN U

on the nervous system in various forms of pain and pruritus in animal models.36, 37, 101 Not only does Phα1β appear to possess a broad range of applications in pain management, but it also shows promise over ziconotide in treatment of neuropathic, inflammatory, post-operative, visceral, and cancer pain in relation to strength and duration of analgesia and mitigation of side effect profile in some but not all pain models.24, 36, 37, 46, 135, 136

TE D

Its proposed mechanism of action implicates the N-type VGCC, which has an important role in the production of pain as evidenced by ziconitide. Typically, VGCCs are activated upon receipt of depolarizing stimulus, causing an intracellular cascade that results in exocytotic release of vesicular

EP

compounds in the synaptic terminal.132 The N-type channel itself is associated with neuronal pain induction,132 exhibiting colocalization with substance P and inhibition through a µ-opioid receptor

AC C

signaling pathway.150, 168

In the management of neuropathic pain, Phα1β shows a significant benefit in chronic constrictive

injury rat models and chemotherapeutic-induced models of neuropathic pain.37, 135, 139 In one study, rats were either intrathecally injected with a single dose of Phα1β 8 days post injection, or continuously administered the toxin for 7 days against a control.139 Results showed significant reduction in CCIinduced neuropathic pain in both cases, with the single injection attenuating pain by up to 100% for 1-6 hours and the continuous administration for 1-7 days by 63% ± 13%.139 In the paclitaxel-induced chronic neuropathic pain model, single intrathecal injection of Phα1β significantly attenuated mechanical 12

ACCEPTED MANUSCRIPT

hyperalgesia in a dose-dependent manner for 4 hours after long term administration of paclitaxel. In the acute pain stage, rats treated with Phα1β showed a maximum inhibition of mechanical pain of 100%, and those in the chronic phase had a maximum inhibition of 81 ± 10% with no statistically significant effect

RI PT

on control rats.135 Importantly, no significant behavioral side effects ensued from neither continuous treatment with Phα1β nor low analgesic doses;135, 139 dynamic allodynia significantly presented in the highest analgesic dose of Phα1β, but the comparative side effect of ziconotide was greater.135 The

Other Toxins of Phoneutria nigriventer

M AN U

harmful side effects associated with ziconotide.37, 135, 139

SC

intrathecal administration of Phα1β displays similar if not greater efficacy than ziconotide, without the

The venom of Phoneutria nigriventer also contains several less-studied toxins with potential pain-alleviating properties. In a mechanistic overview of these toxins, δ-Ctenitoxin-Pn1a, formerly known as PnTx4(6-1), binds to insect axonal voltage-gated sodium channels (VGSCs) to prolong the action

TE D

potential via delay of Na+ channel inactivation, but does not have an effect on the electrophysiology of mammalian Na+ channels.35 PnTx4(5-5) is another toxin from Phoneutria nigriventer that is cited to possess analgesic properties;53 however, aside from one of its recombinant forms having an effect on

EP

increasing depolarization threshold of VGSCs, there has been little research on its relationship with pain.53, 130 Likewise, rPnTx1 acts as a VGSC inhibitor and competes with µ-conotoxin for binding sites,

AC C

but has not yet been studied for pain treatment.102, 154 The toxin Tx3-3 inhibit P/Q, R, and L-type VGCCs (P/Q and R more effectively) and the toxin Tx3-5 inhibits L-type channels.23, 53, 81 The toxins from Phoneutria nigrivente have demonstrated the ability for global inhibition of VGCCs, which is a promising component in understanding pain inhibition.132 Among the aforementioned toxins, δ-CNTX-Pn1a, Tx3-3, and Tx3-5 all indicate potential for neuropathic pain treatment, albeit only via intrathecal administration.34, 53, 127 δ-CNTX-Pn1a produced significant effects on neuropathic rat models with induced CCI, showing brief (20 min) attenuation of hyperalgesia compared to control, although there was no mention of side effects.53 Similarly, in PNL and 13

ACCEPTED MANUSCRIPT

streptozotocin-induced diabetic neuropathy models, Tx3-3 produced significant alleviation of allodynia with no adverse motor effects commonly associated with VGCC blockade.34 Lastly, administration of Tx3-5 in another PNL model produced a maximum inhibition of mechanical hyperalgesia of

RI PT

87±10%, with no noted adverse effects. This study also tested Tx3-5 on a model of cancer-related pain with morphine tolerance in mice showing complete reversal of melanoma-related mechanical

hyperalgesia.127 Much like Phα1β, these toxins contributed to other areas of pain treatment, including

M AN U

largely untapped potential for treatment in pain management.

SC

inflammatory and post-operational pain.53, 127 As a whole, the toxins of Phoneutria nigriventer present a

SNX-482

SNX-482 is a peptide toxin isolated from the giant baboon spider Hysterocrates gigas and a blocker of a variety of calcium, sodium, and potassium channels;6, 57, 76, 116 it has been used to study the VGCC Cav2.3 in particular.76 Although its putative significance to neuropathic pain lies in its ability to

TE D

block this R-type VGCC, which has been shown to contribute at least in part to pain sensation in inflammatory models,109 recent research comparing wild type mice to Cav2.3 knockouts illustrate no significant difference in the development of neuropathic pain in the form of partial sciatic nerve ligation-

EP

induced mechanical allodynia in mice.177 Regardless, the use of SNX-482 in a spinal nerve ligation rat model of neuropathic pain reduced sensitivity to thermal and mechanical allodynia and issued a dose-

AC C

dependent reduction of C and Ad-fiber nociceptor responses after intrathecal administration,103 indicating a potential for further neuropathic pain research.

VOLTAGE-GATED SODIUM CHANNEL INHIBITORS Tetrodotoxin In addition to VGCCs, the literature shows VGSCs play a significant role in pain transmission.31, 137

The significance of VGSCs was alluded to with the mechanisms of PnTx4(5-5) and PnTx4(6-1). The 14

ACCEPTED MANUSCRIPT

premier animal-derived blocker of VGSCs is the puffer fish venom tetrodotoxin (TTX). A non-peptide, its applications fell under the scrutiny of the scientific community early on,158 and it has accumulated a large wealth of research to support its potential as an analgesic. Analysis and use of the venom in an array of

RI PT

experiments has elucidated not only its mechanism of action but the importance of VGSCs in pain.137 VGSCs have even been characterized into TTX-sensitive (Nav1.1, Nav1.2, Nav1.3, Nav1.4, Nav1.6, Nav1.7) and TTX-resistant (Nav1.5, Nav1.8, Nav1.9) depending on the concentration of TTX required to

SC

block the channel. This has allowed for a variety of studies on the comparative importance and effects of TTX-sensitive and TTX-resistant subtypes of VGSCs in pain. Channels Nav1.3, Nav1.7, Nav1.8, and

M AN U

Nav1.9 have all been implicated in contributing to neuropathic pain.137 However, there exists conflicting evidence from knockout studies that exhibit normal neuropathic pain development in the absence of such channels.137

As for the direct effects of TTX on neuropathic pain, early studies have illustrated its ability to dose-dependently reduce abnormal neuronal activity after sciatic nerve transection through an intravenous

TE D

route of administration and mechanical sensitivity in a spinal nerve ligation (SNL) model of neuropathic pain.99, 128 In the latter study, routes of administration were also compared; intraperitoneal injection of TTX did not significantly reduce mechanical allodynia compared to control, but epidural and direct

EP

administration to the dorsal root ganglion did have a significant effect.99 Although no side effects were mentioned, the concentration of TTX used in the study was below that necessary to block action

AC C

potentials.99 In a chemotherapy-induced neuropathic pain mouse model, subcutaneous TTX was shown to attenuate allodynia and thermal hyperalgesia without adverse side effects, with an additional preventative effect on allodynia pre-administration of chemotherapeutic agent.118 Intramuscular TTX additionally displayed antihyperalgesic effects without observable motor impairment for treatment of neuropathic muscle pain induced by another chemotherapeutic agent.5 The effect was time-dependent, with TTX administration 4 hours after oxaliplatin injection producing stronger analgesic effects than administration 15 days after oxaliplatin injection.5 The analgesic effect lasted longer in the 4 hours post-oxaliplatin injection group as well, remaining significant after 2 hours.5 In another model of neuropathic pain, CCI of 15

ACCEPTED MANUSCRIPT

the sciatic and the infraorbital nerve produced mechanical hyperalgesia and allodynia that was attenuated at an optimal dose of 3 µg/kg of subcutaneous TTX in both acute and long term treatment patterns.74 At this and higher dosages of TTX there were no observable adverse effects and no impaired motor

RI PT

performance as evidenced by a rotarod test. This more recent study also indicated that its effect in CCIsciatic nerve rats may involve interaction with the opioid pathway; interestingly, antagonization of the opioid pathway appears to augment TTX’s effects.74 For a comprehensive review of TTX and its impact

SC

on neuropathic and other forms of pain, refer to Nieto et al’s review article.119

Importantly, TTX has already moved through a significant amount of clinical testing. Randomized, double-blind, placebo-controlled studies and longitudinal studies have been conducted on

M AN U

patients with cancer pain.61, 62 For example, one study demonstrated a clinically important use that TTX may provide in the form of analgesia for patients who have persistent moderate to severe cancer pain despite best analgesic care. This study has shown that the mean duration of analgesic response to TTX was 56.7 days compared to 9.9 days for the placebo. Some adverse effects were associated with TTX

TE D

including nausea, dizziness, and oral numbness or tingling, however, there were generally mild to moderate and transient.63 Most common adverse events were nausea, dizziness, and oral numbness or tingling and were generally mild to moderate and transient. Overall, the effects have been promising with

EP

a number of patients reporting analgesic effects, although further study is needed. In addition to completion of Phase III cancer pain trials, TTX has already completed Phase II clinical trials for

AC C

chemotherapy-induced neuropathic pain and is proceeding to Phase III. Thus, of the potential animal toxins and venoms for neuropathic pain management discussed herein, tetrodotoxin is currently the closest to realization.

ProTx-II VGSC Nav1.7 is of particular interest due to evidence regarding its significance in regulating neuropathic pain, alongside a range of human pain disorders.45, 137, 180 Loss of function mutations, for instance, in the gene encoding Nav1.7 can result in diminished pain perception in humans.45 16

ACCEPTED MANUSCRIPT

ProTx-II, a peptide of the inhibitory cysteine knot family isolated from venom of the tarantula Thrixopelma pruriens, binds with strong affinity to voltage-gating domains on sodium channels and Nav1.7 in particular, inhibiting the channel through increase of voltage activation threshold.147, 174, 175

RI PT

Recent evidence has shown that the toxin may do so through a lipid membrane binding mechanism, in which hydrophobic residues interact with the lipid membrane to position the toxin for interaction with its binding site on the channel.66 Its 100-fold selectivity for Nav1.7 over other channels makes it a likely target for development of analgesics; indeed, it has been tested for its ability to regulate acute

SC

inflammatory pain and neuropathic pain.147, 159 Intravenous ProTx-II failed to show any significant

M AN U

increase in paw withdrawal threshold in rats subjected to injections of Complete Freund’s Adjuvant (CFA) for a model of acute inflammatory pain. These rats presented with mechanical allodynia, which was slightly but insignificantly attenuated by the toxin.147 The toxin found more success in alleviating diabetes-induced thermal hyperalgesia in a neuropathic pain mouse model. Following diabetic induction via intravenous injection of streptozotocin, intrathecal administration of increasing doses of ProTx-II

TE D

(0.04 – 4 ng) resulted in a dose-dependent relationship with tail flick latency, indicating an increase in pain threshold that peaked 60 minutes after administration and subsided by 180 minutes.159 This implicates the Nav1.7 channel with thermal hyperalgesia in diabetic mice. However, there was no

EP

mention of side effects, and western blotting illustrated no significant difference in dorsal root ganglion Nav1.7 channels in diabetic versus non-diabetic mice. As damage to the nervous system in neuropathic

AC C

pain has been shown to change expression and regulation of voltage-gated sodium channels,137 the full picture of the interaction between Nav1.7 and other sodium channels and pain is, once again, not fully understood. This leaves room for further analysis of ProTx-II and other VGSC-blocking toxins for insight into neuropathic pain.

µ-theraphotoxin-Hhn1b (HNTX-IV) and µ-theraphotoxin-Hs2a (HWTX-IV) Two families of tarantula toxins, hainantoxins and huwentoxins, present additional potential for use in therapeutic treatment of neuropathic pain. µ-TRTX-Hhn1b (HNTX-IV) is a peptide isolated from 17

ACCEPTED MANUSCRIPT

the venom of Haplopelma hainanum and HWTX-IV is a peptide isolated from the venom of Haplopelma schmidti.170 Much like ProTx-II and other spider toxins,15, 78 HNTX-IV20, 172 and HWTX-IV39, 95, 173, 174 act as inhibitors of the TTX-sensitive VGSC Nav1.7, among others.

RI PT

HNTX-IV significantly reduced neuropathic pain in a rat model.92 The study utilized a spared nerve injury (SNI) model of neuropathic pain. An alternative to CCI, SNI calls for exposure of the sciatic nerve and its three endings but ligation of only two out of the three endings.38 SNI-induced mechanical

SC

allodynia underwent dose-dependent attenuation following intraperitoneal injection of HNTX-IV. The pain-alleviating effect peaked at approximately 60 minutes and lasted beyond 100 minutes with the

M AN U

highest dose of 0.2 mg/kg, in contrast with a dose of 40 mg/kg of mexiletine, a VGSC blocker positive control whose effect became insignificant at 60 minutes. Side effects including inactivity and paralysis were evident in rats given high doses (0.2 mg/kg) of HNTX-IV shown by rotarod.92 HWTX-IV dose dependently produced a longer and more potent pain alleviating effect on allodynia than mexiletine in a spinal nerve model.93 Moreover, HNTX-IV and HWTX-IV both produced significant dose-dependent

TE D

effects on hyperalgesia in inflammatory rat models induced by acetic acid and formalin at a potency comparable to morphine. Rats with spinal nerve injury-induced pain treated with HNTX-IV had a significant increase in withdrawal threshold, 8.8 ± 0.9 g, compared to saline treated rats, 1.4 ± 0.4 g.92, 93

EP

Overall, these two peptides are candidates for further analysis of their effects on alleviating neuropathic pain.

AC C

As there are other hainantoxins85, 96, 176 and huwentoxins165, 166 that show selective inhibition of VGSCs, future research warrants investigation into these and other spider toxins as potential therapeutics.142

µ-SLPTX-Ssm6a Other ion channel blockers include a variety of centipede venoms, whose analgesic and antimicrobial properties pose potential therapeutic benefits.64, 161 Several venoms have been isolated from the Chinese red-headed centipede Scolopendra subspinipes mutilans,178 one of which has a significant 18

ACCEPTED MANUSCRIPT

effect in relieving pain.179 µ-SLPTX-Ssm6a, a potent and highly selective inhibitor of human Nav1.7, significantly alleviates pain in chemically-induced models of inflammatory pain and thermal pain after intraperitoneal injection without notable adverse effects. This effect was observed in a dose dependent

RI PT

relationship and was comparable to morphine in the thermal pain and acetic acid-induced pain models, being even more potent than morphine in the formalin-induced inflammatory pain model.179 However, other studies have shown that the µ-SLPTX-Ssm6a peptide is not a strong binder of Nav1.7, which calls into question the true analgesic mechanism.111 Although µ-SLPTX-Ssm6a has not yet been tested in

SC

neuropathic pain models, its selectivity, relative stability,179 and potential analgesic properties make it a

M AN U

promising therapeutic candidate and likely subject of future experimentation.

GpTx-1

TE D

Venom of Grammostola porteri, the Chilean tarantula, contains GpTx-1. Study of its analogues illustrate an ability to inhibit Nav1.7.110-112 In the only study of its effect on pain, intraplantar coadministration of GpTx-1 with OD1, a scorpion-derived toxin exhibiting delayed inactivation of

EP

Nav1.7, significantly reduced spontaneous pain behavior characterized by control OD1 in a dosedependent manner.40 However, this novel pain model has not been characterized as neuropathic, and

AC C

GpTx-1 failed to produce an effect when injected intraperitoneally at 0.1 mg/kg. Higher doses could not be administered as they resulted in significant motor impairment.40

Anntoxin

The first gene-coded amphibian neurotoxin, anntoxin was isolated from the tree frog Hyla annectans and was shown to inhibit TTX-sensitive VGSCs.186 Anntoxin has antinociceptive effects in models of inflammatory and thermal pain, through a potential anti-inflammatory mechanism.167 Although

19

ACCEPTED MANUSCRIPT

no experiments on animal models of neuropathic pain have yet been conducted, anntoxin is a new

ACID-SENSING ION CHANNEL INHIBITORS PcTx1 and Mambalgins

RI PT

candidate that should be examined in future research.

SC

A more recently studied player in the field of pain is the acid-sensing ion channel (ASIC), whose isoforms (ASIC1a, ASIC1b, ASIC2a, ASIC2b, ASIC3, ASIC4) associate to form homomeric and heteromeric combinations of cation channels in peripheral and central neurons.43 Several studies have

M AN U

arisen to implicate ASICs in the treatment of various forms of pain,43, 105 including postoperative,42 acidic, and primary inflammatory pains.41 Animal toxin agonists of ASIC channels even demonstrate a paininducing effect.13 This evidence has created an impetus for further investigations into its pharmacological benefits.105

TE D

In the context of neuropathic pain, the realm of ASIC inhibitors provides two candidates: PcTx1, a peptide toxin isolated from the venom of Psalmopoeus cambridgei,170 and the mambalgins, a series of peptides (1-3) found in the venom of the black mamba Dendroaspis polylepis.48 Both of these toxins have

EP

been investigated as ASIC inhibitors to examine their potential benefit to therapeutic analgesia.9 The former, PcTx1, was the first inhibitor of ASIC to show high affinity and selectivity for the channel,

AC C

inhibiting ASIC1a and heteromeric combinations of ASIC1a/ASIC2a and ASIC1a/ASIC2b.54, 69 The peptide’s structure and character enable highly sensitive interactions with ASICs,28, 143 and the toxin inhibits ASIC1a through a desensitization mechanism that involves increasing its H+ affinity.27 Intrathecal injection of PcTx1 significantly reduced thermal hyperalgesia and mechanical allodynia in a CCI neuropathic mouse model and thermal and mechanical hyperalgesia in a vincristine (anticancer drug associated with neuropathy) rat model.104, 123 The effects were comparable to morphine and persisted at an hour after injection. The peptide is neither lethal nor causative of secondary effects associated with morphine. As for a potential mechanism of action, a model of thermally-induced acute pain was used to 20

ACCEPTED MANUSCRIPT

elucidate a reliance on the endogenous opioids enkephalins as well as their associated receptors, the µand δ-opioid receptors, with the ASIC1a channel as a potential intermediary.104 However, as the mechanistic findings were not presented in a model of neuropathic pain, it is difficult to assert likewise

RI PT

association of the opioid pathway. These findings present alongside additional comparable analgesic effects to morphine in the areas of thermally and chemically-induced acute and inflammatory pain.104 The ASIC inhibitors mambalgins 1 and 2 have shown promise in the treatment of neuropathic pain and have undergone research in analyzing their binding to ASIC1a as well as in developing an

SC

efficient synthetic procedure.48, 108, 131, 144, 149 Intravenous and intrathecal injections of mambalgin-1

M AN U

demonstrated significant attenuation of mechanical and thermal hyperalgesia in a CCI mouse model.49 The hyperalgesia-reducing effect of intravenous mambalgin-1 diminished earlier than that of intrathecal administration and results indicated that the mechanism of intravenous mambalgin-1 is independent of both ASIC1a and the opioid pathway. Both ASIC1a-KO mice and those pretreated with naloxone, an opioid antagonist, showed no respective significant differences from treatment with mambalgin-1 alone,

TE D

alluding to a mechanistic pathway involving other types of ASICs. However, intrathecal administration of mambalgin-1 produced longer-lasting effects than intravenous mambalgin-1 but revealed naloxonesensitivity as well as total lack of function in ASIC1a-KO mice. No apparent toxicity or motor problems

EP

were observed with the mambalgin-treated mice.49 In addition to indicating the analgesic potential o fmambalgin-1, these results also suggest a significant variability in ASIC expression and role in different

AC C

locations and in different pain pathways, elucidating an underexplored area for further research into ASICS in transmission of neuropathic and other forms of pain. APETx2 is another notable ASIC inhibitor that comes from Anthropleura elegantissima, a sea

anemone. This inhibitor blocks both ASIC3 and TTX-R Nav1.8 channels,12, 47 and evidence shows dose dependent attenuation of pain through intramuscular and intraplantar routes of injection in acid-induced muscle pain and CFA-induced inflammatory pain models, respectively.73 No studies on APETx2 and its action on neuropathic pain have yet been conducted.

21

ACCEPTED MANUSCRIPT

MECHANOSENSITIVE ION CHANNEL INHIBITOR

RI PT

GsMTx4 GsMTx4, a toxin from the Chilean rose tarantula Grammostola rosea, is the only known inhibitor of mechanosensitive ion channels.16 In a therapeutic context, these channels have mainly been studied for

SC

their involvement in conditions such as cardiac arrhythmias and muscular dystrophy.10, 16, 157 Despite results indicating that GsMTx4 is ineffective in reducing mechanically-activated currents in cultured DRG neurons expressing stretch-activated ion channels, analgesic effects have been evidenced in vivo and

M AN U

implicate the stretch activated ion channels TRPV4, TRPC1, and TRPC6.3, 50, 133 The resulting decrease in mechanical hyperalgesia was only tested using TRPV4-deficient mouse models that had pain induced by inflammatory soup.3 Research has revealed this toxin to be effective in reducing mechanical allodynia in a sciatic nerve injury rat model through intraperitoneal injection, indicating a potential inhibition of

TE D

mechanosensitive ion channels.133 Moreover, in a paclitaxel chemotherapy-induced neuropathic pain model, intradermal GsMTx4 reversed mechanical hyperalgesia after single injection, but this effect did not persist after 24 hours.3 It is important to note that GsMTx4 has activity on a variety of channels

EP

involved in pain (Kv and Piezo1) which may contribute to its analgesic effects.8, 120 Future research on this toxin and its ability to induce analgesia in the context of mechanosensitive receptors should be

AC C

explored.

OPIOID PATHWAY Crotalphine

Crotalphine, a small 14-amino acid long peptide, is a venom isolated from Crotalus durissus terrificus that has been shown to provide significant analgesic effects in models of inflammatory, neuropathic, and cancer pain.19, 59, 60, 80 Recent studies of crotalphine have primarily focused on deducing 22

ACCEPTED MANUSCRIPT

the pathway by which the toxin takes its effect. These studies have implicated a joint cannabinoid-opioid pathway, in which peripheral pain is alleviated through activation of the CB2 cannabinoid receptor by crotalphine and release of endogenous opioid A-dynorphin, which acts on the κ-opioid receptor.100, 187

RI PT

Additionally, crotalphine was found to desensitize TRPA1 channels and reduced chemically-induced inflammatory hyperalgesia in mouse models.18 This desensitizing effect disappeared in TRPA-1 deficient mice. These mechanistic results were obtained through models of inflammatory pain, although κ and δ-

SC

opioid receptor antagonists did inhibit crotalphine’s effect in CCI rats.60 In terms of its effect on

neuropathic pain itself, CCI rat models illustrated crotalphine’s analgesic effect in dose dependently

M AN U

reducing mechanical hyperalgesia and mechanical allodynia after oral administration for more than 3 days. Pain threshold was increased to above pre-surgery levels in tests of mechanical hyperalgesia, and crotalphine proved to be more effective than both morphine and gabapentin in alleviating pain.59 A similar effect was found in CCI rats, in which both intraplantar and oral crotalphine significantly reduced mechanical hyperalgesia and allodynia.60 Overall, there was no significant effect on motor activity after

TE D

oral administration of crotalphine.59 Moreover, oral administration of crotalphine every 3 days for 75 days and evaluating its analgesic effect 60 minutes after each administration did not reveal any tolerance development.59 This suggests a need for further studies investigating its uses in neuropathic pain.

EP

The ability of this isolated animal toxin to decrease neuropathic pain even when administered orally makes it unique. Stability to this degree has not been previously observed from a peptide toxin

AC C

given orally, and the mechanism by which crotalphine functions when administered orally has not been determined.80 Non-central routes of administration that effectively alleviate neuropathic pain provide new opportunities to advance future pain therapies.

Najanalgesin Pain reduction resulting from the najanalgesin toxin peptide, isolated from the venom of the Chinese cobra Naja naja atra, demonstrates a distinctive mechanism of action.68 The toxin may produce its analgesic effects through a glial cell related pathway reminiscent of diluted bee venom, but it had 23

ACCEPTED MANUSCRIPT

initially been shown to involve the opioid and cholinergic pathways.68, 87, 88, 91 Recent evidence of najanalgesin toxin peptide’s mechanism of action indicates that it may occur through the c-Jun N-terminal kinase mitogen-activated protein kinase (MAPK) pathway, one of the several MAPK pathways activated

RI PT

in response to nerve damage.55, 89 Intrathecal and intraperitoneal injections of najanalgesin produced significant antinociceptive effects in an SNL rat model.87, 88 The intraperitoneal experiments showed a dose dependent analgesic effect on mechanical allodynia lasting for over 10 days after a single injection,

SC

which persisted longer than the effects of morphine. The reduction on thermal allodynia, however, was not dose dependent. Intrathecal injections of najanalgesin produced significant dose dependent alleviation of mechanical allodynia, but for a shorter duration than intraperitoneal injection; this effect was not

M AN U

compared to morphine.88 Intrathecally administered najanalgesin also reduced SNL-induced hyperalgesia.87 Continued research on najanalgesin and its exact mechanism of action could further

TE D

support its candidacy as a target of neuropathic pain treatment.

NOREPINEPHRINE TRANSPORTER INHIBITOR Xen2174

EP

Isolated from the marbled cone, Conus marmoreus, Xen2174 is a more stable analog of the cconotoxin MrIA. It is a noncompetitive inhibitor of the norepinephrine transporter (NET) and thus affects

AC C

the noradrenergic pathway.151 Importantly, it was shown to have potential in neuropathic pain treatment through intrathecal injections in CCI rat models. Xen2174 dose dependently alleviated mechanical allodynia with mild side effects and had a longer duration of effect than morphine.117 These and other findings have propelled Xen2174 into clinical trials for postoperative and cancer pain, and there is promise for its use in neuropathic pain.84, 124 However, Xen2174 has halted its clinical development in 2012. The invasive administration of Xen2174 for examining its therapeutic effects further complicates its use in clinical trials.

24

ACCEPTED MANUSCRIPT

DISCUSSION Numerous potential avenues for isolated toxin peptides exist for alleviating neuropathic pain but

RI PT

require further exploration in terms of safety and efficacy (see supplementary Table 1). There are common themes among the toxin peptides in terms of mechanism of action with many concentrating on VGSCs (see supplementary Table 2) and VGCCs (see supplementary Table 3).83, 189 This is expected,

SC

as ziconotide, currently the only synthetically produced and FDA-approved prescription toxin peptide for pain relief, is a VGCC inhibitor. Nav1.1-1.19 are VGSCs believed to contribute to neuropathic pain.

M AN U

However, the data has not been pharmacologically tested in humans, which limits the ability to draw conclusions regarding their role in neuropathic pain. In the literature, Nav1.7 stands out as the foremost target of the VGSC inhibitors. All the VGSC-inhibiting toxins discussed act on TTX-sensitive VGSCs, and the toxins TTX, ProTx-II, HNTX-IV, HWTX-IV, µ-SLPTX-Ssm6a, and GpTx-1 all possess painreducing effects that may arise through their high selectivity for Nav1.7. Although the only FDA-

TE D

approved animal toxin for neuropathic pain management is the VGCC inhibitor ziconotide, the research on other VGCC inhibitors is surprisingly less focused than that on sodium channels. Spider venoms SNX482, Pha1b, Tx3-3, and Tx5-5 act on VGCCs to produce their cited effects on neuropathic pain, but only

EP

when administered intrathecally. Unfortunately, intrathecal injection proves far too invasive to be done routinely and an alternative procedure or route of administration would need to be sought out for these

AC C

toxin peptides. If a more favorable method of introducing the peptide could be demonstrated, then it appears that the isolated spider venom peptides would have an improved side effect profile when compared to other frequently used drugs. Though these toxin peptides hold promise in treating chronic pain, few of these molecules have been formally tested on humans and not all underwent testing for side effects in animal models (e.g. rotarod test, open field test). There exists high variability in the pathways that could be used to alleviate neuropathic pain. In addition to action upon the classic VGCCs, VGSCs, and opioid receptor-mediated pathways, the potential candidacies of nAChRs and NETs (inhibited by α-conotoxins Vc1.1/RgIA and c-conotoxin MrIA, 25

ACCEPTED MANUSCRIPT

respectively) in neuropathic pain sensation warrants a re-evaluation of these pathways. In fact, recent findings have established ASICs and MSICs as putative modulators of neuropathic pain sensation. Neuropathic pain is understood as a complex phenomenon that likely has as many methods of alleviation

field of venoms and toxins that may be examined in the future.

RI PT

as sources of induction. The scope of species touched upon in this review can attest to the unexplored

The copious number of pathways through which neuropathic pain can be relieved is further

SC

augmented by the variety of administration methods recognized in rat models. Although the majority of treatments showing attenuation of neuropathic pain occur through the intrathecal route, some studies

M AN U

show success using less invasive procedures. The α-conotoxins Vc1.1 and RgIA as well as the aOconotoxins GeXIVA [1,2] and [1,4] produce their effects through intramuscular administration; the blowfish toxin TTX reduced abnormal neuronal activity intravenously and pain subcutaneously and intramuscularly; DBV’s effects were observed using the subcutaneous method; the tarantula toxin HNTXIV was intraperitoneally injected; mambalgin-1 was intravenously administered; the tarantula toxin

TE D

GsMTx1 was effective through both intraperitoneal and intradermal injections; the cobra toxin najanalgesin was intraperitoneal; crotoxin was subcutaneously administered; and GpTx-1 and crotalphine were both effective after intraplantar injection. Crotalphine’s stability imbued it with effectiveness

EP

through an oral route of administration in spite of its status as a peptide, the only tested toxin peptide to demonstrate this unique characteristic. The significance of this finding should not be lost on the ongoing

AC C

search for a less invasive method of neuropathic pain relief, a leap to the ideal route of drug administration. Nonetheless, the other methods of injection remain improvements on the central route. Further research on administration methods, as well as on prolonging the stability of these toxins, is needed.

CONCLUSION Overall, it seems that there is a high potential among the venoms and toxin peptides discussed 26

ACCEPTED MANUSCRIPT

herein for a few to be developed into therapeutic entities. Many show positive effects in significantly attenuating or reversing neuropathic pain in an array of animal models but need further testing. Researchers in these experiments employ a variety of administration methods, which show promise in

RI PT

improving treatment procedures that move away from invasive practices. Adverse side effects were either not observed or limited for a large portion of the tested compounds. More intensive research into these toxins’ impact on animals, as well as optimization of administration methods and side effect profiles, may

SC

likely advance some compounds to consideration for clinical trials. Neuropathic pain, albeit a complex and largely invisible inducer of suffering, is a condition that has the potential to be fought with the advent

AC C

EP

TE D

M AN U

of new and more effective drugs derived from naturally occurring venoms.

27

ACCEPTED MANUSCRIPT

References:

1. Abdulsalam MA, Ebrahim BE Abdulsalam AJ: Immune thrombocytopenia after bee venom therapy: a case report. BMC Complement Altern Med 16 :107, 2016

RI PT

2. Akbayram S, Akgun C, Dogan M, Caksen H Oner AF: Acute ITP due to insect bite: report of 2 cases. Clin Appl Thromb Hemost 17 4:408-409, 2011 3. Alessandri-Haber N, Dina OA, Chen X Levine JD: TRPC1 and TRPC6 channels cooperate with TRPV4 to mediate mechanical hyperalgesia and nociceptor sensitization. J Neurosci 29 19:6217-6228, 2009

SC

4. Alford DP, Liebschutz J, Chen IA, Nicolaidis C, Panda M, Berg KM, Gibson J, Picchioni M Bair MJ: Update in pain medicine. J Gen Intern Med 23 6:841-845, 2008

M AN U

5. Alvarez P Levine JD: Antihyperalgesic effect of tetrodotoxin in rat models of persistent muscle pain. Neuroscience 311 :499-507, 2015 6. Arroyo G, Aldea M, Fuentealba J, Albillos A Garcia AG: SNX482 selectively blocks P/Q Ca2+ channels and delays the inactivation of Na+ channels of chromaffin cells. Eur J Pharmacol 475 1-3:11-18, 2003

TE D

7. Azam L, Papakyriakou A, Zouridakis M, Giastas P, Tzartos SJ McIntosh JM: Molecular interaction of alpha-conotoxin RgIA with the rat alpha9alpha10 nicotinic acetylcholine receptor. Mol Pharmacol 87 5:855-864, 2015 8. Bae C, Sachs F Gottlieb PA: The mechanosensitive ion channel Piezo1 is inhibited by the peptide GsMTx4. 50 29:6295-6300, 2011

EP

9. Baron A, Diochot S, Salinas M, Deval E, Noel J Lingueglia E: Venom toxins in the exploration of molecular, physiological and pathophysiological functions of acid-sensing ion channels. Toxicon 75 :187204, 2013

AC C

10. Barrabes JA, Inserte J, Agullo L, Rodriguez-Sinovas A, Alburquerque-Bejar JJ Garcia-Dorado D: Effects of the Selective Stretch-Activated Channel Blocker GsMtx4 on Stretch-Induced Changes in Refractoriness in Isolated Rat Hearts and on Ventricular Premature Beats and Arrhythmias after Coronary Occlusion in Swine. PLoS One 10 5:e0125753, 2015 11. Berecki G, Motin L, Haythornthwaite A, Vink S, Bansal P, Drinkwater R, Wang CI, Moretta M, Lewis RJ, Alewood PF, Christie MJ Adams DJ: Analgesic ω-Conotoxins CVIE and CVIF Selectively and Voltage-Dependently Block Recombinant and Native N-Type Calcium Channels. 77 2:139-148, 2010 12. Blanchard MG, Rash LD Kellenberger S: Inhibition of voltage-gated Na(+) currents in sensory neurones by the sea anemone toxin APETx2. Br J Pharmacol 165 7:2167-2177, 2012 13. Bohlen CJ, Chesler AT, Sharif-Naeini R, Medzihradszky KF, Zhou S, King D, Sanchez EE, Burlingame AL, Basbaum AI Julius D: A heteromeric Texas coral snake toxin targets acid-sensing ion channels to produce pain. Nature 479 7373:410-414, 2011 28

ACCEPTED MANUSCRIPT

14. Boivie J, Leijon G Johansson I: Central post-stroke pain--a study of the mechanisms through analyses of the sensory abnormalities. Pain 37 2:173-185, 1989 15. Bosmans F Swartz KJ: Targeting voltage sensors in sodium channels with spider toxins. Trends Pharmacol Sci 31 4:175-182, 2010

RI PT

16. Bowman CL, Gottlieb PA, Suchyna TM, Murphy YK Sachs F: Mechanosensitive ion channels and the peptide inhibitor GsMTx-4: history, properties, mechanisms and pharmacology. Toxicon 49 2:249270, 2007 17. Breivik H, Collett B, Ventafridda V, Cohen R Gallacher D: Survey of chronic pain in Europe: prevalence, impact on daily life, and treatment. Eur J Pain 10 4:287-333, 2006

SC

18. Bressan E, Touska F, Vetter I, Kistner K, Kichko TI, Teixeira NB, Picolo G, Cury Y, Lewis RJ, Fischer M, Zimmermann K Reeh PW: Crotalphine desensitizes TRPA1 ion channels to alleviate inflammatory hyperalgesia. 157 11:2504, 2016

M AN U

19. Brigatte P, Konno K, Gutierrez VP, Sampaio SC, Zambelli VO, Picolo G, Curi R Cury Y: Peripheral kappa and delta opioid receptors are involved in the antinociceptive effect of crotalphine in a rat model of cancer pain. Pharmacol Biochem Behav 109 :1-7, 2013 20. Cai T, Luo J, Meng E, Ding J, Liang S, Wang S Liu Z: Mapping the interaction site for the tarantula toxin hainantoxin-IV (beta-TRTX-Hn2a) in the voltage sensor module of domain II of voltage-gated sodium channels. Peptides 68 :148-156, 2015

TE D

21. Callaghan B, Haythornthwaite A, Berecki G, Clark RJ, Craik DJ Adams DJ: Analgesic alphaconotoxins Vc1.1 and Rg1A inhibit N-type calcium channels in rat sensory neurons via GABAB receptor activation. J Neurosci 28 43:10943-10951, 2008 22. Callaghan B Adams DJ: Analgesic alpha-conotoxins Vc1.1 and RgIA inhibit N-type calcium channels in sensory neurons of alpha9 nicotinic receptor knockout mice. Channels (Austin) 4 1:51-54, 2010

EP

23. Cardoso FC, Pacifico LG, Carvalho DC, Victoria JM, Neves AL, Chavez-Olortegui C, Gomez MV Kalapothakis E: Molecular cloning and characterization of Phoneutria nigriventer toxins active on calcium channels. Toxicon 41 7:755-763, 2003

AC C

24. Castro-Junior CJ, Milano J, Souza AH, Silva JF, Rigo FK, Dalmolin G, Cordeiro MN, Richardson M, Barros AG, Gomez RS, Silva MA, Kushmerick C, Ferreira J Gomez MV: Phalpha1beta toxin prevents capsaicin-induced nociceptive behavior and mechanical hypersensitivity without acting on TRPV1 channels. Neuropharmacology 71 :237-246, 2013 25. Chen B Ji Y: Antihyperalgesia effect of BmK AS, a scorpion toxin, in rat by intraplantar injection. Brain Res 952 2:322-326, 2002 26. Chen J Lariviere WR: The nociceptive and anti-nociceptive effects of bee venom injection and therapy: a double-edged sword. Prog Neurobiol 92 2:151-183, 2010 27. Chen X, Kalbacher H Grunder S: The tarantula toxin psalmotoxin 1 inhibits acid-sensing ion channel (ASIC) 1a by increasing its apparent H+ affinity. J Gen Physiol 126 1:71-79, 2005 29

ACCEPTED MANUSCRIPT

28. Chen X, Kalbacher H Grunder S: Interaction of acid-sensing ion channel (ASIC) 1 with the tarantula toxin psalmotoxin 1 is state dependent. J Gen Physiol 127 3:267-276, 2006

RI PT

29. Chen ZX, Zhang HL, Gu ZL, Chen BW, Han R, Reid PF, Raymond LN Qin ZH: A long-form alphaneurotoxin from cobra venom produces potent opioid-independent analgesia. Acta Pharmacol Sin 27 4:402-408, 2006 30. Clark RJ, Fischer H, Nevin ST, Adams DJ Craik DJ: The synthesis, structural characterization, and receptor specificity of the alpha-conotoxin Vc1.1. J Biol Chem 281 32:23254-23263, 2006 31. Cummins TR, Sheets PL Waxman SG: The roles of sodium channels in nociception: Implications for mechanisms of pain. Pain 131 3:243-257, 2007

SC

32. Cuny H, de Faoite A, Huynh TG, Yasuda T, Berecki G Adams DJ: gamma-Aminobutyric acid type B (GABAB) receptor expression is needed for inhibition of N-type (Cav2.2) calcium channels by analgesic alpha-conotoxins. J Biol Chem 287 28:23948-23957, 2012

M AN U

33. Cura JE, Blanzaco DP, Brisson C, Cura MA, Cabrol R, Larrateguy L, Mendez C, Sechi JC, Silveira JS, Theiller E, de Roodt AR Vidal JC: Phase I and pharmacokinetics study of crotoxin (cytotoxic PLA(2), NSC-624244) in patients with advanced cancer. Clin Cancer Res 8 4:1033-1041, 2002 34. Dalmolin GD, Silva CR, Rigo FK, Gomes GM, Cordeiro Mdo N, Richardson M, Silva MA, Prado MA, Gomez MV Ferreira J: Antinociceptive effect of Brazilian armed spider venom toxin Tx3-3 in animal models of neuropathic pain. Pain 152 10:2224-2232, 2011

TE D

35. de Lima ME, Stankiewicz M, Hamon A, de Figueiredo SG, Cordeiro MN, Diniz CR, MartinEauclaire M Pelhate M: The toxin Tx4(6-1) from the spider Phoneutria nigriventer slows down Na(+) current inactivation in insect CNS via binding to receptor site 3. J Insect Physiol 48 1:53-61, 2002

EP

36. de Souza AH, Lima MC, Drewes CC, da Silva JF, Torres KC, Pereira EM, de Castro Junior, C. J., Vieira LB, Cordeiro MN, Richardson M, Gomez RS, Romano-Silva MA, Ferreira J Gomez MV: Antiallodynic effect and side effects of Phalpha1beta, a neurotoxin from the spider Phoneutria nigriventer: comparison with omega-conotoxin MVIIA and morphine. Toxicon 58 8:626-633, 2011

AC C

37. de Souza AH, Castro CJ,Jr., Rigo FK, de Oliveira SM, Gomez RS, Diniz DM, Borges MH, Cordeiro MN, Silva MA, Ferreira J Gomez MV: An evaluation of the antinociceptive effects of Phalpha1beta, a neurotoxin from the spider Phoneutria nigriventer, and omega-conotoxin MVIIA, a cone snail Conus magus toxin, in rat model of inflammatory and neuropathic pain. Cell Mol Neurobiol 33 1:59-67, 2013 38. Decosterd I Woolf CJ: Spared nerve injury: an animal model of persistent peripheral neuropathic pain. Pain 87 2:149-158, 2000 39. Deng M, Luo X, Jiang L, Chen H, Wang J, He H Liang S: Synthesis and biological characterization of synthetic analogs of Huwentoxin-IV (Mu-theraphotoxin-Hh2a), a neuronal tetrodotoxin-sensitive sodium channel inhibitor. Toxicon 71 :57-65, 2013 40. Deuis JR, Wingerd JS, Winter Z, Durek T, Dekan Z, Sousa SR, Zimmermann K, Hoffmann T, Weidner C, Nassar MA, Alewood PF, Lewis RJ Vetter I: Analgesic Effects of GpTx-1, PF-04856264 and CNV1014802 in a Mouse Model of NaV1.7-Mediated Pain. Toxins (Basel) 8 3, 2016 30

ACCEPTED MANUSCRIPT

41. Deval E, Noel J, Lay N, Alloui A, Diochot S, Friend V, Jodar M, Lazdunski M Lingueglia E: ASIC3, a sensor of acidic and primary inflammatory pain. EMBO J 27 22:3047-3055, 2008 42. Deval E, Noel J, Gasull X, Delaunay A, Alloui A, Friend V, Eschalier A, Lazdunski M Lingueglia E: Acid-sensing ion channels in postoperative pain. J Neurosci 31 16:6059-6066, 2011

RI PT

43. Deval E Lingueglia E: Acid-Sensing Ion Channels and nociception in the peripheral and central nervous systems. Neuropharmacology 94 :49-57, 2015

44. Di Cesare Mannelli L, Cinci L, Micheli L, Zanardelli M, Pacini A, McIntosh JM Ghelardini C: alphaconotoxin RgIA protects against the development of nerve injury-induced chronic pain and prevents both neuronal and glial derangement. Pain 155 10:1986-1995, 2014

SC

45. Dib-Hajj SD, Cummins TR, Black JA Waxman SG: From genes to pain: Na v 1.7 and human pain disorders. Trends Neurosci 30 11:555-563, 2007

M AN U

46. Diniz DM, de Souza AH, Pereira EM, da Silva JF, Rigo FK, Romano-Silva MA, Binda N, Castro CJ,Jr., Cordeiro MN, Ferreira J Gomez MV: Effects of the calcium channel blockers Phalpha1beta and omega-conotoxin MVIIA on capsaicin and acetic acid-induced visceral nociception in mice. Pharmacol Biochem Behav 126 :97-102, 2014 47. Diochot S, Baron A, Rash LD, Deval E, Escoubas P, Scarzello S, Salinas M Lazdunski M: A new sea anemone peptide, APETx2, inhibits ASIC3, a major acid-sensitive channel in sensory neurons. EMBO J 23 7:1516-1525, 2004

TE D

48. Diochot S, Baron A, Salinas M, Douguet D, Scarzello S, Dabert-Gay AS, Debayle D, Friend V, Alloui A, Lazdunski M Lingueglia E: Black mamba venom peptides target acid-sensing ion channels to abolish pain. Nature 490 7421:552-555, 2012

EP

49. Diochot S, Alloui A, Rodrigues P, Dauvois M, Friend V, Aissouni Y, Eschalier A, Lingueglia E Baron A: Analgesic effects of mambalgin peptide inhibitors of acid-sensing ion channels in inflammatory and neuropathic pain. Pain 157 3:552-559, 2016

AC C

50. Drew LJ, Rugiero F, Cesare P, Gale JE, Abrahamsen B, Bowden S, Heinzmann S, Robinson M, Brust A, Colless B, Lewis RJ Wood JN: High-threshold mechanosensitive ion channels blocked by a novel conopeptide mediate pressure-evoked pain. PLoS One 2 6:e515, 2007 51. Ellison M, Haberlandt C, Gomez-Casati ME, Watkins M, Elgoyhen AB, McIntosh JM Olivera BM: Alpha-RgIA: a novel conotoxin that specifically and potently blocks the alpha9alpha10 nAChR. Biochemistry 45 5:1511-1517, 2006 52. Ellison M, Feng ZP, Park AJ, Zhang X, Olivera BM, McIntosh JM Norton RS: Alpha-RgIA, a novel conotoxin that blocks the alpha9alpha10 nAChR: structure and identification of key receptor-binding residues. J Mol Biol 377 4:1216-1227, 2008 53. Emerich BL, Ferreira RC, Cordeiro MN, Borges MH, Pimenta AM, Figueiredo SG, Duarte ID de Lima ME: delta-Ctenitoxin-Pn1a, a Peptide from Phoneutria nigriventer Spider Venom, Shows Antinociceptive Effect Involving Opioid and Cannabinoid Systems, in Rats. Toxins (Basel) 8 4:106, 2016

31

ACCEPTED MANUSCRIPT

54. Escoubas P, De Weille JR, Lecoq A, Diochot S, Waldmann R, Champigny G, Moinier D, Menez A Lazdunski M: Isolation of a tarantula toxin specific for a class of proton-gated Na+ channels. J Biol Chem 275 33:25116-25121, 2000 55. Gao YJ Ji RR: Activation of JNK pathway in persistent pain. Neurosci Lett 437 3:180-183, 2008

RI PT

56. Garry EM, Delaney A, Anderson HA, Sirinathsinghji EC, Clapp RH, Martin WJ, Kinchington PR, Krah DL, Abbadie C Fleetwood-Walker SM: Varicella zoster virus induces neuropathic changes in rat dorsal root ganglia and behavioral reflex sensitisation that is attenuated by gabapentin or sodium channel blocking drugs. Pain 118 1-2:97-111, 2005

SC

57. Giessel AJ Sabatini BL: Boosting of synaptic potentials and spine Ca transients by the peptide toxin SNX-482 requires alpha-1E-encoded voltage-gated Ca channels. PLoS One 6 6:e20939, 2011

M AN U

58. Gong S, Liang Q, Zhu Q, Ding D, Yin Q, Tao J Jiang X: Nicotinic acetylcholine receptor alpha7 subunit is involved in the cobratoxin-induced antinociception in an animal model of neuropathic pain. Toxicon 93 :31-36, 2015 59. Gutierrez VP, Konno K, Chacur M, Sampaio SC, Picolo G, Brigatte P, Zambelli VO Cury Y: Crotalphine induces potent antinociception in neuropathic pain by acting at peripheral opioid receptors. Eur J Pharmacol 594 1-3:84-92, 2008 60. Gutierrez VP, Zambelli VO, Picolo G, Chacur M, Sampaio SC, Brigatte P, Konno K Cury Y: The peripheral L-arginine-nitric oxide-cyclic GMP pathway and ATP-sensitive K(+) channels are involved in the antinociceptive effect of crotalphine on neuropathic pain in rats. Behav Pharmacol 23 1:14-24, 2012

TE D

61. Hagen NA, Fisher KM, Lapointe B, du Souich P, Chary S, Moulin D, Sellers E, Ngoc AH Canadian Tetrodotoxin Study G: An open-label, multi-dose efficacy and safety study of intramuscular tetrodotoxin in patients with severe cancer-related pain. J Pain Symptom Manage 34 2:171-182, 2007

EP

62. Hagen NA, du Souich P, Lapointe B, Ong-Lam M, Dubuc B, Walde D, Love R, Ngoc AH Canadian Tetrodotoxin Study G: Tetrodotoxin for moderate to severe cancer pain: a randomized, double blind, parallel design multicenter study. J Pain Symptom Manage 35 4:420-429, 2008

AC C

63. Hagen NA, Cantin L, Constant J, Haller T, Blaise G, Ong-Lam M, du Souich P, Korz W Lapointe B: Tetrodotoxin for Moderate to Severe Cancer-Related Pain: A Multicentre, Randomized, Double-Blind, Placebo-Controlled, Parallel-Design Trial. Pain Res Manag 2017 :7212713, 2017 64. Hakim MA, Yang S Lai R: Centipede venoms and their components: resources for potential therapeutic applications. Toxins (Basel) 7 11:4832-4851, 2015 65. Hannon HE Atchison WD: Omega-conotoxins as experimental tools and therapeutics in pain management. Mar Drugs 11 3:680-699, 2013 66. Henriques ST, Deplazes E, Lawrence N, Cheneval O, Chaousis S, Inserra M, Thongyoo P, King GF, Mark AE, Vetter I, Craik DJ Schroeder CI: Interaction of Tarantula Venom Peptide ProTx-II with Lipid Membranes Is a Prerequisite for Its Inhibition of Human Voltage-gated Sodium Channel NaV1.7. J Biol Chem 291 33:17049-17065, 2016

32

ACCEPTED MANUSCRIPT

67. Huynh TG, Cuny H, Slesinger PA Adams DJ: Novel mechanism of voltage-gated N-type (Cav2.2) calcium channel inhibition revealed through alpha-conotoxin Vc1.1 activation of the GABA(B) receptor. Mol Pharmacol 87 2:240-250, 2015

RI PT

68. Jiang WJ, Liang YX, Han LP, Qiu PX, Yuan J Zhao SJ: Purification and characterization of a novel antinociceptive toxin from Cobra venom (Naja naja atra). Toxicon 52 5:638-646, 2008 69. Joeres N, Augustinowski K, Neuhof A, Assmann M Grunder S: Functional and pharmacological characterization of two different ASIC1a/2a heteromers reveals their sensitivity to the spider toxin PcTx1. Sci Rep 6 :27647, 2016

SC

70. Kang SY, Roh DH, Park JH, Lee HJ Lee JH: Activation of Spinal alpha2-Adrenoceptors Using Diluted Bee Venom Stimulation Reduces Cold Allodynia in Neuropathic Pain Rats. Evid Based Complement Alternat Med 2012 :784713, 2012

M AN U

71. Kang SY, Roh DH, Yoon SY, Moon JY, Kim HW, Lee HJ, Beitz AJ Lee JH: Repetitive treatment with diluted bee venom reduces neuropathic pain via potentiation of locus coeruleus noradrenergic neuronal activity and modulation of spinal NR1 phosphorylation in rats. J Pain 13 2:155-166, 2012 72. Kang SY, Roh DH, Choi JW, Ryu Y Lee JH: Repetitive Treatment with Diluted Bee Venom Attenuates the Induction of Below-Level Neuropathic Pain Behaviors in a Rat Spinal Cord Injury Model. Toxins (Basel) 7 7:2571-2585, 2015

TE D

73. Karczewski J, Spencer RH, Garsky VM, Liang A, Leitl MD, Cato MJ, Cook SP, Kane S Urban MO: Reversal of acid-induced and inflammatory pain by the selective ASIC3 inhibitor, APETx2. Br J Pharmacol 161 4:950-960, 2010 74. Kayser V, Viguier F, Ioannidi M, Bernard JF, Latremoliere A, Michot B, Vela JM, Buschmann H, Hamon M Bourgoin S: Differential anti-neuropathic pain effects of tetrodotoxin in sciatic nerve- versus infraorbital nerve-ligated rats--behavioral, pharmacological and immunohistochemical investigations. Neuropharmacology 58 2:474-487, 2010

EP

75. Kim W, Kim MJ, Go D, Min BI, Na HS Kim SK: Combined Effects of Bee Venom Acupuncture and Morphine on Oxaliplatin-Induced Neuropathic Pain in Mice. Toxins (Basel) 8 2:33, 2016

AC C

76. Kimm T Bean BP: Inhibition of A-type potassium current by the peptide toxin SNX-482. J Neurosci 34 28:9182-9189, 2014 77. Klimis H, Adams DJ, Callaghan B, Nevin S, Alewood PF, Vaughan CW, Mozar CA Christie MJ: A novel mechanism of inhibition of high-voltage activated calcium channels by alpha-conotoxins contributes to relief of nerve injury-induced neuropathic pain. Pain 152 2:259-266, 2011 78. Klint JK, Senff S, Rupasinghe DB, Er SY, Herzig V, Nicholson GM King GF: Spider-venom peptides that target voltage-gated sodium channels: pharmacological tools and potential therapeutic leads. Toxicon 60 4:478-491, 2012 79. Koh WU, Choi SS, Lee JH, Lee SH, Lee SK, Lee YK, Leem JG, Song JG Shin JW: Perineural pretreatment of bee venom attenuated the development of allodynia in the spinal nerve ligation injured neuropathic pain model; an experimental study. BMC Complement Altern Med 14 :431, 2014 33

ACCEPTED MANUSCRIPT

80. Konno K, Picolo G, Gutierrez VP, Brigatte P, Zambelli VO, Camargo AC Cury Y: Crotalphine, a novel potent analgesic peptide from the venom of the South American rattlesnake Crotalus durissus terrificus. Peptides 29 8:1293-1304, 2008

RI PT

81. Leao RM, Cruz JS, Diniz CR, Cordeiro MN Beirao PS: Inhibition of neuronal high-voltage activated calcium channels by the omega-phoneutria nigriventer Tx3-3 peptide toxin. Neuropharmacology 39 10:1756-1767, 2000 82. Lee JH, Li DX, Yoon H, Go D, Quan FS, Min BI Kim SK: Serotonergic mechanism of the relieving effect of bee venom acupuncture on oxaliplatin-induced neuropathic cold allodynia in rats. BMC Complement Altern Med 14 :471, 2014

SC

83. Lees G Shipton E: Voltage-gated sodium channels in nociception and their potential as targets for new drugs in treatment of chronic neuropathic pain. Curr Anaesth Crit Care 20 5-6:204-208, 2009

M AN U

84. Lewis RJ, Dutertre S, Vetter I Christie MJ: Conus venom peptide pharmacology. Pharmacol Rev 64 2:259-298, 2012 85. Li D, Xiao Y, Hu W, Xie J, Bosmans F, Tytgat J Liang S: Function and solution structure of hainantoxin-I, a novel insect sodium channel inhibitor from the Chinese bird spider Selenocosmia hainana. FEBS Lett 555 3:616-622, 2003 86. Li X, Hu Y, Wu Y, Huang Y, Yu S, Ding Q, Zhangsun D Luo S: Anti-hypersensitive effect of intramuscular administration of alphaO-conotoxin GeXIVA[1,2] and GeXIVA[1,4] in rats of neuropathic pain. Prog Neuropsychopharmacol Biol Psychiatry 66 :112-119, 2016

TE D

87. Liang Y, Jiang W, Zhang Z, Yu J, Tao L Zhao S: Behavioral and morphological evidence for the involvement of glial cells in the antinociceptive effect of najanalgesin in a rat neuropathic pain model. Biol Pharm Bull 35 6:850-854, 2012

EP

88. Liang YX, Jiang WJ, Han LP Zhao SJ: Peripheral and spinal antihyperalgesic activity of najanalgesin isolated from Naja naja atra in a rat experimental model of neuropathic pain. Neurosci Lett 460 3:191195, 2009

AC C

89. Liang YX, Zhang ZY Zhang R: Antinociceptive Effect of Najanalgesin from Naja Naja Atra in a Neuropathic Pain Model via Inhibition of c-Jun NH2-terminal Kinase. Chin Med J (Engl) 128 17:23402345, 2015 90. Lim BS, Moon HJ, Li DX, Gil M, Min JK, Lee G, Bae H, Kim SK Min BI: Effect of bee venom acupuncture on oxaliplatin-induced cold allodynia in rats. Evid Based Complement Alternat Med 2013 :369324, 2013 91. Lin Q, Jiang W, Liang Y, Han L, Zhang C Zhao S: [Influence of najanalgesin from Naja naja on GLT-1 in spinal cord of rat in neuropathic pain]. Zhongguo Zhong Yao Za Zhi 36 7:903-907, 2011 92. Liu Y, Tang J, Zhang Y, Xun X, Tang D, Peng D, Yi J, Liu Z Shi X: Synthesis and analgesic effects of mu-TRTX-Hhn1b on models of inflammatory and neuropathic pain. Toxins (Basel) 6 8:2363-2378, 2014

34

ACCEPTED MANUSCRIPT

93. Liu Y, Wu Z, Tang D, Xun X, Liu L, Li X, Nie D, Xiang Y, Yi J Yi J: Analgesic effects of Huwentoxin-IV on animal models of inflammatory and neuropathic pain. Protein Pept Lett 21 2:153-158, 2014

RI PT

94. Liu YL, Lin HM, Zou R, Wu JC, Han R, Raymond LN, Reid PF Qin ZH: Suppression of complete Freund's adjuvant-induced adjuvant arthritis by cobratoxin. Acta Pharmacol Sin 30 2:219-227, 2009 95. Liu Z, Dai J, Chen Z, Hu W, Xiao Y Liang S: Isolation and characterization of hainantoxin-IV, a novel antagonist of tetrodotoxin-sensitive sodium channels from the Chinese bird spider Selenocosmia hainana. Cell Mol Life Sci 60 5:972-978, 2003

SC

96. Liu Z, Cai T, Zhu Q, Deng M, Li J, Zhou X, Zhang F, Li D, Li J, Liu Y, Hu W Liang S: Structure and function of hainantoxin-III, a selective antagonist of neuronal tetrodotoxin-sensitive voltage-gated sodium channels isolated from the Chinese bird spider Ornithoctonus hainana. J Biol Chem 288 28:20392-20403, 2013

M AN U

97. Livett BG, Sandall DW, Keays D, Down J, Gayler KR, Satkunanathan N Khalil Z: Therapeutic applications of conotoxins that target the neuronal nicotinic acetylcholine receptor. Toxicon 48 7:810829, 2006 98. Luo S, Zhangsun D, Harvey PJ, Kaas Q, Wu Y, Zhu X, Hu Y, Li X, Tsetlin VI, Christensen S, Romero HK, McIntyre M, Dowell C, Baxter JC, Elmslie KS, Craik DJ McIntosh JM: Cloning, synthesis, and characterization of alphaO-conotoxin GeXIVA, a potent alpha9alpha10 nicotinic acetylcholine receptor antagonist. Proc Natl Acad Sci U S A 112 30:E4026-35, 2015

TE D

99. Lyu YS, Park SK, Chung K Chung JM: Low dose of tetrodotoxin reduces neuropathic pain behaviors in an animal model. Brain Res 871 1:98-103, 2000 100. Machado FC, Zambelli VO, Fernandes AC, Heimann AS, Cury Y Picolo G: Peripheral interactions between cannabinoid and opioid systems contribute to the antinociceptive effect of crotalphine. Br J Pharmacol 171 4:961-972, 2014

EP

101. Maciel IS, Azevedo VM, Pereira TC, Bogo MR, Souza AH, Gomez MV Campos MM: The spinal inhibition of N-type voltage-gated calcium channels selectively prevents scratching behavior in mice. Neuroscience 277 :794-805, 2014

AC C

102. Martin-Moutot N, Mansuelle P, Alcaraz G, Dos Santos RG, Cordeiro MN, De Lima ME, Seagar M Van Renterghem C: Phoneutria nigriventer toxin 1: a novel, state-dependent inhibitor of neuronal sodium channels that interacts with micro conotoxin binding sites. Mol Pharmacol 69 6:1931-1937, 2006 103. Matthews EA, Bee LA, Stephens GJ Dickenson AH: The Cav2.3 calcium channel antagonist SNX482 reduces dorsal horn neuronal responses in a rat model of chronic neuropathic pain. Eur J Neurosci 25 12:3561-3569, 2007 104. Mazzuca M, Heurteaux C, Alloui A, Diochot S, Baron A, Voilley N, Blondeau N, Escoubas P, Gelot A, Cupo A, Zimmer A, Zimmer AM, Eschalier A Lazdunski M: A tarantula peptide against pain via ASIC1a channels and opioid mechanisms. Nat Neurosci 10 8:943-945, 2007

35

ACCEPTED MANUSCRIPT

105. McEntire DM, Kirkpatrick DR, Dueck NP, Kerfeld MJ, Smith TA, Nelson TJ, Reisbig MD Agrawal DK: Pain transduction: a pharmacologic perspective. Expert Rev Clin Pharmacol 9 8:1069-1080, 2016

RI PT

106. Mitchell AA, Sapienza-Crawford AJ, Hanley KL, Lokey KJ, Wells L, McDowell GC,2nd StantonHicks M: Administering ziconotide and monitoring patients treated with ziconotide: expert opinions. Pain Manag Nurs 14 3:e84-94, 2013 107. Mohammadi S Christie MJ: alpha9-nicotinic acetylcholine receptors contribute to the maintenance of chronic mechanical hyperalgesia, but not thermal or mechanical allodynia. Mol Pain 10 :64, 2014

SC

108. Mourier G, Salinas M, Kessler P, Stura EA, Leblanc M, Tepshi L, Besson T, Diochot S, Baron A, Douguet D, Lingueglia E Servent D: Mambalgin-1 Pain-relieving Peptide, Stepwise Solid-phase Synthesis, Crystal Structure, and Functional Domain for Acid-sensing Ion Channel 1a Inhibition. J Biol Chem 291 6:2616-2629, 2016

M AN U

109. Murakami M, Nakagawasai O, Suzuki T, Mobarakeh I, Sakurada Y, Murata A, Yamadera F, Miyoshi I, Yanai K, Tan-No K, Sasano H, Tadano T Iijima T: Antinociceptive effect of different types of calcium channel inhibitors and the distribution of various calcium channel alpha 1 subunits in the dorsal horn of spinal cord in mice. Brain Res 1024 1-2:122-129, 2004 110. Murray JK, Biswas K, Holder JR, Zou A, Ligutti J, Liu D, Poppe L, Andrews KL, Lin FF, Meng SY, Moyer BD, McDonough SI Miranda LP: Sustained inhibition of the NaV1.7 sodium channel by engineered dimers of the domain II binding peptide GpTx-1. Bioorg Med Chem Lett 25 21:4866-4871, 2015

TE D

111. Murray JK, Ligutti J, Liu D, Zou A, Poppe L, Li H, Andrews KL, Moyer BD, McDonough SI, Favreau P, Stocklin R Miranda LP: Engineering potent and selective analogues of GpTx-1, a tarantula venom peptide antagonist of the Na(V)1.7 sodium channel. J Med Chem 58 5:2299-2314, 2015

EP

112. Murray JK, Long J, Zou A, Ligutti J, Andrews KL, Poppe L, Biswas K, Moyer BD, McDonough SI Miranda LP: Single Residue Substitutions That Confer Voltage-Gated Sodium Ion Channel Subtype Selectivity in the NaV1.7 Inhibitory Peptide GpTx-1. J Med Chem 59 6:2704-2717, 2016 113. Namdev R, Dutta SR Singh H: Acute immune thrombocytopenic purpura triggered by insect bite. J Indian Soc Pedod Prev Dent 27 1:58-61, 2009

AC C

114. Napier IA, Klimis H, Rycroft BK, Jin AH, Alewood PF, Motin L, Adams DJ Christie MJ: Intrathecal alpha-conotoxins Vc1.1, AuIB and MII acting on distinct nicotinic receptor subtypes reverse signs of neuropathic pain. Neuropharmacology 62 7:2202-2207, 2012 115. Nevin ST, Clark RJ, Klimis H, Christie MJ, Craik DJ Adams DJ: Are alpha9alpha10 nicotinic acetylcholine receptors a pain target for alpha-conotoxins? Mol Pharmacol 72 6:1406-1410, 2007 116. Newcomb R, Szoke B, Palma A, Wang G, Chen X, Hopkins W, Cong R, Miller J, Urge L, TarczyHornoch K, Loo JA, Dooley DJ, Nadasdi L, Tsien RW, Lemos J Miljanich G: Selective peptide antagonist of the class E calcium channel from the venom of the tarantula Hysterocrates gigas. Biochemistry 37 44:15353-15362, 1998

36

ACCEPTED MANUSCRIPT

117. Nielsen CK, Lewis RJ, Alewood D, Drinkwater R, Palant E, Patterson M, Yaksh TL, McCumber D Smith MT: Anti-allodynic efficacy of the chi-conopeptide, Xen2174, in rats with neuropathic pain. Pain 118 1-2:112-124, 2005

RI PT

118. Nieto FR, Entrena JM, Cendan CM, Pozo ED, Vela JM Baeyens JM: Tetrodotoxin inhibits the development and expression of neuropathic pain induced by paclitaxel in mice. Pain 137 3:520-531, 2008 119. Nieto FR, Cobos EJ, Tejada MA, Sanchez-Fernandez C, Gonzalez-Cano R Cendan CM: Tetrodotoxin (TTX) as a therapeutic agent for pain. Mar Drugs 10 2:281-305, 2012

SC

120. Nishizawa M Nishizawa K: Molecular Dynamics Simulations of a Stretch-Activated Channel Inhibitor GsMTx4 with Lipid Membranes: Two Binding Modes and Effects of Lipid Structure. 92 12:4233-4243, 2007

M AN U

121. Nogueira-Neto Fde, Amorim RL, Brigatte P, Picolo G, Ferreira WA,Jr., Gutierrez VP, Conceicao IM, Della-Casa MS, Takahira RK, Nicoletti JL Cury Y: The analgesic effect of crotoxin on neuropathic pain is mediated by central muscarinic receptors and 5-lipoxygenase-derived mediators. Pharmacol Biochem Behav 91 2:252-260, 2008 122. Novakovic SD, Tzoumaka E, McGivern JG, Haraguchi M, Sangameswaran L, Gogas KR, Eglen RM Hunter JC: Distribution of the tetrodotoxin-resistant sodium channel PN3 in rat sensory neurons in normal and neuropathic conditions. 18 6:2174-2187, 1998 123. Nozaki-Taguchi N, Chaplan SR, Higuera ES, Ajakwe RC Yaksh TL: Vincristine-induced allodynia in the rat. Pain 93 1:69-76, 2001

TE D

124. Obata H, Conklin D Eisenach JC: Spinal noradrenaline transporter inhibition by reboxetine and Xen2174 reduces tactile hypersensitivity after surgery in rats. Pain 113 3:271-276, 2005 125. Ochoa J, Torebjörk HE, Culp WJ Schady W: Abnormal spontaneous activity in single sensory nerve fibers in humans. 5 9:7, 1982

EP

126. O'Connor AB Dworkin RH: Treatment of neuropathic pain: an overview of recent guidelines. Am J Med 122 10:S22-32, 2009

AC C

127. Oliveira SM, Silva CR, Trevisan G, Villarinho JG, Cordeiro MN, Richardson M, Borges MH, Castro CJ,Jr., Gomez MV Ferreira J: Antinociceptive effect of a novel armed spider peptide Tx3-5 in pathological pain models in mice. Pflugers Arch 468 5:881-894, 2016 128. Omana-Zapata I, Khabbaz MA, Hunter JC, Clarke DE Bley KR: Tetrodotoxin inhibits neuropathic ectopic activity in neuromas, dorsal root ganglia and dorsal horn neurons. Pain 72 1-2:41-49, 1997 129. Pacini A, Micheli L, Maresca M, Branca JJ, McIntosh JM, Ghelardini C Di Cesare Mannelli L: The alpha9alpha10 nicotinic receptor antagonist alpha-conotoxin RgIA prevents neuropathic pain induced by oxaliplatin treatment. Exp Neurol 282 :37-48, 2016 130. Paiva AL, Matavel A, Peigneur S, Cordeiro MN, Tytgat J, Diniz MR de Lima ME: Differential effects of the recombinant toxin PnTx4(5-5) from the spider Phoneutria nigriventer on mammalian and insect sodium channels. Biochimie 121 :326-335, 2016 37

ACCEPTED MANUSCRIPT

131. Pan M, He Y, Wen M, Wu F, Sun D, Li S, Zhang L, Li Y Tian C: One-pot hydrazide-based native chemical ligation for efficient chemical synthesis and structure determination of toxin Mambalgin-1. Chem Commun (Camb) 50 44:5837-5839, 2014 132. Park J Luo ZD: Calcium channel functions in pain processing. Channels (Austin) 4 6:510-517, 2010

RI PT

133. Park SP, Kim BM, Koo JY, Cho H, Lee CH, Kim M, Na HS Oh U: A tarantula spider toxin, GsMTx4, reduces mechanical and neuropathic pain. Pain 137 1:208-217, 2008

134. Premkumar LS: Targeting TRPV1 as an alternative approach to narcotic analgesics to treat chronic pain conditions. AAPS J 12 3:361-370, 2010

SC

135. Rigo FK, Dalmolin GD, Trevisan G, Tonello R, Silva MA, Rossato MF, Klafke JZ, Cordeiro Mdo N, Castro Junior CJ, Montijo D, Gomez MV Ferreira J: Effect of omega-conotoxin MVIIA and Phalpha1beta on paclitaxel-induced acute and chronic pain. Pharmacol Biochem Behav 114-115 :16-22, 2013

M AN U

136. Rigo FK, Trevisan G, Rosa F, Dalmolin GD, Otuki MF, Cueto AP, de Castro Junior, C. J., RomanoSilva MA, Cordeiro Mdo N, Richardson M, Ferreira J Gomez MV: Spider peptide Phalpha1beta induces analgesic effect in a model of cancer pain. Cancer Sci 104 9:1226-1230, 2013 137. Rogers M, Tang L, Madge DJ Stevens EB: The role of sodium channels in neuropathic pain. Semin Cell Dev Biol 17 5:571-581, 2006

TE D

138. Roh DH, Kwon YB, Kim HW, Ham TW, Yoon SY, Kang SY, Han HJ, Lee HJ, Beitz AJ Lee JH: Acupoint stimulation with diluted bee venom (apipuncture) alleviates thermal hyperalgesia in a rodent neuropathic pain model: involvement of spinal alpha 2-adrenoceptors. J Pain 5 6:297-303, 2004

EP

139. Rosa F, Trevisan G, Rigo FK, Tonello R, Andrade EL, Cordeiro Mdo N, Calixto JB, Gomez MV Ferreira J: Phalpha1beta, a peptide from the venom of the spider Phoneutria nigriventer shows antinociceptive effects after continuous infusion in a neuropathic pain model in rats. Anesth Analg 119 1:196-202, 2014

AC C

140. Roy A, Zhou X, Chong MZ, D'Hoedt D, Foo CS, Rajagopalan N, Nirthanan S, Bertrand D, Sivaraman J Kini RM: Structural and functional characterization of a novel homodimeric three-finger neurotoxin from the venom of Ophiophagus hannah (king cobra). J Biol Chem 285 11:8302-8315, 2010 141. Sadeghi M, Murali SS, Lewis RJ, Alewood PF, Mohammadi S Christie MJ: Novel ω-conotoxins from C. catus reverse signs of mouse inflammatory pain after systemic administration. 9 1:1-9, 2013 142. Saez NJ, Senff S, Jensen JE, Er SY, Herzig V, Rash LD King GF: Spider-venom peptides as therapeutics. Toxins (Basel) 2 12:2851-2871, 2010 143. Salinas M, Rash LD, Baron A, Lambeau G, Escoubas P Lazdunski M: The receptor site of the spider toxin PcTx1 on the proton-gated cation channel ASIC1a. J Physiol 570 :339-354, 2006 144. Salinas M, Besson T, Delettre Q, Diochot S, Boulakirba S, Douguet D Lingueglia E: Binding site and inhibitory mechanism of the mambalgin-2 pain-relieving peptide on acid-sensing ion channel 1a. J Biol Chem 289 19:13363-13373, 2014 38

ACCEPTED MANUSCRIPT

145. Sandall DW, Satkunanathan N, Keays DA, Polidano MA, Liping X, Pham V, Down JG, Khalil Z, Livett BG Gayler KR: A novel alpha-conotoxin identified by gene sequencing is active in suppressing the vascular response to selective stimulation of sensory nerves in vivo. Biochemistry 42 22:6904-6911, 2003

RI PT

146. Satkunanathan N, Livett B, Gayler K, Sandall D, Down J Khalil Z: Alpha-conotoxin Vc1.1 alleviates neuropathic pain and accelerates functional recovery of injured neurones. Brain Res 1059 2:149-158, 2005 147. Schmalhofer WA, Calhoun J, Burrows R, Bailey T, Kohler MG, Weinglass AB, Kaczorowski GJ, Garcia ML, Koltzenburg M Priest BT: ProTx-II, a selective inhibitor of NaV1.7 sodium channels, blocks action potential propagation in nociceptors. Mol Pharmacol 74 5:1476-1484, 2008

SC

148. Scholz J Woolf CJ: Can we conquer pain? Nat Neurosci 5 Suppl :1062-1067, 2002

M AN U

149. Schroeder CI, Rash LD, Vila-Farres X, Rosengren KJ, Mobli M, King GF, Alewood PF, Craik DJ Durek T: Chemical synthesis, 3D structure, and ASIC binding site of the toxin mambalgin-2. Angew Chem Int Ed Engl 53 4:1017-1020, 2014 150. Seward E, Hammond C Henderson G: Mu-opioid-receptor-mediated inhibition of the N-type calcium-channel current. Proc Biol Sci 244 1310:129-135, 1991 151. Sharpe IA, Palant E, Schroeder CI, Kaye DM, Adams DJ, Alewood PF Lewis RJ: Inhibition of the norepinephrine transporter by the venom peptide chi-MrIA. Site of action, Na+ dependence, and structure-activity relationship. J Biol Chem 278 41:40317-40323, 2003

TE D

152. Shi GN, Liu YL, Lin HM, Yang SL, Feng YL, Reid PF Qin ZH: Involvement of cholinergic system in suppression of formalin-induced inflammatory pain by cobratoxin. Acta Pharmacol Sin 32 10:12331238, 2011

EP

153. Shin JS, Lee J, Kim MR, Jung J, Shin BC, Lee MS Ha IH: The Short-Term Effect of Integrated Complementary and Alternative Medicine Treatment in Inpatients Diagnosed with Lumbar Intervertebral Disc Herniation: A Prospective Observational Study. J Altern Complement Med 22 7:533-543, 2016 154. Silva AO, Peigneur S, Diniz MR, Tytgat J Beirao PS: Inhibitory effect of the recombinant Phoneutria nigriventer Tx1 toxin on voltage-gated sodium channels. Biochimie 94 12:2756-2763, 2012

AC C

155. Sobral F, Sampaio A, Falcao S, Queiroz MJ, Calhelha RC, Vilas-Boas M Ferreira IC: Chemical characterization, antioxidant, anti-inflammatory and cytotoxic properties of bee venom collected in Northeast Portugal. Food Chem Toxicol 94 :172-177, 2016 156. Son DJ, Lee JW, Lee YH, Song HS, Lee CK Hong JT: Therapeutic application of anti-arthritis, painreleasing, and anti-cancer effects of bee venom and its constituent compounds. Pharmacol Ther 115 2:246-270, 2007 157. Suchyna TM Sachs F: Mechanosensitive channel properties and membrane mechanics in mouse dystrophic myotubes. J Physiol 581 :369-387, 2007 158. Suehiro M: [Historical review on chemical and medical studies of globefish toxin before World War II]. Yakushigaku Zasshi 29 3:428-434, 1994 39

ACCEPTED MANUSCRIPT

159. Tanaka K, Sekino S, Ikegami M, Ikeda H Kamei J: Antihyperalgesic effects of ProTx-II, a Nav1.7 antagonist, and A803467, a Nav1.8 antagonist, in diabetic mice. J Exp Pharmacol 7 :11-16, 2015 160. Tanphaichitr VS Tuchinda M: Severe thrombocytopenic purpura following a bee sting. Ann Allergy 49 4:229-231, 1982

RI PT

161. Undheim EA, Fry BG King GF: Centipede venom: recent discoveries and current state of knowledge. Toxins (Basel) 7 3:679-704, 2015

162. Vieira LB, Kushmerick C, Hildebrand ME, Garcia E, Stea A, Cordeiro MN, Richardson M, Gomez MV Snutch TP: Inhibition of high voltage-activated calcium channels by spider toxin PnTx3-6. J Pharmacol Exp Ther 314 3:1370-1377, 2005

SC

163. Vincler M, Wittenauer S, Parker R, Ellison M, Olivera BM McIntosh JM: Molecular mechanism for analgesia involving specific antagonism of alpha9alpha10 nicotinic acetylcholine receptors. Proc Natl Acad Sci U S A 103 47:17880-17884, 2006

M AN U

164. Vincler M McIntosh JM: Targeting the alpha9alpha10 nicotinic acetylcholine receptor to treat severe pain. Expert Opin Ther Targets 11 7:891-897, 2007 165. Wang M, Guan X Liang S: The cross channel activities of spider neurotoxin huwentoxin-I on rat dorsal root ganglion neurons. Biochem Biophys Res Commun 357 3:579-583, 2007 166. Wang RL, Yi S Liang SP: Mechanism of action of two insect toxins huwentoxin-III and hainantoxin-VI on voltage-gated sodium channels. J Zhejiang Univ Sci B 11 6:451-457, 2010

TE D

167. Wei L, Dong L, Zhao T, You D, Liu R, Liu H, Yang H Lai R: Analgesic and anti-inflammatory effects of the amphibian neurotoxin, anntoxin. Biochimie 93 6:995-1000, 2011 168. Westenbroek RE, Hoskins L Catterall WA: Localization of Ca2+ channel subtypes on rat spinal motor neurons, interneurons, and nerve terminals. J Neurosci 18 16:6319-6330, 1998

EP

169. Wolz-Richter S, Esser KH Hess A: Antinociceptive activity of crotoxin in the central nervous system: a functional Magnetic Resonance Imaging study. Toxicon 74 :44-55, 2013

AC C

170. Wood DL, Miljenovic T, Cai S, Raven RJ, Kaas Q, Escoubas P, Herzig V, Wilson D King GF: ArachnoServer: a database of protein toxins from spiders. BMC Genomics 10 :375, 2009 171. Wright AB, Norimatsu Y, McIntosh JM Elmslie KS: Limited efficacy of alpha-conopeptides, Vc1.1 and RgIA, to inhibit sensory neuron CaV current. eNeuro 2 1, 2015 172. Xiao Y Liang S: Inhibition of neuronal tetrodotoxin-sensitive Na+ channels by two spider toxins: hainantoxin-III and hainantoxin-IV. Eur J Pharmacol 477 1:1-7, 2003 173. Xiao Y, Bingham JP, Zhu W, Moczydlowski E, Liang S Cummins TR: Tarantula huwentoxin-IV inhibits neuronal sodium channels by binding to receptor site 4 and trapping the domain ii voltage sensor in the closed configuration. J Biol Chem 283 40:27300-27313, 2008

40

ACCEPTED MANUSCRIPT

174. Xiao Y, Blumenthal K, Jackson JO,2nd, Liang S Cummins TR: The tarantula toxins ProTx-II and huwentoxin-IV differentially interact with human Nav1.7 voltage sensors to inhibit channel activation and inactivation. Mol Pharmacol 78 6:1124-1134, 2010

RI PT

175. Xiao Y, Blumenthal K Cummins TR: Gating-pore currents demonstrate selective and specific modulation of individual sodium channel voltage-sensors by biological toxins. Mol Pharmacol 86 2:159167, 2014 176. Xiao YC Liang SP: Purification and characterization of Hainantoxin-V, a tetrodotoxin-sensitive sodium channel inhibitor from the venom of the spider Selenocosmia hainana. Toxicon 41 6:643-650, 2003

SC

177. Yang L Stephens GJ: Effects of neuropathy on high-voltage-activated Ca(2+) current in sensory neurones. Cell Calcium 46 4:248-256, 2009

M AN U

178. Yang S, Liu Z, Xiao Y, Li Y, Rong M, Liang S, Zhang Z, Yu H, King GF Lai R: Chemical punch packed in venoms makes centipedes excellent predators. Mol Cell Proteomics 11 9:640-650, 2012 179. Yang S, Xiao Y, Kang D, Liu J, Li Y, Undheim EA, Klint JK, Rong M, Lai R King GF: Discovery of a selective NaV1.7 inhibitor from centipede venom with analgesic efficacy exceeding morphine in rodent pain models. Proc Natl Acad Sci U S A 110 43:17534-17539, 2013 180. Yang Y, Wang Y, Li S, Xu Z, Li H, Ma L, Fan J, Bu D, Liu B, Fan Z, Wu G, Jin J, Ding B, Zhu X Shen Y: Mutations in SCN9A, encoding a sodium channel alpha subunit, in patients with primary erythermalgia. J Med Genet 41 3:171-174, 2004

TE D

181. Yeo JH, Yoon SY, Kwon SK, Kim SJ, Lee JH, Beitz AJ Roh DH: Repetitive Acupuncture Point Treatment with Diluted Bee Venom Relieves Mechanical Allodynia and Restores Intraepidermal Nerve Fiber Loss in Oxaliplatin-Induced Neuropathic Mice. J Pain 17 3:298-309, 2016

EP

182. Yoon H, Kim MJ, Yoon I, Li DX, Bae H Kim SK: Nicotinic Acetylcholine Receptors Mediate the Suppressive Effect of an Injection of Diluted Bee Venom into the GV3 Acupoint on Oxaliplatin-Induced Neuropathic Cold Allodynia in Rats. Biol Pharm Bull 38 5:710-714, 2015

AC C

183. Yoon J, Jeon JH, Lee YW, Cho CK, Kwon KR, Shin JE, Sagar S, Wong R Yoo HS: Sweet bee venom pharmacopuncture for chemotherapy-induced peripheral neuropathy. J Acupunct Meridian Stud 5 4:156-165, 2012 184. Yoon SY, Roh DH, Kwon YB, Kim HW, Seo HS, Han HJ, Lee HJ, Beitz AJ Lee JH: Acupoint stimulation with diluted bee venom (apipuncture) potentiates the analgesic effect of intrathecal clonidine in the rodent formalin test and in a neuropathic pain model. J Pain 10 3:253-263, 2009 185. Yoon SY, Yeo JH, Han SD, Bong DJ, Oh B Roh DH: Diluted bee venom injection reduces ipsilateral mechanical allodynia in oxaliplatin-induced neuropathic mice. Biol Pharm Bull 36 11:17871793, 2013 186. You D, Hong J, Rong M, Yu H, Liang S, Ma Y, Yang H, Wu J, Lin D Lai R: The first gene-encoded amphibian neurotoxin. J Biol Chem 284 33:22079-22086, 2009

41

ACCEPTED MANUSCRIPT

187. Zambelli VO, Fernandes AC, Gutierrez VP, Ferreira JC, Parada CA, Mochly-Rosen D Cury Y: Peripheral sensitization increases opioid receptor expression and activation by crotalphine in rats. PLoS One 9 3:e90576, 2014

RI PT

188. Zamponi GW: Antiallodynic effects of a confused alpha-conotoxin: Vc1.1 relieves neuropathic pain via off target actions on GABA(B) receptors and N-type channels. Pain 152 2:241-242, 2011 189. Zamponi GW, Striessnig J, Koschak A Dolphin AC: The Physiology, Pathology, and Pharmacology of Voltage-Gated Calcium Channels and Their Future Therapeutic Potential. Pharmacol Rev 67 4:821870, 2015

AC C

EP

TE D

M AN U

SC

190. Zhang HL, Han R, Chen ZX, Chen BW, Gu ZL, Reid PF, Raymond LN Qin ZH: Opiate and acetylcholine-independent analgesic actions of crotoxin isolated from crotalus durissus terrificus venom. Toxicon 48 2:175-182, 2006

42

ACCEPTED MANUSCRIPT

Potential Uses of Isolated Toxin Peptides in Neuropathic Pain Relief: A Literature Review Highlights

EP

TE D

M AN U

SC

RI PT

Drugs derived from naturally occurring toxins may aid in treating neuropathic pain. Researching toxins’ impact on animals may lead to consideration for clinical trials. Ziconotide is an available toxin drug used for management of severe chronic pain. Many toxin peptides have only undergone animal studies. It is essential to continue research for safer alternatives to opioid analgesia.

AC C

• • • • •

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

Abbreviation List: ASIC - Acid-sensing ion channel Ca – Calcium CCI - chronic constriction injury cDNA - complementary Deoxyribonucleic acid CFA - Complete Freund’s Adjuvant DBV - Diluted bee venom DRG – Dorsal Root Ganglion FDA – Food and Drug Administration GABA - gamma-Aminobutyric acid GFAP - glial fibrillary acidic protein HNTX-IV - μ-TRTX-Hhn1b ITP - induced immune thrombocytopenia KO – Knock out MAPK - mitogen-activated protein kinase MWT - Mechanical Withdrawal Threshold MSIC – Mechanosensitive Ion Channels mRNA – Messenger Ribonucleic acid Na – Sodium nAChR – Nicotinic Acetylcholine Receptor NET - norepinephrine transporter NSAID – Nonsteroidal Anti-Inflammatory Drug PNL - partial nerve ligation SNI - spared nerve injury SNL - spinal nerve ligation SNRI –Serotonin Norepinephrine Reuptake Inhibitor TTX - Tetrodotoxin TWL - Thermal Withdrawal Latency US – United States V1 – Ophthalmic nerve V2 – Maxillary Nerve V3 – Mandibular nerve VGCC – Voltage gated calcium channel VGSCs - voltage-gated sodium channels μg/kg – Microgram per kilogram

SC

August 12, 2017

RI PT

Mousa K Hamad Albert Einstein College of Medicine Department of Neurosurgery 1300 Morris Park Ave Bronx, NY 1046 [email protected]

ACCEPTED MANUSCRIPT

Thank you for your consideration.

AC C

EP

TE D

M AN U

SC

RI PT

Very Respectfully, Mousa K Hamad, MD Corresponding Author Albert Einstein College of Medicine Department of Neurosurgery