Review

Expert opinion on emerging drugs: chronic low back pain

Expert Opin. Emerging Drugs Downloaded from informahealthcare.com by Selcuk Universitesi on 12/24/14 For personal use only.

Eugene Hsu, Sunberri Murphy, David Chang & Steven P Cohen† †

Johns Hopkins School of Medicine and Uniformed Services University of the Health Sciences, Bethesda, MD, USA

1.

Background

2.

Medical need

3.

Existing treatments

4.

Market review

5.

Current research goals

6.

Scientific rationale for mechanism-based treatment of LBP

7.

Competitive environment

8.

Potential development issues

9.

Conclusions

10.

Expert opinion

Introduction: It is difficult to overestimate the personal and socioeconomic impact of chronic low back pain (CLBP). It is the leading cause of years lost to disability and poses the highest economic toll among chronic illnesses. Despite the strong need for extensive research efforts, few drugs have consistently demonstrated effectiveness for this condition. Areas covered: In this review, the epidemiology, rationale for mechanismbased treatment, competitive environment and market trends, and the preclinical and clinical evidence supporting over 15 different classes of analgesic medications studied for CLBP or related pain conditions are discussed. Treatments are divided by drug category, type of CLBP they are likely to treat (e.g., neuropathic or mechanical), and whether they are new formulations of existing treatments, new indications for existing treatments or represent novel mechanisms of action. Databases searched included MEDLINE, Embase, Pharmaprojects and various clinical trial registries. Expert opinion: Many barriers exist for the development of medications for CLBP including difficulties in identifying pathophysiological mechanisms, biologic resiliency secondary to multiple concurrent pain pathways and offtarget and sometimes serious side effects. Nevertheless, the volume and diversity of novel molecular entities has continued to surge and includes possible disease-modifying therapies such as gene and stem cell therapy. Keywords: low back pain, mechanical back pain, mechanism-based treatment, pharmacotherapy, radiculopathy, sciatica Expert Opin. Emerging Drugs [Early Online]

1.

Background

One would be hard-pressed to overestimate the societal and economic tolls exacted by low back pain (LBP). LBP is the leading cause of disability in the world, with a lifetime prevalence rate ranging between 50 and 90% [1-3]. In the US alone, the annual cost of LBP by some estimates exceeds $100 billion, with over one-half stemming from lost productivity [3-5]. LBP is sometimes considered to be a ‘diagnosis’ but is more accurately defined as a symptom characterized by multiple and often overlapping mechanisms. One of the most common classification schemes distinguishes LBP as being either neuropathic (pain caused by a disease or lesion affecting the somatosensory system) or nociceptive/mechanical pain (which is caused by peripheral stimulation of painful nerve endings embedded in structures contained within or supporting the lumbar spinal column such as muscles, intervertebral discs, facet joints, etc.) [6,7]. Although we will use this mechanism-based definition for the purpose of this review, it should be noted that several authors have emphasized a broader definition for chronic LBP (CLBP). Waddell was one of the first to utilize the biopsychosocial model, which postulates that biological, psychological and social factors all play a role in disease, as an operational model to assess the impact of physical rehabilitation approaches for CLBP [8].

10.1517/14728214.2015.993379 © 2014 Informa UK, Ltd. ISSN 1472-8214, e-ISSN 1744-7623 All rights reserved: reproduction in whole or in part not permitted

1

Expert Opin. Emerging Drugs Downloaded from informahealthcare.com by Selcuk Universitesi on 12/24/14 For personal use only.

E. Hsu et al.

The treatment of pain, in general, and LBP, in particular, is notoriously challenging. Indeed, the effect sizes of most nonsurgical treatments for nonspecific LBP are low to moderate (0.2 -- 0.6) and unlikely to result in complete pain relief [9]. Therefore, pain management is aimed at reducing dysfunction in physical, psychological and social dimensions while improving quality of life for patients and minimizing risks and adverse effects. Studies have found a 30% pain reduction to constitute clinically meaningful benefit [10-13], but a decrease of as little as 10% compared to ‘placebo’ is what is often required for regulatory drug approval [14]. For acute LBP, the PACE study of paracetamol, a first-line analgesic, showed that scheduled or as needed paracetamol did not affect recovery time compared to placebo [15]. For neuropathic back pain, clinical trials evaluating first-line medications such as tricyclic antidepressants (TCAs) and gabapentinoids have generally found very small differences between treatment and control groups [16,17]. For CLBP, systematic reviews have found small effect sizes supporting NSAIDs, opioids and muscle relaxants, with mixed results for antidepressants [18-21]. The combination of the high prevalence rate and economic burden, manifold mechanisms and recalcitrance to treatment has led to an onslaught of research aimed at alleviating LBP. Whereas the purpose of this review is to evaluate emerging pharmacologic therapies for LBP based on clinical trials, the important role of physical, psychological and rehabilitation treatments cannot be understated [22]. 2.

Medical need

There is a strong need for new and novel treatments for CLBP, as existing treatments have shown very modest efficacy and tend to be characterized by significant side effects, risks and costs. Moreover, despite the multiple pharmacological and non-pharmacological treatments that have emerged over the past two decades, the number of patients reporting LBP [23,24] and disability claims have continued to rise [25]. Over the past decade, there have been concomitant increases in sales of prescription and illicit (i.e., diversion) opioid medications with a trend toward more potent opioids, as well as deaths from prescription drugs [26-28]. The 2011 Centers for Disease Control and Prevention Morbidity and Mortality Report estimates that there were 20,044 deaths due to prescription drugs in 2008, with 73% due to opioids [28]. In a systematic review and treatment guideline for CLBP published by the American Pain Society (APS), Chou and Huffman reviewed 40 pharmacological randomized controlled trials (RCTs) with just one recommendation based on ‘good evidence’, namely TCAs are moderately more effective than placebo for CLBP. The APS review underscores the great need for safe, effective and non-addictive analgesics [29]. 3.

Existing treatments

Multiple interventions including physical therapy, psychological therapy, injection therapy, surgery and multimodality 2

therapy are used to treat CLBP [30,31]. Although not the focus of this review, it should be emphasized that the evidence for multimodal treatment programs shows significant reduction in pain and prescription drug use, healthcare resources and income support, and an increase in rates of people who return to work. Injections tend to provide only short- or intermediate-term relief to a subset of individuals, and in some instances it may be associated with catastrophic complications [11,32,33]. Most randomized studies of decompression surgery for neurogenic LBP have shown benefit for shortterm (< 1 year) but not long-term (> 2 years) relief [34,35] and most randomized studies of fusion or disc replacement surgery suggest than less than one-third of people can expect clinically meaningful pain relief or functional improvement with the benefits diminishing over time [36]. Psychological and physical and rehabilitation approaches have been shown to provide modest treatment effects in two Cochrane reviews and will not be discussed here [37,38]. Duloxetine is the only non-opioid analgesic to receive FDA approval for the treatment of mechanical LBP, yet clinical trials have shown only modest (£ 1-point) improvement compared to placebo [14,39], and one systematic review found no significant difference between duloxetine and other oral pharmacotherapeutics [40]. A recent responder analysis conducted by Moore et al. demonstrated that patients with CLBP receiving duloxetine responded in a bimodal fashion, experiencing either very good or very poor pain relief [41]. There are currently no drugs approved for neurogenic LBP.

4.

Market review

The overall global market for pain therapeutics grew at a compounded annual growth rate (CAGR) of 5.3% between 2002 and 2010 to an estimated market size of $28.6 billlion [42]. The top eight therapeutic indications by annual sales for pain therapies included postoperative pain, LBP, neuropathic pain, fibromyalgia, osteoarthritis (OA) pain, rheumatoid arthritis pain, cancer pain and migraines. Of these indications, LBP accounted for ~ 17% of the worldwide pain management therapeutics market with $4.9 billion in annual sales [42]. Despite impending patent expirations of the two top selling agents for neuropathic pain (pregabalin with $4.595 billion in annual sales for all indications in 2013 and duloxetine with $5.084 billion in annual sales for all indications in 2013), the expected ‘blockbuster’ sales for new pain drugs have not been realized [43,44]. For example, tapentadol, a µ-opioid agonist and norepinephrine reuptake inhibitor, obtained FDA approval for diabetic peripheral neuropathy in 2011, but annual sales in 2013 are still relatively small at $86 million [45]. The overall market is expected to grow more slowly in the next few years at a CAGR of 3.84%, with a forecast to be ~ $35 billion in size by 2017 [42].

Expert Opin. Emerging Drugs (2014) 20(1)

Expert opinion on emerging drugs: chronic low back pain

Expert Opin. Emerging Drugs Downloaded from informahealthcare.com by Selcuk Universitesi on 12/24/14 For personal use only.

Glial cell antagonists lon channel antagonists

α-2 agonists Bisphosphonates Cytokine inhibitors Gene therapy NGF antagonists NMDA antagonists Opioid agonists ORL-1 agonists PDE inhibitors SNRI, SDRI Stem cell therapy TLR antagonists TRPV1 modulators

NSAIDs

Nociceptive pain Neuropathic pain

Figure 1. Venn diagram showing analgesic class by mechanism of action. NGF: Nerve growth factor; NMDA; N-methyl-D-aspartate; NSAID: Nonsteroidal anti-inflammatory drug; ORL: Opioid receptor like; PDE: Phosphodiesterase inhibitor; SDRI: Serotonin-Dopamine reuptake inhibitor; SNDRI: Serotonin-Norepinephrine-Dopamine reuptake inhibitor; SNRI: Serotonin-Norepinephrine reuptake inhibitor; TLR: Toll-like receptor.

5.

Current research goals

Current research goals include discovering and identifying medications with improved efficacy, longer duration of action and fewer side effects and unwanted sequelae such as addiction. Patients with CLBP could benefit both from the development of novel treatments targeting specific mechanisms as well as diagnostic and prognostic tools for proper risk stratification and personalized therapy. Finally, further study of the underlying pathogenesis of CLBP could better enable the development of disease-modifying therapies.

Scientific rationale for mechanism-based treatment of LBP

6.

About 15 years ago, in a review published in the New England Journal of Medicine, Deyo and Weinstein estimated that neuropathic pain comprised < 10% of LBP [46]. In the intervening years, several instruments have been published that facilitate classification of pain into neuropathic, nociceptive and mixed [7,47]. Multiple studies have since been published using these instruments to categorize CLBP patients into neuropathic or nociceptive and have found that neuropathic pain accounts for between 17 and 55% of cases, with a median rate of around 41% [6,7,10,11]. The distinction is important because it affects treatment decisions at all levels of care (e.g., pharmacotherapy, type of injection or surgery) (Figures 1 and 2)

It is widely acknowledged that the mechanism-based treatment of pain is superior to etiologic or disease-based treatments, yet in practice this is difficult to implement [12]. Identifying the precise mechanism(s) can be challenging, if not impossible. For example, intravenous infusion tests have been used for nearly 20 years as tools to identify pain mechanisms and guide treatment, but these tests generally suffer from low specificity and indeterminate sensitivity and at best have marginal predictive value [48]. Many treatments considered to be effective only for neuropathic pain such as gabapentinoids have proven to be beneficial for nociceptive pain [49], and drugs designed and tested primarily for nociceptive pain such as NSAIDs have shown efficacy for neuropathic pain [50,51]. Part of the reason for this is that there is considerable overlap in pain mechanisms such that the taxonomic classification of pain may bear little relationship to clinical considerations. Consequently, many experts consider it more practical to consider the various types of pain as different points on the same continuum rather than distinct entities [6]. 7.

Competitive environment

A search for compounds in development for CLBP was performed on the clinicalTrials.gov clinical trials registry. Additional developmental compounds were identified through database search of keywords relating to therapeutic targets for the treatment of CLBP (Pharmaprojects, MEDLINE, Embase, Google Scholar). Keyword search terms were

Expert Opin. Emerging Drugs (2014) 20(1)

3

E. Hsu et al.

Cortex

Pain perception and amplification: • Placebo Descending modulation: • Anti-depressants • Opioids, tramadol • Muscle relaxants • Cannabinoids

Expert Opin. Emerging Drugs Downloaded from informahealthcare.com by Selcuk Universitesi on 12/24/14 For personal use only.

Thalamus

Synaptic transmission and central sensitization: • Anti-convulsants • α2 adrenegic agonists • Opioids • NMDA blockers (e.g., ketamine) • Nerve growth factor inhibitors • Cytokine inhibitors • Muscle relaxants • Cannabinoids

PAG

RVM

Peripheral stimulation,transduction, transmission and amplification: • Anti-inflammatory drugs • Topical agents • Anti-convulsants • Cytokine inhibitors • Anti-depressants • Nerve growth factor inhibitors • Cannabinoids

Figure 2. Illustration showing the site(s) of action of various classes of analgesics. Adapted from [6] drawing by Shizuka Aoki. PAG: Periaqueductal gray; RVM: Rostral ventromedial medulla.

identified from the current understanding of mechanisms involved in pain processing. Results were limited to compounds in Phase II or Phase III of development, which were classified based on whether they were new formulations of existing compounds, new indications for existing compounds or represented novel mechanisms of action (Tables 1, 2 and 3). Each compound is categorized by its mechanistic contribution to either nociceptive or neuropathic pain (Figure 1).

New formulations of existing compounds Perhaps in an effort to minimize the enormous cost and risk associated with developing novel compounds, most new pain therapeutics approved in recent years are new formulations of existing drugs [52]. New formulations can be subdivided into 7.1

4

novel delivery mechanisms and formulations designed to improve the safety profile of known adverse effects. The latter approach highlights recent actions by the FDA emphasizing drug safety. In 2007, the FDA Amendments Act granted the FDA the authority to decide when to require a Risk Evaluation and Mitigation Strategy (REMS) from drug sponsors [53]. REMS are risk management programs that use risk minimization strategies to ensure that the benefits of a drug outweigh its risks. Each REMS contains specific measures unique to the safety risks associated with a particular drug or class of drugs. In response to the alarming increase in opioid prescriptions and opioid-related overdose deaths, in February 2009, the FDA began requiring manufacturers of high-risk opioids to incorporate REMS to address abuse, misuse and exposure of non-opioid-tolerant persons to opioids [28,54,55]. In 2012, the FDA approved a

Expert Opin. Emerging Drugs (2014) 20(1)

Expert Opin. Emerging Drugs (2014) 20(1)

Nociceptive pain, neuropathic pain

Pre-registration FDA has accepted for filing the NDA for MNK-155) n = 155 RCT; n = 52

Route of administration

Study design

COX-1 inhibitor COX-2 inhibitor Opioid d-receptor agonist Opioid k-receptor agonist opioid µ-receptor agonist

Sodium channel antagonist

Oral

Transdermal

Intranasal

6h

12 wk

12 wk

Follow-up period

12 wk

Single-center, randomized, double-blind,

N/A

Open-label, single-group- 5 wk assignment study in patients with OA of the knee or hip or moderateto-severe CLBP

Randomized, doubleblind, placebo-controlled in patients with CLBP

Randomized, doubleN/A blind, placebo-controlled, three-period, crossover study in patients with CLBP

Randomized, doubleblind, placebo-controlled in opioid-experienced subjects with CLBP Randomized, doubleblind, placebo-controlled in opioid-naı¨ve subjects with moderate-to-severe CLBP requiring opioid analgesia. a2 adrenoreceptor Oral, spray Oral, Double-blind, placeboagonist sublingual controlled, crossover study in patients with CLBP

Opioid k-receptor Oral, buccal antagonist, Opioid soluble film µ-receptor agonist/ antagonist, ORL 1 agonist

Mechanism of action

Both intact and crushed MNK-155 showed lower

Statistically significant decrease in pain up to 6 h after dosing In Phase II trials for chronic pain [210] Statistically significant improvement in pain symptoms within 30 min of administration and sustained improvement in pain symptoms for up to 4 h In Phase II trials for postoperative and cancer breakthrough pain [210] Did not achieve a statistically significant decrease in pain intensity [211] ClinicalTrials.gov Identifier: NCT01096966. Phase III trial initiated by Impax is expected in late 2014 [212] No results available for Phase III study

May be effective in patients with CLBP who experience opioid-related adverse effects. ClinicalTrials.gov Identifier: NCT01633944 NCT01675167

Comment

CLBP: Chronic low back pain; ER: Extended release; HAL: Human abuse liability; IR: Immediate release; N/A: Not available; NDA: New Drug Application; NRS: Numeric rating score; OA: Osteoarthritis; ORL: Opioid receptor ligand; OXN: Oxycodone/naloxone controlled release; RCT: Randomized controlled trial; wk: Weeks.

Acetaminophe- Mallinckrodt n+hydrocodone, MNK-155 (COV-155) ER oral formulation of hydrocodone

Phase II; n = 263

Nociceptive pain, neuropathic pain

DUR-843

Durect

Phase Ib; n = 24

Phase Ib; n = 21

Phase III; n = 462

Phase III; n = 511

Stage of development

Dex-IN, Dexmedotomidine (sublingual)

Nociceptive pain, neuropathic pain

Dex-SL, Dexme- Orion Pharma dotomidine (sublingual)

Pain mechanism Nociceptive pain, neuropathic pain

Company

Buprenorphine, BioDelivery BEMA LA Sciences (EN-3409)

Compound

Table 1. New formulations of existing compounds.

Expert Opin. Emerging Drugs Downloaded from informahealthcare.com by Selcuk Universitesi on 12/24/14 For personal use only.

Expert opinion on emerging drugs: chronic low back pain

5

6

Company

Pain mechanism

Expert Opin. Emerging Drugs (2014) 20(1)

RCT; n = 24

Phase III; n = 600

RCT; n = 44

Phase III n = 389

Stage of development

Oral

Route of administration

Opioid µ-receptor Oral agonist competitive antagonist at µ, d and k-opioid receptors

Opioid µ-receptor agonist

Mechanism of action

Follow-up period

Single-center, randomized, double-blind, placebo- and activecontrolled study in

Randomized, doubleblind, placebo-controlled study in opioid-exposed subjects with moderateto-severe CLBP

N/A

12 wk

Mechanical manipulation N/A and pharmacokinetic studies in healthy male and female volunteers

double-dummy, activeand placebo-controlled seven-way crossover HAL study assessing the abuse potential of orallyadministered MNK-155 in healthy male and female nondependent recreational opioid users Multicenter, randomized, 12 wk double-blind, placebocontrolled in opioid-naı¨ve and -experienced patients with CLBP

Study design

Sustained release, abusedeterrent oral formulation of oxycodone showed a significant reduction in pain compared with placebo [214]; It may be useful in high-risk patients on opioids. ClinicalTrials.gov Identifier: NCT01685684 DETERx beads retained their ER properties after mechanical tampering and chewing by study subjects [215] Three additional compounds utilizing the DETERx bead technology in preclinical stages of development (COL-172, COL-195, COL-196) [216] Significant decreased in pain scores at wk 12. May be useful in high-risk patients on opioids. ClinicalTrials.gov Identifier: NCT01358526 Significant reductions in drug-liking following administration of OXN compared with oxycodone;

subjective abuse-related effects than an IR hydrocodone bitartrate/ acetaminophen formulation [213]

Comment

CLBP: Chronic low back pain; ER: Extended release; HAL: Human abuse liability; IR: Immediate release; N/A: Not available; NDA: New Drug Application; NRS: Numeric rating score; OA: Osteoarthritis; ORL: Opioid receptor ligand; OXN: Oxycodone/naloxone controlled release; RCT: Randomized controlled trial; wk: Weeks.

Targiniq ER, Purdue Pharma Nociceptive OXN, LP pain, neurooxycodone + pathic pain naloxone ER formulation of oxycodone and naloxone in a fixed 2:1 ratio

Oxycodone Collegium Nociceptive DETERx, Pharmaceutical pain, neuroCOL-003 pathic pain Tamper-resistant, sustainedrelease oral formulation of oxycodone

and acetaminophen

Compound

Table 1. New formulations of existing compounds (continued).

Expert Opin. Emerging Drugs Downloaded from informahealthcare.com by Selcuk Universitesi on 12/24/14 For personal use only.

E. Hsu et al.

Expert Opin. Emerging Drugs (2014) 20(1)

Allodynic Therapeutics

Alpharma, Pfizer

Company

Nociceptive pain, neuropathic pain

Nociceptive pain, neuropathic pain

Pain mechanism

Phase II; n = 78

RCT, n = 41; RCT, n = 32

Phase III; n = 281

Stage of development

Route of administration

Opioid µ-receptor Oral agonist Opioid k-receptor agonist a 2 adrenoreceptor agonist competitive antagonist at µ, d and k- opioid receptors

Opioid µ-receptor Oral agonist competitive antagonist at µ, d and k- opioid receptors

Mechanism of action

Follow-up period

Two abuse potential N/A studies randomized, double-blind, double-dummy, placeboand active-controlled, 6-way crossover study in healthy, nondependent, recreational opioid users with oral administration; randomized, double-blind, placebo- and activecontrolled, 4-way crossover study in healthy, nondependent, recreational opioid with intranasal administration Randomized, double3 wk blind, placebo-controlled trial in subjects with chronic spinal pain in the lumbar, thoracic or cervical regions

Multicenter, randomized, 12 wk double-blind, placebocontrolled study of subjects with moderateto-severe CLBP

nondependent recreational opioid users

Study design

Completed Phase II for CLBP. Results not available. ClinicalTrials.gov Identifier: NCT01415895 Phase III trials for trigeminal neuralgia were withdrawn prior to enrollment for undisclosed reason. ClinicalTrials.gov Identifier: NCT01920087

comparable to placebo [217] Available in 29 countries outside US [218] 57.5% had at least 30% decrease in NRS of pain compared to 44% patients treated with placebo; it may be useful in high-risk patients on opioids. 39.7% patients had at least a 50% decrease in NRS of pain compared to 29.9% patients in the placebo group Statistically significant lower scores for drug-liking and ‘high’ than equivalent doses of crushed IR oxycodone. Statistically significant lower scores for drug-liking and ‘high’ relative to crushed IR oxycodone [219]

Comment

CLBP: Chronic low back pain; ER: Extended release; HAL: Human abuse liability; IR: Immediate release; N/A: Not available; NDA: New Drug Application; NRS: Numeric rating score; OA: Osteoarthritis; ORL: Opioid receptor ligand; OXN: Oxycodone/naloxone controlled release; RCT: Randomized controlled trial; wk: Weeks.

Naltrexone + clonidine, ATNC-05

Oxycodone + naltrexone, ALO-02

Compound

Table 1. New formulations of existing compounds (continued).

Expert Opin. Emerging Drugs Downloaded from informahealthcare.com by Selcuk Universitesi on 12/24/14 For personal use only.

Expert opinion on emerging drugs: chronic low back pain

7

8

Company

Expert Opin. Emerging Drugs (2014) 20(1)

Nociceptive pain

Nociceptive pain, neuropathic pain

Pain mechanism

Phase II; n = 127

Phase II

Stage of development

NSAIDs

Opioid µ-receptor agonist, opioid d-receptor agonist, opioid k-receptor agonist

Mechanism of action

Transdermal

Oral

Route of administration

Comment

May have few side effects. No results published . ClinicalTrials.gov Identifier: NCT00759330. Drug does not appear on webpage of company pipeline, another compound TPH-023 appears, but no other information of registered trials is available [221]

No information Ongoing Phase II trials for available chronic pain [220] may be useful in high-risk patients on opioids

Follow-up period

Multicenter, randomized, 1 wk double-blind, placebocontrolled study in patients with CLBP

No information available

Study design

CLBP: Chronic low back pain; ER: Extended release; HAL: Human abuse liability; IR: Immediate release; N/A: Not available; NDA: New Drug Application; NRS: Numeric rating score; OA: Osteoarthritis; ORL: Opioid receptor ligand; OXN: Oxycodone/naloxone controlled release; RCT: Randomized controlled trial; wk: Weeks.

Morphine+oxyc- QRxPharma odone, Q-8011CR (MoxDuoCR) abuse-deterrent formulation of oral controlled release combination of the opioids morphine and oxycodone Flurbiprofen Teikoku Tape

Compound

Table 1. New formulations of existing compounds (continued).

Expert Opin. Emerging Drugs Downloaded from informahealthcare.com by Selcuk Universitesi on 12/24/14 For personal use only.

E. Hsu et al.

Expert Opin. Emerging Drugs (2014) 20(1)

N/A

Pregnenolone

Nociceptive pain, neuropathic pain

Nociceptive pain, neuropathic pain

Nociceptive pain, neuropathic pain

Pain mechanism

Route of administration

Oral

Intravenous

Serotonin and Oral norepinephrine reuptake inhibitor

Mechanism of action

Bisphosphonate; mechanism of analgesic effect possibly related to inhibition of Phase II; osteoclasts and painn = 44 inducing substances (IL-1, PGE-2, lactic acid, etc.) Phase II and III; GABAa receptor n = 90 modulation, antiinflammatory effects

Phase II; n = 0/48

Phase II; n = 35

Phase IV; n = 18

Stage of development

CLBP: Chronic low back pain; LBP: Low back pain; mo: months; PGE: Prostaglandin E; VAS: Visual Analog Scale; wk: Weeks.

N/A

Forest/Cypress

Milnicipran (Savella)

Pamidronate

Company

Compound

Table 2. New indications for existing compounds.

Randomized, doubleblind, placebo-controlled in patients with radicular pain related to lumbosacral disc disease Randomized, doubleblind, placebo-controlled study in 35 patients with chronic neuropathic LBP Randomized, doubleblind, placebo-controlled in patients with degenerative disk disease Randomized, double blind, placebo-controlled in patients with nonspecific, mechanical predominantly axial CLBP Randomized, double blind, placebo-controlled in patients with CLBP

Study design

6 wk

Currently recruiting; ClinicalTrials.gov Identifier: NCT01898013

Significant reduction in pain reported compared to placebo [67]

6 mo

6 mo

6 wk

Reduction in pain scores observed but no statistical analysis provided. ClinicalTrials.gov Identifier: NCT01777581 Effect size of VAS pain effect was 0.22. ClinicalTrials.gov Identifier: NCT01225068 Currently recruiting; ClinicalTrials.gov Identifier: NCT01799616

Comment

10 wk

Follow-up period

Expert Opin. Emerging Drugs Downloaded from informahealthcare.com by Selcuk Universitesi on 12/24/14 For personal use only.

Expert opinion on emerging drugs: chronic low back pain

9

10

Expert Opin. Emerging Drugs (2014) 20(1)

Pfizer, Lilly

Tanezumab (RN624)

TNF a inhibitor

Phase II -- III; n = 84

Nociceptive pain Phase II; n = 389

Phase II; n = 1347

Epidural

Oral

NGF antagonist Subcutaneous

NGF antagonist Intravenous or subcutaneous

ORL-1 receptor agonist

Mechanism of Route of action administration

Phase II; n = 1089

Stage of development

Nociceptive pain Phase II; n = 225

Nociceptive pain, neuropathic Pain Nociceptive pain, neuropathic pain

Indication

Follow-up period

Multicenter, randomized, double-blind, placebocontrolled study in patients with moderateto-severe CLBP.

Multicenter, randomized, double-blind, placeboand active-controlled study in patients with CLBP

12 wk

16 wk

Randomized, double14 wk blind, placebo-controlled study in patients with moderate-to-severe CLBP Multicenter, randomized, 6 mo double-blind, placebocontrolled study in patients with lumbosacral radiculopathy comparing etanercept to steroid and saline Multicenter, randomized, 12 wk double-blind, placeboand active-controlled study in patients with CLBP requiring regular use of analgesic medication

Study design

CLBP: Chronic low back pain; d: Days; LBP: Low back pain; mo: Months; NGF: Nerve growth factor; OA: Osteoarthritis; ORL: Opioid receptor ligand; wk: Weeks.

Amgen

Amgen, Pfizer

Etanercept

Fulranumab (AMG-403, JNJ-42160443)

Grunthal

Company

Cebranopadol (GRT 6005)

Compound

Table 3. Novel mechanisms of action.

Expert Opin. Emerging Drugs Downloaded from informahealthcare.com by Selcuk Universitesi on 12/24/14 For personal use only.

Greater proportions of patients reported ‡ 30 and ‡ 50% reduction in acute LBP with tanezumab versus naproxen (p £ 0.013) and placebo (p < 0.001) [79] ClinicalTrials.gov Identifier: NCT00584870 Tanezumab 10 and 20 mg had similar efficacy profiles and significantly improved pain scores versus both placebo and naproxen [80]. ClinicalTrials.gov Identifier: NCT00876187. Clinical trials suspended at request of FDA over concerns of rapidly progressive OA leading to joint replacement [81]. Pfizer received notification from the FDA on 19 July 2013 that the clinical hold for tanezumab had been lifted pending the submission and review of nonclinical data [82] Phase II trial terminated due to lack of efficacy; ClinicalTrials.gov Identifier: NCT00973024

Modest improvement in leg and back pain, but worse disability index related to sleep and sex [94]. ClinicalTrials.gov Identifier: NCT00733096

Study completed. Not published. ClinicalTrials.gov Identifier: NCT01725087

Comment

E. Hsu et al.

Mesoblast, Ltd

Red de Terapia Nociceptive Celular (Vallapain, neurodolid University) pathic pain

Mesenchymal stem cell

Expert Opin. Emerging Drugs (2014) 20(1)

Mesenchymal stem cell

Phase I; n = 0/24

Phase I; n = 0/100

Phase II; n = 141

Phase II; n = 165

Phase III; n = 325

Phase II; n = 39

Stage of development Oral

Mesenchymal stem cell

Mesenchymal stem cell

N-type calcium channel inhibitor

Glial cell modulator

Intradiscal

Intradiscal

Oral

Intravenous

Triple reuptake Oral inhibitor of dopamine, norepinephrine, serotonin, low NMDA receptor affinity

NMDA partial agonist

Mechanism of Route of action administration

Follow-up period

Multicenter, randomized, double-blind, placebocontrolled study in patients with lumbar radiculopathy Randomized, doubleblind, placebo-controlled study in patients with lumbosacral radiculopathy Multicenter, randomized, double-blind, placebocontrolled study in patients with chronic discogenic lumbar back pain Randomized, doubleblind, placebo-controlled study in patients with degenerative disc disease

12 mo

36 mo

6 wk

10 d

Randomized, double12 wk blind, placebo-controlled study in patients with neuropathic CLBP Three multicenter, 3 mo randomized, double-blind, placebo-controlled trials in patients with CLBP

Study design

CLBP: Chronic low back pain; d: Days; LBP: Low back pain; mo: Months; NGF: Nerve growth factor; OA: Osteoarthritis; ORL: Opioid receptor ligand; wk: Weeks.

Nociceptive pain, neuropathic pain

Neuropathic pain

Zalicus

Neuropathic pain

Neuropathic pain

Z-160

DOV pharmaceutical

Bicifadine

Neuropathic pain

biogen idec

Sponsored by Northwestern University

D-cycloserine

Indication

Neublastin (BG00010)

Company

Compound

Table 3. Novel mechanisms of action (continued).

Expert Opin. Emerging Drugs Downloaded from informahealthcare.com by Selcuk Universitesi on 12/24/14 For personal use only.

Not yet recruiting; ClinicalTrials.gov Identifier: NCT01860417

Not yet recruiting; ClinicalTrials.gov Identifier: NCT01290367

Completed, results unpublished. ClinicalTrials.gov Identifier: NCT01655849

ClinicalTrials.gov Identifier: NCT00295711; NCT00295724; NCT00281645; Phase III completed in 2011 but found no significant reduction in pain intensity; results confounded by poor compliance [101] Currently recruiting for Phase II trial. ClinicalTrials.gov Identifier: NCT01873404

Study is ongoing; ClinicalTrials.gov Identifier: NCT00125528

Comment

Expert opinion on emerging drugs: chronic low back pain

11

Expert Opin. Emerging Drugs Downloaded from informahealthcare.com by Selcuk Universitesi on 12/24/14 For personal use only.

E. Hsu et al.

REMS for extended release (ER) and long-acting opioid medications [56]. Two abuse-deterrent formulations of opioids have gained recent approval, morphine/naltrexone combination (Embeda; King Pharma) and oxycodone/niacin (Oxecta/Acurox; King Pharma/Acura Pharmaceuticals), with an additional four compounds currently in Phase II or Phase III development. Similar measures have been undertaken with NSAID-class compounds to minimize the risk of serious gastrointestinal (GI) ulceration and hemorrhage by combining an NSAID with a GI protectant. Cost analyses have determined that in certain populations (e.g., the elderly and those on anticoagulants), the cost of treating GI-related adverse events approaches or exceeds the cost of therapy [57-61]. Systematic reviews have determined that combination therapy (i.e., NSAID plus protein pump inhibitor, H2 blocker or misoprostol) may be a more cost-effective strategy than the use of a COX-2 inhibitor [62]. Two compounds have gained recent approval: naproxen/esomeprazole (Vimovo; Pozen/ AstraZeneca) and ibuprofen/famotidine (Duexis/HZT-501; Horizon Pharma). In clinical trials, both formulations significantly reduced the incidence of NSAID-related GI ulcers [52]. New indications for existing compounds The recent success of the antidepressant duloxetine/Cymbalta (Eli-Lilly) in the treatment of neuropathic and nociceptive pain (including LBP) spurred the development of the antidepressant Milnacipran/Savella (Forest/Cypress). Savella is a selective serotonin and norepinephrine reuptake inhibitor currently approved for the treatment of fibromyalgia. Additional compounds with new indications on the horizon include pamidronate, a bisphosphonate whose class has been shown in clinical studies to alleviate pain from osteoporosis, bone metastases and complex regional pain syndrome, suggesting that it may be effective for both neuropathic and nociceptive CLBP [63,64], and the neurosteroid pregnenolone, which has been shown in animal models to alleviate neuropathic and nociceptive pain [65,66]. Pamidronate recently completed a small (n = 44) Phase II trial for nociceptive CLBP in which two infusions were found to provide significant pain relief compared to placebo through 6-month follow up [67]. Another small prospective study in elderly postmenopausal women with osteoporosis and CLBP found that risedronate therapy over 4 months improved LBP in the absence of vertebral fractures, suggesting that bone resorption due to osteoporosis may be a cause of LBP. This is bolstered by the apparent efficacy of bisphosphonates in relieving pain due to bony metastases [68]. A recent meta-analysis of RCTs in patients with bone pain due to metastatic disease showed that the likelihood of experiencing bone pain was significantly lower in patients taking zoledronate compared with placebo [69]. 7.2

Novel mechanisms of action Despite the plethora of potential drug targets represented by the inflammatory milieu underlying CLBP, the actual number of compounds in development is small. In contrast, there are at 7.3

12

least 25 compounds in development for neuropathic pain [70]. Of these 25 compounds, only cebranopadol (Grunenthal), an opioid-like receptor -1 (ORL-1) receptor agonist, is being studied in trials as a possible treatment for CLBP. A similar scenario exists when considering OA-associated pain, whereby nine compounds are currently in development [71]. To place this in context, facet joint (e.g., arthritis) pain comprises ~ 10 -- 15% of CLBP, with the prevalence increasing with age [72]. There are a total of nine compounds currently being developed for CLBP including five anti-nerve growth factor (NGF) antibodies, namely, Tanezumab/RN624 (Pfizer, Eli-Lilly), Fulranumab/AMG-403/JHJ-42160443 (Amgen, Johnson & Johnson), REGN-475 (Sanofi-Aventis/Regeneron), MEDI-578 (AstraZeneca) and PG110 (Abbott).

NGF inhibitors NGF plays a role in the development and maintenance of nociceptive and neuropathic pain. Rodent models of NGF injected locally and systemically in animal models have demonstrated hyperalgesia and allodynia, and inflamed peripheral tissues exhibit elevated concentrations of NGF [73-75]. Furthermore, NGF inhibition and knockout models reveal reduced pain perception in models of acute local inflammation, chronic inflammatory arthritis, OA, postoperative, visceral and neuropathic pain [76]. In RCTs, NGF inhibitors have been effective in different pain conditions including OA, interstitial cystitis and diabetic neuropathy. Four RCTs of three different compounds have been conducted in patients with CLBP, two with tanezumab, one with fulranumab and one with REGN-475. Thus far, only the two trials of tanezumab have been published in peer-reviewed journals, with both demonstrating statistically significant reduction in pain over placebo [77]. Despite demonstrating efficacy in both pain and functional measures for hip and knee arthritis, a Phase II trial evaluating fulranumab as an adjunctive treatment in 389 patients with CLBP inadequately controlled with conventional medications did not demonstrate a statistically significant difference over placebo at week 12 of treatment [78]. Both tanezumab trials studied patients with non-neuropathic CLBP and found superior pain relief and functional improvement over naproxen and placebo at 12 and 16 weeks of follow up, respectively [79,80]. In 2010, all clinical trials for anti-NGF compounds were suspended at the request of the FDA over concerns of rapidly progressive OA and reports of osteonecrosis leading to joint replacement [81]. A 2012 Arthritis Advisory Committee voted unanimously to restart clinical trials scheduled to resume in 2015 due to a lack of evidence for a direct molecular mechanism linking anti-NGF antibodies to joint destruction [82]. Moreover, it was reported that Pfizer received notification from the FDA on 19 July 2013 that the ‘clinical hold’ for tanezumab had been lifted pending the submission and review of nonclinical data [83]. 7.3.1

Expert Opin. Emerging Drugs (2014) 20(1)

Expert opinion on emerging drugs: chronic low back pain

Expert Opin. Emerging Drugs Downloaded from informahealthcare.com by Selcuk Universitesi on 12/24/14 For personal use only.

7.3.2

Glial cell modulators

Following nerve injury, glial cells proliferate at the dorsal root ganglia (DRG) and spinal cord levels where they stimulate the complement component of the immune system, resulting in the release of cytokines, chemokines and cytotoxic substances such as nitric oxide and free radicals, all of which are integrally involved in the development and maintenance of neuropathic pain [6,84]. Much attention has been focused on glial cell inhibitors due to their potential in reducing abuse-related reward and potentiating the analgesic effects of opioids [85]. Although promising in preclinical animal models, glial cell modulators such as pentoxifylline, propentofylline, ibudilast have not demonstrated significant efficacy in clinical trials for neuropathic pain conditions other than CLBP [6,86]. Adverse reactions of FDA-approved glial cell modulators include nausea, vomiting, anaphylaxis and hemorrhage in combination with anticoagulants including NSAIDs. Cytokine inhibitors Cytokine inhibitors have been extensively studied in neuropathic LBP. TNF-a) is the prototypical proinflammatory cytokine due to its prominent role in initiating the activation cascade of other cytokines and growth factors in the inflammatory response. As a drug class, the TNF-a inhibitors are associated with multiple potential adverse events, which include injection-site reactions, infusion reactions, neutropenia, infections, demyelinating disease, heart failure and cutaneous reactions such as psoriasis, malignancy and autoimmune reactions. In preclinical studies, the preemptive administration of TNF inhibitors has been shown to prevent the pathological and behavioral changes associated with application of nucleus pulposus to spinal nerve roots, suggesting a possible role in radiculopathy [87,88]. TNF inhibitors have also been shown in clinical trials to be effective for inflammatory arthritis, including ankylosing spondylitis, indicating that they may be beneficial for mechanical spinal disorders [89]. However, the results of clinical trials in mechanical spinal pain have been mixed. After positive findings from a case series involving 10 patients with acute sciatica, a randomized controlled study by Korhonen et al. found no benefit from a single infusion of the TNF inhibitor infliximab in 40 patients with unilateral sciatica < 12 weeks in duration [90,91]. In another placebo-controlled trial, Genevay et al. demonstrated a modest benefit for adalimumab 6 month after two subcutaneous injections in 61 patients with acute radiculopathy [92]. The results from injections of TNF inhibitors have yielded similar conflicting results. A small, double-blind study comparing perispinal, intramuscular etanercept with placebo found no benefit in 15 patients with acute radicular pain [93]. Two placebo-controlled studies evaluating epidural etanercept failed to show any benefit in patients with neuropathic spinal pain, whereas two other randomized studies demonstrated significant benefit over epidural dexamethasone at 4 weeks and epidural saline through 6 months [94-97]. 7.3.3

Collectively, these findings suggest the possibility of a modest effect for TNF inhibitors in individuals with acute or subacute radicular LBP. Serotonin, norepinephrine and/or dopamine reuptake inhibitors

7.3.4

Inhibition of the reuptake of neurotransmitters involved in pain modulation is the rationale for prescribing antidepressants to treat CLBP. The descending fibers of the dorsolateral funiculus in the spinal cord are responsible for suppressing nociceptive neurotransmission. These fibers contain serotonergic projections from the raphe nuclei, dopaminergic projections from the ventral tegmental area and noradrenergic projections from the locus coeruleus [98]. Several subclasses of antidepressants have being used for treating chronic pain including tetracyclic antidepressants (amoxapine, maprotiline), serotonin and noradrenaline reuptake inhibitors (SNRIs; duloxetine, venlafaxine, milnacipran), atypical antidepressants (bupropion, trazodone, mirtazapine, nefazodone), selective serotonin reuptake inhibitors (SSRIs; citalopram, fluoxetine and paroxetine), serotonin, dopamine and noradrenaline reuptake inhibitors (SNDRIs; bicifadine) and TCAs (amitriptyline, doxepin, imipramine, desipramine and nortriptyline) [99]. This class of medications, particularly TCAs, may be associated with anticholinergic side effects (dry mouth, double vision), drowsiness, insomnia/delirium, orthostatic hypotension, QTc prolongation, GI toxicity, weight gain and sexual dysfunction. Bicifadine, a SNDRI with low NMDA receptor affinity that has shown preclinical [100] and clinical efficacy in postoperative pain [101,102], failed to show an effect in patients with CLBP in a Phase III clinical trial. However, a subsequent analysis stratifying patients according to compliance found significant reduction in pain scores compared to placebo in those with higher plasma levels [103]. Based on the APS guidelines for the treatment of CLBP, paroxetine and trazodone were not effective compared to placebo, TCAs were slightly to moderately more effective than placebo, and there was insufficient evidence to judge the effectiveness of TCAs versus SSRIs [29]. Investigational compounds on the horizon include TD-9855, Theravance, a SNRI, in Phase II development in patients with fibromyalgia [104]. Toll-like receptor antagonists Toll-like receptors (TLR) are transmembrane patternrecognition receptors found both intracellularly and extracellularly. Activation of these receptors causes a conformational change and the production and release of proinflammatory cytokines such as IL-6, IL-1b and TNF-a, which are implicated in initiating and maintaining chronic pain states. Additionally, endogenous ligands, damage-associated molecular patterns or ‘alarmins’, released from damaged tissue are hypothesized to be associated with activation of TLR in neuropathic pain. Most research has focused on TLRs role in neuropathic pain, particularly TLR2, 3 and 4, but more recent studies also demonstrate a role in nociceptive pain [105,106]. 7.3.5

Expert Opin. Emerging Drugs (2014) 20(1)

13

E. Hsu et al.

Currently, AV411 (Ibudilast), a TLR4 antagonist which also acts as a glial cell inhibitor, is in clinical trials for chronic migraine and various withdrawal syndromes including from opioids, alcohol and methamphetamines [107]. In clinical trials, the most frequently reported adverse effects were GI in nature including nausea, diarrhea and dyspepsia. Although there is no evidence of efficacy yet for analgesia in clinical trials, this category of drugs has tremendous promise given its safety profile and theoretical role in reducing substance dependence. Novel opioid agonists Opioid drugs are defined by their target receptor types and act both peripherally and centrally to decrease nociceptive pain. They are also used in neuropathic pain but have reduced efficacy in this context and are associated with a wide range of side effects including constipation, nausea and emesis, sedation, respiratory depression, tolerance, dependence and addiction. There are several different endogenous opioid receptors and subtypes including, µ, k, d and ORL-1 which are targets for emerging pharmacotherapy. Receptors are found in both the peripheral system and CNS, with variable distribution. ORL-1 receptors are not expressed in the same areas as µ-, k- and d-opioid receptors. Instead, they are located in the amygdala septum, hypothalamus, thalamus and perikarya in the DRG and spinal cord [108]. Currently, clinically available opioids all act on µ-receptors; there are several opioids in clinical trials that act on other opioid receptors. The potential benefits of opioids such as k-opioid receptor agonists are less sedation, less constipation, less respiratory depression and less addiction [109]. Emerging therapies in this class of drugs include NKTR 181, a µ-opioid in Phase II trials for knee pain for OA. Buprenorphine, which is a partial µ-agonist/antagonist and k-antagonist, is in Phase III trials for efficacy in opioid-tolerant patients with CLBP. Previous RCTs have demonstrated improvement in pain and quality of life with buprenorphine in opioid-naı¨ve patients with CLBP [110,111]. Two Phase II trials have been completed evaluating CR845, an intravenous k-opioid agonist, for postoperative pain after hysterectomy and bunionectomy. Six Phase II studies have been completed evaluating cebranopadol (GRT 6005), an ORL-1 receptor agonist, in patients with pain symptoms ranging from CLBP, knee OA, diabetic neuropathy and bunionectomy pain. In recruitment is a Phase III trial comparing cebranopadol with morphine-prolonged release in patients with chronic cancer pain [70].

Expert Opin. Emerging Drugs Downloaded from informahealthcare.com by Selcuk Universitesi on 12/24/14 For personal use only.

7.3.6

a-2 agonists Direct a-2 adrenergic agonists such as clonidine and dexmedetomidine bind to a-2 adrenergic receptors found in central (supraspinal and spinal) and peripheral (smooth muscle) sites. Their clinical utility lies in their ability to provide analgesia, sedation and sympatholysis. There are three different receptor subtypes -- a2A, a2B, and a2C, whose effects are mediated through G-protein receptor coupling to effectors with different mechanisms. Analgesia is mediated through all three 7.3.7

14

receptor subtypes and may result in the alleviation of neuropathic pain [112]. AGN 2038181 (Rezatomidine) is an a2B adrenoreceptor agonist in Phase II clinical trials for diabetic peripheral neuropathy [104]. Although these drugs are widely used in regional anesthesia, their side effects, including bradycardia, hypotension and sedation, will likely limit their use in the chronic setting. Ion channel antagonists Ion channels are ubiquitous components of the plasma membrane of almost all cells in the human body and are characterized by selective permeability to specific electrically charged ions whose entry or exit modulates neurotransmission. Passage through ion channels is governed by a ‘gate’, which may be opened or closed in response to temperature (i.e., transient receptor potential vanilloid type 1 [TRPV1]), ligand binding (i.e., NMDA receptor) or chemical or electrical signals (i.e., voltage-gated sodium channels). 7.3.8

Vanilloid receptor TRPV1 modulators The vanilloid receptor or TRPV1 receptor is a nonselective cation channel expressed both centrally and peripherally [113-117]. It is abundantly expressed on C-fibers and A-delta fibers and localized to DRG and trigeminal ganglia cells [113,114]. Intrinsically, TRPV1 responds to a variety of endogenous ligands as well as changes in pH (< 6), temperature (> 42 C) and membrane depolarization, and receptor activation ultimately leads to the influx of Na+ and Ca2+ ions into cells [116-118]. TRPV1 is upregulated during inflammation and plays a role in inflammation-induced pain behaviors [119-124]. TRPV1 knockout mice exhibit decreased thermal hypersensitivity after inflammation [125,126]. Interestingly, both agonists and antagonists have the ability to provide analgesia, with the TRPV1 antagonist class offering a novel mechanistic treatment for both nociceptive and neuropathic pain. Agonists such as capsaicin (active ingredient in hot chili peppers) and the vanilloid analog resiniferatoxin cause desensitization of TRPV1 channels and relief of pain behaviors in inflammatory, neuropathic and cancer pain models [114,127]. However, human studies have found systemic administration problematic due to the prevalence of side effects (e.g., high blood pressure, respiratory effects). Efforts to create a nonpungent and lower potency agonist (olvanil) did not progress to clinical trials due to poor pharmacokinetics and limited bioavailability [128]. High-dose topical therapy and local injections have shown positive pain relief in some human trials [129]. 4975 (Adlea) developed by Anesiva demonstrated significant reductions in post-bunionectomy, intermetatarsal neuroma, and lateral epicondylitis with a single local injection [130-132]. Two clinical trials evaluating intra-articular injections in patients with end-stage knee OA have been conducted with conflicting results [133,134]. Another compound, Civamide (Zucapsaicin), is an intranasal formulation under investigation for the treatment of cluster and migraine headaches, with mixed results thus far [128]. 7.3.8.1

Expert Opin. Emerging Drugs (2014) 20(1)

Expert Opin. Emerging Drugs Downloaded from informahealthcare.com by Selcuk Universitesi on 12/24/14 For personal use only.

Expert opinion on emerging drugs: chronic low back pain

The potential to treat a broad range of acute and chronic pain conditions has set the stage for an aggressive search for clinically effective compounds [128,135-143]. Current Phase II compounds include DWP-05195 (Daewoong Pharm), AZD-1386 (AstraZeneca), GRC-6211 (Glenmark/Lilly), JTS-653 (Japan Tobacco), MK-2295 (Merck/Neurogen) and SB-705498 (GlaxoSmithKline). Hyperthermia is a potential side effect of TRPV1 agents, and previous trials were either terminated for hyperthermia (AMG 517) [126] or exhibited a significant hyperthermic effect (ABT 102) [124,127,128,144]. Additional Phase II studies evaluating GRC-6211 and AZD-1386 were suspended for undisclosed reasons and elevated liver enzymes, respectively [145]. The emergence of these adverse effects has generated some concern regarding the complexity of TRPV1 and its role in temperature homeostasis [142]. Recently, Trevisani and Gatti described five compounds tested in preclinical animal models that achieved anti-nociception without affecting core body temperature -- BCTP, GRC6211, PHE377, AS1928370 and AS1928370 [143,145-148]. Glutamate receptor subtype NMDA Glutamate is a neurotransmitter with a well-established role in somatosensory pain processing [149]. A noxious peripheral stimuli triggers primary afferent nociceptive terminals to release glutamate which binds to two receptor subtypes: i) kainite; and ii) (R,S)-a- AMPA receptors. With continued activation of nociceptive signaling pathways as occurs in chronic pain, the release of glutamate leads to prolonged membrane depolarization and removal of the tonic magnesium inhibition of the third glutamate receptor subtype, the NMDA receptor [150-153]. Unlike AMPA and kainate receptors, the ability of NMDA receptors to initiate Ca2+-mediated intracellular signaling pathways results in long-term potentiation, opioid tolerance and the development of chronic neuropathic pain [153]. The NMDA receptor ion channel is a complex pharmacological target with a heterotetrameric structure that consists of up to seven subunits, each of which contains numerous allosteric sites which serve as potential targets [154]. The NMDA receptor is widely distributed throughout the CNS and is involved in multiple important physiological (learning and memory) and pathological processes (chronic pain, neurodegeneration and dementia), complicating the development of useful pharmacological agents [153]. Whereas the most obvious target is the ligand binding site, and highly selective and potent NMDA antagonists are available, this class has not shown much utility due to excessive side effects including dissociative anesthesia (amnesia, catalepsy), hallucinations, confusion, agitation and psychosis [153,155]. Attempts to decrease off-target effects have centered on identification of low-affinity channel blockers which can inhibit ‘wind-up’ without affecting physiologically appropriate responses to noxious stimuli [153,156]. However, clinical results have been inconsistent. High-dose dextromethorphan achieved a modest (24%) reduction in pain intensity in 7.3.8.2

patients with diabetic neuropathy but failed to exhibit a significant effect in postherpetic neuralgia patients [157,158]. A double-blind, placebo-controlled trial assessing the antiparkinsonian drug amantadine demonstrated relief of postsurgical neuropathic pain in cancer patients; however, its structurally related analog memantine yielded no difference compared to placebo in patients with diabetic neuropathy, postherpetic neuralgia and phantom limb pain [159,160]. A major challenge in analgesic clinical trials involves inadequate dosing in the face of a narrow therapeutic ratio, and the class of NMDA receptor antagonists is a good example of this -- hence the better responses shown with higher doses of low-affinity NMDA antagonists. Studies by Sang et al. and Nelson et al. showed an effect for high-dose dextromethorphan compared to an inert as well as an active placebo (lorazepam) [160,161]. A 2012 review of novel NMDA receptor modulators by Santangelo et al. discussed two novel agents: i) a PCP analog, bicyclo-4-F-PCP, which demonstrated decreased acute cell line toxicity; and ii) AQ444 compound 8, for use in the treatment or prevention of diseases characterized by impairment of higher cerebral functions [158]. In an effort to develop more selective compounds with potentially reduced side-effect profiles, research has focused on targets such as the NR2B subunit, which is expressed in DRG cells and superficial layers of the dorsal spinal horn -- critical areas of pain processing [162]. Two early drugs ifenprodil and eliprodil failed in development due to crossreactivity with other molecular targets (serotonin receptors, a1-adrenergic receptors and cardiac ion channels) [163]. The development of more selective compounds has met greater success in animal pain models of inflammatory and neuropathic pain, with decreased cross-reactivity profiles [163,164]. One compound, traxoprodil (CP-101,606), provided relief in a clinical trial to patients suffering from both central (spinal cord injury) and peripheral (monoradiculopathy) neuropathic pain, without significant psychomimetic symptoms [165]. The most serious adverse effects were centrally mediated (depression, dizziness, hypoesthesia). Glycine is an obligatory NMDA receptor co-agonist, binding to a site on the NR1 subunit [154]. The binding of glycine potentiates NMDA receptor activity; thus, glycine B antagonists enhance desensitization and reduce NMDA responsiveness. Additionally, glycine B antagonists appear to be devoid of the typical psychomimetic and neurotoxic effects associated with NMDA receptor antagonists. However, these compounds can cause ataxia and motor relaxation [166]. Several glycine antagonists have shown positive effects in preclinical neuropathic and inflammatory pain models [167]. Only one clinical trial has been reported in which the oral administration of GV196771 reduced areas of allodynia but did not improve evoked pain intensity or provide pain relief [167]. Voltage-gated sodium channels Within the human CNS, sodium channels play a role in the generation of nerve impulses in excitable cells such as neurons, 7.3.8.3

Expert Opin. Emerging Drugs (2014) 20(1)

15

Expert Opin. Emerging Drugs Downloaded from informahealthcare.com by Selcuk Universitesi on 12/24/14 For personal use only.

E. Hsu et al.

glial cells and cardiac myocytes. Existing sodium channel blocking drugs such as lidocaine, mexiletine and tocainide are effective for neuropathic pain, but they are nonspecific and therefore characterized by myriad side effects. Pain research over the past two decades has been focused on identifying sodium channels that are upregulated and heterotopically expressed in DRG and injured axons following nerve injury. There are currently nine different genes that encode nine distinct sodium channel isoforms (NaV1.1 -- NaV1.9), with different amino acid sequences, different physiological properties and different distributions within the nervous system. Of these, five isoforms, NaV1.3, 1.6, 1.7, 1.8 and 1.9 have emerged as attractive molecular targets for pain therapies. NaV1.7 has attracted special interest because of its key role in producing human pain syndromes such as erythromelalgia, an inherited disorder characterized by severe burning pain of the hands and feet caused by a gain-of-function mutation in NaV 1.7, and congenital insensitivity to pain, a disorder in which individuals with a loss-of-function mutation in NaV 1.7 cannot feel painful stimuli. Some NaV1.7 mutations can alter the sensitivity of sodium channels to antagonists such as mexiletine and carbamazepine [168,169]. Several clinically available nonselective sodium channel antagonists belong to the class I antiarrhythmic category and therefore possess negative inotropic and proarrhythmic effects. Other side effects include seizures, dermatitis and hypersensitivity syndromes. In January 2014, the European Commission issued a grant for e4.8 million for the PROPANE study, a multisite, international prospective cohort study that aims to stratify patients with painful neuropathy using targeted genomic sequencing [170]. Although not yet completed, the results could usher in a new era for personalized genomically targeted pain pharmacotherapeutics [171]. Voltage-gated calcium channels Membrane depolarization leads to an opening of voltagesensitive calcium channels, calcium influx into the cell and subsequent neuronal, glial cell and muscle excitation. Preclinical studies have focused on compounds that inhibit N, T and a2d subunit-type calcium channels, thus modulating the release of neurotransmitters at presynaptic terminals in the spinal cord. The most widely used class of agents for CLBP remain gabapentinoids such as gabapentin (including an ER formulation), pregabalin and enacarbil, which act on the a2d subunit. All of these agents were FDA-approved for CNS diseases or neuropathic pain states other than CLBP, and clinical trials evaluating gabapentinoids for CLBP have been decidedly mixed. Although two small randomized trials demonstrated benefit for gabapentin in neuropathic LBP [172,173], a larger randomized controlled study evaluating pregabalin in patients with chronic lumbosacral radiculopathy failed to demonstrate any benefit compared to placebo [16]. For combination therapy, a small randomized trial found the combination of celecoxib and pregabalin to be superior to either drug alone in a mixed CLPB population with 7.3.8.4

16

nociceptive and neuropathic pain, with the most marked benefit noted in those with neuropathic pain [174]. However, a much larger study conducted in patients with neuropathic LBP found no difference between tapentadol prolonged release as a stand-alone therapy and tapentadol prolonged release in combination with gabapentin [175]. A major challenge to the development of drugs within this class is the ubiquitous distribution of calcium channels in neurons, glial cells, pacemaker cells, myocytes and even osteocytes [176]. Current pharmaceutical development has focused on N- and T-type calcium channel blockers which illustrate off-target effects. For example, ziconotide, a selective N-type calcium channel blocker FDA approved in 2004, is restricted to intrathecal delivery, contraindicated in psychosis and associated with a high incidence of side effects, including neurological and psychiatric [177]. Several calcium channel blockers used for other indications have been withdrawn from the market or fallen out of use due to safety concerns such as mibefradil, pimozide and penfluridol. Several agents targeting calcium channel blockers aiming to overcome the limitations in drug delivery and safety are currently in Phase II clinical trials for various neuropathic pain states, including DS-5565 (a2d), CNV-2197944 (N-type) and ABT-639 (T-type). The only calcium channel modulating agent (Z-160) studied in lumbar radiculopathy failed to show efficacy in a Phase II trial [70]. Voltage-gated potassium channels Voltage-gated potassium channels are also involved in the generation of action potential in neurotransmission. Even more so than calcium channel blockers, development of a targeted treatment for pain is challenged by the ubiquity of potassium channels in multiple tissues. Furthermore, potassium channels are encoded by > 70 genes with a high degree of sequence similarity among subclasses of channels [178]. Drug--drug interactions also represent a major challenge to potassium channel analgesics since opioid receptor agonists open potassium channels and NMDA receptor antagonists close potassium channels; this may lead to or potentiate adverse effects such as sedation. Due to these challenges, development of clinically ready potassium channel modulators is still at a preclinical stage and is focused on selection and validation of pain-specific ion channels such as KCNQ, Kv2 and Kv9.1 [178]. 7.3.8.5

Nitrous oxide and PDEs Nitrous oxide released from damaged afferent nerve endings activates soluble guanylyl cyclase, catalyzing the formation of second messenger cGMP, resulting in the sensitization of nearby neurons and hyperalgesia [179]. This effect is terminated by cGMP-specific PDE hydrolysis of cGMP. Several animal studies have demonstrated clinically significant antinociceptive effects with local, systemic and intrathecal administration of PDE5 inhibitors [180,181]. These therapeutic effects have been observed in both nociceptive and neuropathic pain models. Rocha et al. demonstrate dose-dependent 7.3.9

Expert Opin. Emerging Drugs (2014) 20(1)

Expert opinion on emerging drugs: chronic low back pain

Expert Opin. Emerging Drugs Downloaded from informahealthcare.com by Selcuk Universitesi on 12/24/14 For personal use only.

analgesia in rodent OA models treated with tadalafil, suggesting possible benefit for facetogenic CLBP [181]. Huang et al. were able to show analgesic effects for intravenous sildenafil in models of neuropathic pain induced by spinal nerve ligation [182]. Two other studies found a synergistic effect between sildenafil and morphine for both oral and intrathecal administration [180,183]. Human studies demonstrating analgesic effects are lacking. Side effects of this drug class include hypotension, hypertension, angina, priapism and vision loss. To date, only a single Pfizer-sponsored Phase IV clinical trial has been conducted in the treatment of diabetic neuropathy pain in the mid-2000s, with no results reported. Gene therapy An ideal therapeutic would be one that inhibits neurotransmission at a functionally specific site such as peripheral neurons or spinal cord/DRG without off-target side effects. Viral vector-based gene therapy has demonstrated safety in one Phase I clinical trial for cancer pain [184]. Promising data from a Phase I study of a DNA-decoy drug AYX-1 for preventing post-surgical pain following total knee replacement offers the possibility of preventing the transition between acute and chronic post-surgical pain using gene therapy [185]. However, several following factors deserve consideration when designing a suitable therapy: i) therapeutic gene selection; ii) specificity of the viral vector to the appropriate target cells; and iii) vector production quality and cost. The current state of evidence for efficacy of various viral transduction methodologies is limited to animal studies [186]. In the short term, gene therapy requires further validation of efficacy and safety in CLBP. However, the ability to specifically target the mechanism (nociceptive, neuropathic, mixed) and tissue (disc) offers substantial promise. 7.4

Stem cell therapy The promising results of feasibility studies using stem cell therapy in treating neurodegenerative conditions such as amyotrophic lateral sclerosis and multiple sclerosis has spurred interest in using stem cell therapy to treat neuropathic pain [187,188]. The evidence for efficacy of this modality is still preclinical [189], but two human studies using allogeneic mesenchymal stem cell intradiscal transplants were recently submitted to the ClinicalTrials.gov registry [190,191]. We believe there is still significant uncertainty over using stem cell therapy for CLBP including safety and quality concerns such as immunogenicity, bacterial or viral contamination and misdirected or uncontrolled growth. However, over the long term, gene therapy offers unprecedented opportunities to move pain management from symptom mitigation to disease modification. 7.4.1

8.

Potential development issues

There are several challenges related to the development of novel analgesics for CLBP. A substantial number of clinical

trials exist for analgesic drugs that, even though they have previously shown efficacy, have yielded negative results [192-195]. One explanation for this phenomenon is that intrinsic characteristics of clinical trials can compromise their ability to reveal efficacy [196]. In this context, one objective is to increase the ‘ability to distinguish an effective from a less or ineffective treatment’, a concept known as assay sensitivity [197]. The Initiative on Methods, Measurement, and Pain Assessment in Clinical Trials has developed consensus guidelines to identify factors that influence assay sensitivity with the goal of reducing the rate of false-negative trials [196]. This report identified several areas of patient, study design, study site and outcome measurement factors that could affect the assay sensitivity and efficiency of chronic pain clinical studies such as psychological distress, which was associated with a higher placebo response and less treatment improvement in one clinical trial examining opioids in the treatment of LBP [198]. Another barrier to clinical development is adverse effects. Whereas reports of serious adverse events associated with the use of medications for LBP are sparse [199-201], many analgesic agents are centrally acting and common side effects such as sedation, dizziness and fatigue can lead to a high rate of withdrawal from studies [202,203]. In one study comparing topiramate with diphenhydramine for lumbar radiculopathy, topiramate was associated with more than twice the rate of withdrawal due to adverse events such as sedation and diarrhea [204]. This led the investigators to not recommend using a medication that was more efficacious than an active placebo. The cost of clinical trials in CLBP represents another barrier to the development of novel analgesic medications. In the case of clinical trials for LBP, the cost of conducting expensive trials is compounded by the poor predictive value of preclinical animal models, which increases the false-positive rate of drug candidates advanced to human testing [104,205,206]. Improving the efficiency of studies that advance into latestage testing is, therefore, a worthwhile endeavor [196]. 9.

Conclusions

It is important to note the importance of accounting for effect size and nonspecific effects that may confound study results. It is difficult to compare treatment studies in CLBP due to variability in the type and precision of pain outcome measures. For example, efficacy studies of novel analgesics for CLBP may use different measures of pain severity (Numeric Rating Scale, Visual Analog Scale, percentage decrease from baseline, etc.). Because each measure contains different scales, comparing the magnitude of an effect can be challenging. Computing an effect size is critical to prove that there is a difference between a study drug and comparator. Two well-accepted measures of effect size are commonly used, namely: i) the standardized mean difference, which is used for continuous measures such as a pain intensity rating scale; and ii) the number needed to treat,

Expert Opin. Emerging Drugs (2014) 20(1)

17

Expert Opin. Emerging Drugs Downloaded from informahealthcare.com by Selcuk Universitesi on 12/24/14 For personal use only.

E. Hsu et al.

which is used for binary outcomes such as responder versus nonresponder [207,208]. Interestingly, regardless of treatment type or measurement, improvement in LBP symptoms follows a distinct pattern. In a recent meta-analysis of both RCT and observational studies of patients with LBP, Artus et al. showed that patients experienced rapid improvement in the short term then smaller improvement in the intermediate and long term [209]. Drug discovery in CLBP must weigh the potential to create substantial value against the risk of causing individual and societal harm. CLBP is particularly challenging given the wide spectrum of different and often overlapping etiologies. Furthermore, the concept of LBP as a biopsychosocial phenomenon warrants repeating. Utilizing this definition helps to explain the limited long-term effects of all single modality treatments and interventions for LBP including pharmacotherapy and underscores the need to develop better physical, psychological and social interventions in addition to novel drug therapy. Challenges to developing better medicines specific to CLBP include: 1) heterogeneity of pain phenotypes within a given pain diagnostic category; 2) biological resiliency of multiple concurrent nociceptive pathways such that unimodal therapy is often inadequate in modulating established chronic pain conditions; 3) inability to identify pathophysiological disease mechanisms specific to an individual patient; 4) lack of consensus over outcome measures; 5) off-target and sometimes dangerous side effects; and 6) inconsistent treatment delivery approaches, which are often empirical rather than mechanism-based. [5]. Bibliography Papers of special note have been highlighted as either of interest () or of considerable interest () to readers. 1.

US Burden of Disease Collaborators. The state of US health, 1990-2010: burden of diseases, injuries, and risk factors. JAMA 2013;310:591-608

2.

Hoy D, Bain C, Williams G, et al. A systematic review of the global prevalence of low back pain. Arthritis Rheum 2012;64:2028-37

3.

Juniper M, Le T, Mladsi D. The epidemiology, economic burden, and pharmacological treatment of chronic low back pain in France, Germany, Italy, Spain and the UK: a literature-based review. Expert Opin Pharmacother 2009;10:2581-92

18

4.

5.

..

6.

10.

Expert opinion

Despite these challenges, the volume and diversity of novel molecular entities spanning chemical, biological, gene- and cell-based modalities reflect the level of interest and investment in this field. The paradigm of mechanism-based rather than empirical treatment underscores the need for drugs with diverse mechanisms of action. Genotype- or phenotype-specific therapy based on the results of multinational collaborative research such as the PROPANE study offers the prospect of targeted, individualized therapy [170]. Early stage efforts in gene- and stem cell-based therapies may ultimately lead to diseasemodifying therapy for CLBP rather than symptom alleviation alone. In the effort to improve collaboration around pain care, several national and international collaborations are ongoing, spanning topics from clinical trial design, patient-reported outcomes registries and more effective strategies to deliver and reimburse safe pain care [5].Continued collaborative efforts from multidisciplinary centers, and aligning research and clinical care around validated clinically appropriate outcome measures is critical in making progress toward identifying highvalue treatments and treatment plans for patients with CLBP.

Declaration of interest This paper was funded in part by the Centers for Rehabilitation Sciences Research, Uniformed Services University of the Health Sciences, Bethesda, MD, USA. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Crow W, Willis D. Estimating cost of care for patients with acute low back pain: a retrospective review of patient records. J Am Osteopath Assoc 2009;109:229-33 Pizzo PA, Clark NM, Carter-Pokras O, et al. Relieving pain in America. A blueprint for transforming prevention, care, education and research. Institute of Medicine. National Academies Press; 2011 The 2010 Patient Protection and Affordable Care Act required the Department of Health and Human Services to enlist the Institute of Medicine in examining pain as a public health problem; this report summarizes their findings. Cohen SP, Mao J. Mechanisms of neuropathic pain and their

Expert Opin. Emerging Drugs (2014) 20(1)

.

clinical implications. BMJ 2014;348:f7656 A recent overview explaining the rationale and importance of mechanism-based treatment over disease-based treatment.

7.

Freynhagen R, Baron R, Gockel U, et al. painDETECT: a new screening questionnaire to identify neuropathic components in patients with back pain. Curr Med Res Opin 2006;22:1911-20

8.

Waddell G. A new clinical model for the treatment of low-back pain. Spine (Phila Pa 1976) 1987;12(7):631

9.

Keller A, Hayden J, Bombardier C, van Tulder M. Effect sizes of nonsurgical treatments of non-specific lowback pain. Eur Spine J 2007;16:1776-88

Expert opinion on emerging drugs: chronic low back pain

10.

11.

Expert Opin. Emerging Drugs Downloaded from informahealthcare.com by Selcuk Universitesi on 12/24/14 For personal use only.

12.

13.

14.

.

15.

16.

17.

18.

.

19.

.

20.

Hassan A, Saleh H, Baroudy Y, et al. Prevalence of neuropathic pain among patients suffering from chronic low back pain in Saudi Arabia. Saudi Med J 2004;25:1986-90 Cohen SP, Jamison D, Bicket M, et al. Epidural steroids: a comprehensive, evidence-based review. Reg Anesth Pain Med 2013;38:175-200 Woolf CJ, Max MB. Mechanism-based pain diagnosis: issues for analgesic drug development. Anesthesiology 2001;95:241-9 Farrar JT. What is clinically meaningful: outcome measures in pain clinical trials. Clin J Pain 2000;16(2 Suppl):S106-12 Skljarevski V, Ossanna M, Liu-Seifert H, et al. A double-blind, randomized trial of duloxetine versus placebo in the management of chronic low back pain. Eur J Neurol 2009;16:1041-8 A clinical trial demonstrating efficacy of duloxetine in treatment of chronic low back pain (CLBP). Williams CM, Maher CG, Latimer J, et al. Efficacy of paracetamol for acute low-back pain: a double-blind, randomised controlled trial. Lancet 2014;384(9954):1586-96 Khoromi S, Cui L, Nackers L, et al. Morphine, nortriptyline and their combination vs. placebo in patients with chronic lumbar root pain. Pain 2007;130:66-75 Baron R, Freynhagen R, T€olle TR, et al. The efficacy and safety of pregabalin in the treatment of neuropathic pain associated with chronic lumbosacral radiculopathy. Pain 2010;150:420-7 Roelofs PD, Deyo RA, Koes BW, et al. Non-steroidal anti-inflammatory drugs for low back pain. Cochrane Database Syst Rev 2008;CD000396 A Cochrane systematic review showing small effect size for NSAIDs in the treatment of nociceptive CLBP. Deshpande A, Furlan A, Mailis-Gagnon A, et al. Opioids for chronic low-back pain. Cochrane Database Syst Rev 2007;CD004959 A Cochrane systematic review showing small effect size for opioids in the treatment of CLBP. Urquhart DM, Hoving JL, Assendelft WW, et al. Antidepressants for non-specific low back pain.

.

21.

22.

Cochrane Database Syst Rev 2008;CD001703 A Cochrane systematic review showing mixed efficacy of pain-relieving antidepressants for the treatment of CLBP. Toth PP, Urtis J. Commonly used muscle relaxant therapies for acute low back pain: a review of carisoprodol, cyclobenzaprine hydrochloride, and metaxalone. Clin Ther 2004;26:1355-67 Morlion B. Chronic low back pain: pharmacological, interventional and surgical strategies. Nat Rev Neurol 2013;9(8):462-73

23.

Freburger JK, Homes GM, Agans RP, et al. The rising prevalence of chronic low back pain. Arch Intern Med 2009;169(3):251-8

24.

Smith M, Davis MA, Stano M, Whedon JM. Aging baby boomers and the rising cost of chronic back pain: secular trend analysis of longitudinal medical expenditures panel survey data for years to 2000 to 2007. J Manipulative Physiol Ther 2013;36:2-11

from the American College of Physicians and the American Pain Society. 30.

Manchikanti L, Falco FJ, Singh V, et al. Utilization of interventional techniques in managing chronic pain in the Medicare population: analysis of growth patterns from 2000 to 2011. Pain Physician 2012;15:E969-82

31.

Deyo RA, Gray DT, Kreuter W, et al. United States trends in lumbar fusion surgery for degenerative conditions. Spine (Phila Pa 1976) 2005;30(12):1441-5

32.

Cohen SP, Chen Y, Neufeld NJ. Sacroiliac joint pain: a comprehensive review of epidemiology, diagnosis and treatment. Expert Rev Neurother 2013;13:99-116

33.

Cohen SP, Huang JH, Brummett C. Facet joint pain- advances in patient selection and management. Nat Rev Rheumatol 2013;9:101-16

34.

Jacobs WC, van Tulder M, Arts M, et al. Surgery versus conservative management of sciatica due to a lumbar herniated disc: a systematic review. Eur Spine J 2011;20:513-22

35.

Kovacs FM, Urr utia G, Alarco´n JD. Surgery versus conservative treatment for symptomatic lumbar spinal stenosis: a systematic review of randomized controlled trials. Spine (Phila Pa 1976) 2011;36:E1335-51

25.

Driscoll T, Jacklyn G, Orchard J, et al. The global burden of occupationally related low back pain: estimates from the Global Burden of Disease 2010 study. Ann Rheum Dis 2014;3(6):975-81

26.

Luo X, Pietrobon R, Hey L. Patterns and trends in opioid use among individuals with back pain in the united states. Spine 2004;29:884-91

36.

Caudill-Slosberg M, Schwartz L, Woloshin S. Office visits and analgesic prescriptions for musculoskeletal pain in US: 1980 vs 2000. Pain 2004;109(3):514-19

Brox JI, Nygaard ØP, Holm I, et al. Four-year follow-up of surgical versus non-surgical therapy for chronic low back pain. Ann Rheum Dis 2010;69:1643-8

37.

Paulozzi LJ, Jones CM, Mack KA, Rudd RA. Vital signs: overdose of prescription opioid pain relievers -United States, 1999 -- 2008. MMWR Morb Mortal Wkly Rep 2011;60:1487-92

Henschke N, Ostelo RW, van Tulder MW, et al. Behavioural treatment for chronic low-back pain. Cochrane Database Syst Rev 2010;7(7):CD002014

38.

van Tulder M, Malmivaara A, Esmail R, et al. Exercise therapy for low back pain: a systematic review within the framework of the Cochrane Collaboration Back Review Group. Spine 2000;25:2784-96

39.

Skjarevski V, Zhang S, Desaiah D, et al. Duloxetine versus placebo in patients with chronic low back pain: a 12-week, fixed-dose, randomized, double-blind trial. J Pain 2010;11(12):1282-90

40.

Cawston H, Davie A, Paget MA, et al. Efficacy of duloxetine versus alternative oral therapies: an indirect comparison of randomised clinical trials in chronic low

27.

28.

29.

..

Chou R, Huffman LH; American Pain Society; American College of Physicians. Medications for acute and chronic low back pain: a review of the evidence for an American Pain Society/American College of Physicians clinical practice guideline. Ann Intern Med; 2007;147(7):505-14 Comprehensive review of the evidence and guidelines on the use of pharmacotherapy for low back pain Expert Opin. Emerging Drugs (2014) 20(1)

19

E. Hsu et al.

41.

Expert Opin. Emerging Drugs Downloaded from informahealthcare.com by Selcuk Universitesi on 12/24/14 For personal use only.

42.

43.

44.

45.

Moore RA, Cai N, Skjarevski V, et al. Duloxetine use in chronic painful conditions -- individual patient data responder analysis. Eur J Pain 2014;18:67-75 Pain Management Therapeutics Market to 2017 -- Price Competition to Intensify Following Patent Expiries of Lyrica and Cymbalta. GBI Research, London, UK. Available from: http://www.gbiresearch. com/Report.aspx?ID=Pain-ManagementTherapeutics-Market-to-2017-PriceCompetition-to-Intensify-FollowingPatent-Expiries-of-Lyrica-and-Cymbalta& [Last accessed 1 October 2014] Pfizer Annual Report 2013. Pfizer, Inc., New York, NY. Available from: http:// www.pfizer.com/files/investors/ financial_reports/annual_reports/2013/ stories_fight.htm [Last accessed 1 October 2014] Eli Lilly and Company Annual Report 2013. Eli Lilly and Company. Indianapolis, IN. Available from: https:// investor.lilly.com/releasedetail.cfm? ReleaseID=822044 [Last accessed 1 October 2014]

61.

51.

Cohen KL, Harris S. Efficacy and safety of nonsteroidal anti-inflammatory drugs in the therapy of diabetic neuropathy. Arch Intern Med 1987;147:1442-4

Bloom BS. Cost of treating arthritis and NSAID-related gastrointestinal side-effects. Aliment Pharmacol Ther 1988;2(Suppl 1):131-8

62.

52.

Melnikova I. Pain market. Nat Rev Drug Discov 2010;9:589-90

53.

A Brief Overview of Risk Evaluation & Mitigation Strategies (REMS). FDA Review. FDA/Center for Drug Evaluation and Research, Washington, DC; 2012. Available from: http://www.fda. gov/downloads/AboutFDA/Transparency/ Basics/UCM328784.pdf [Last accessed 1 October 2014]

Chen YF, Jobanputra P, Barton P, et al. Cyclooxygenase-2 selective non-steroidal anti-inflammatory drugs (etodolac, meloxicam, celecoxib, rofecoxib, etoricoxib, valdecoxib and lumiracoxib) for osteoarthritis and rheumatoid arthritis: a systematic review and economic evaluation. Health Technol Assess 2008;12(11):1-278.iii

63.

Le Goff B, Berthelot JM, Maugars Y, et al. Alternative use of bisphosphonate therapy for rheumatic disease. Curr Pharm Des 2010;16(27):3045-52

64.

O’Connell NE, Wand BM, McAuley J, et al. Interventions for treating pain and disability in adults with complex regional pain syndrome. Cochrane Database Syst Rev 2011(11):CD009416

65.

Patte-Mensah C, Cherkaouia K, Boudard D, et al. Neurogenic pain and steroid synthesis in the spinal cord. J Mol Neurosci 2006;28(1):17-31

66.

Chen SC, Liu BC, Chen CW, et al. Intradermal pregnenolone sulfate attenuates capsaicin-induced nociception in rats. Biochem Biophys Res Commun 2006;349(2):626-33

67.

Pappagallo M, Breuer B, Lin HM, et al. A pilot trial of intravenous pamidronate for chronic low back pain. Pain 2014;1):108-17 Recent Phase II clinical trial demonstrating efficacy of intravenous pamidronate for nociceptive low back pain.

Deyo RA, Weinstein JN. Low back pain. N Engl J Med 2001;344(5):363-70

47.

Bennett MI, Smith BH, Torrance N, et al. The S-LANSS score for identifying pain of predominantly neuropathic origin: validation for use in clinical and postal research. J Pain 2005;6(3):149-58

48.

Cohen SP, Kapoor SG, Rathmell JP. Intravenous infusion tests have limited utility for selecting long-term drug therapy in patients with chronic pain: a systematic review. Anesthesiology 2009;111:416-31

49.

Clarke H, Bonin RP, Orser BA, et al. The prevention of chronic postsurgical pain using gabapentin and pregabalin: a combined systematic review and meta-analysis. Anesth Analg 2012;115:428-42

50.

20

54.

55.

56.

Actavis Confirms Generic Nucynta ER Patent Challenge. PR Newswire, New York, NY. Available from: http://www. prnewswire.com/news-releases/actavisconfirms-generic-nucynta-er-patentchallenge-217368391.html [Last accessed 1 October 2014]

46.

Vo T, Rice AS, Dworkin RH. Non-steroidal anti-inflammatory drugs

induced gastrointestinal damage. Eur J Rheumatol Inflamm 1993;13(1):33-40

for neuropathic pain: how do we explain continued widespread use? Pain 2009;143:169-71

back pain. Eur Spine J 2013;22(9):1996-2009

57.

58.

59.

60.

Sullivan MD, Howe CQ. Opioid therapy for chronic pain in the United States: promises and perils. Pain 2014;154(1):S94-100 Vokow ND, Frieden TR, Hyde PS, et al. Medication-assisted therapies -- tackling the opioid-overdose epidemic. N Engl J Med 2014;370:2063-6 Extended-Release and Long-Acting Opioid Analgesics - RISK EVALUATION AND MITIGATION STRATEGY (REMS). FDA Review. FDA/Center for Drug Evaluation and Research, Washington, DC; 2012. Available from: http://www.fda. gov/downloads/Drugs/DrugSafety/ InformationbyDrugClass/UCM348818. pdf [Last accessed 1 October 2014] Marco JL, Amariles P, Bosca´et B, et al. Risk factors associated with NSAIDinduced upper gastrointestinal bleeding resulting in hospital admissions: a crosssectional, retrospective, case series analysis in valencia, spain. Curr Ther Res Clin Exp 2007;68(2):107-19 Rahme E, Joseph L, Kong S, et al. Gastrointestinal health care resource use and costs associated with nonsteroidal antiinflammatory drugs versus acetaminophen: retrospective cohort study of an elderly population. Arthritis Rheum 2000;43:917-24 Johnson RE, Hornbrook MC, Hooker RS, et al. Analysis of the costs of NSAID-associated gastropathy Experience in a US health maintenance organisation. Pharmacoeconomics 1997;12(1):76-88 de Pouvourville G, Tasch RF. The economic consequences of NSAID-

Expert Opin. Emerging Drugs (2014) 20(1)

.

68.

Ohtori S, Akazawa T, Murata Y, et al. Risedronate decreases bone resorption and improves low back pain in postmenopausal osteoporosis patients without vertebral fractures. J Clin Neurosci 2010;17(2):209-13

69.

Zhu M, Liang R, Pan LH, et al. Zoledronate for metastatic bone disease and pain: a meta-analysis of randomized clinical trials. Pain Med 2013;14(2):257-64

70.

Gilron I, Dickenson A. Emerging drugs for neuropathic pain. Expert Opin Emerg Drugs 2014;1-13

Expert opinion on emerging drugs: chronic low back pain

71.

Matthews GL, Hunter DJ. Emerging drugs for osteoarthritis. Expert Opin Emerg Drugs 2011;16(3):479-91

72.

Cohen SP, Raja SN. Pathogenesis, Diagnosis, and Treatment of Lumbar Zygapophysial (Facet) Joint Pain. Anesthesiology 2007;106(3):591-614

73.

Expert Opin. Emerging Drugs Downloaded from informahealthcare.com by Selcuk Universitesi on 12/24/14 For personal use only.

74.

Siedel MF, Herguijuela M, Forkert R, et al. Nerve growth factor in rheumatic diseases. Semin Arthritis Rheum 2010;40(2):109-26 Hefti FF, Rosenthal A, Walicke PA, et al. Novel class of pain drugs based on antagonism of nerve growth factor. Trends Pharmacol Sci 2006;27(2):85-91

75.

Watson JJ, Allen SJ, Dawbarn DD, et al. Targeting nerve growth factor in pain. BioDrugs 2008;22(6):349-59

76.

Seidel MF, Wise BL, Lane NE. Nerve growth factor: update on science and therapy. Osteoarthritis Cartilage 2013;21(9):1223-8

77.

78.

Lewin GR, Lechner SG, Smith ES. Nerve growth factor and nociception: from experimental embryology to new analgesic therapy. Handb Exp Pharmacol 2014;220:251-82 Sanga P, Katz N, Polverejan E, et al. Efficacy, safety, and tolerability of fulranumab, an anti-nerve growth factor antibody, in the treatment of patients with moderate to severe osteoarthritis pain. Pain 2013;154(10):1910-19

79.

Katz N, Borenstein DG, Birbara C, et al. Efficacy and safety of tanezumab in the treatment of chronic low back pain. Pain 2011;152(10):2248-58

80.

Kivitz AJ, Gimbel JS, Bramson C, et al. Efficacy and safety of tanezumab versus naproxen in the treatment of chronic low back pain. Pain 2013;154(7):1009-21 Clinical trial showing efficacy of an anti-nerve growth factor therapy for the treatment of non-neuropathic LBP.

.

81.

82.

Pfizer Suspends Chronic Pain Studies In Tanezumab Clinical Trial Program; Some Studies Continue In Areas Of Unmet Medical Need. Pfizer Corporate Press Release, New York, 2010. Available from: http://pfizer. mediaroom.com/index.php? s=5149&item=11206 [Last accessed 1 October 2014] Eli Lilly and Company 2nd Qtr Result. Indianapolis, IN. 2014. Slide 14 Available from: http:// files.shareholder.com/downloads/LLY/

3119382919x0x770508/e97384ea-95d84661-81ae-3284a6ffec30/ Q2_2014_Slides.pdf [Last accessed 1 October 2014] 83.

Bannwarth B, Kostine M. Targeting nerve growth factor (NGF) for pain management: what does the future hold for NGF antagonists? Drugs 2014;74(6):619-26

84.

Mika J, Zychowska M, Popiolk-Barczyk K, et al. Importance of glial activation in neuropathic pain. Eur J Pharmacol 2013;716(1-3):106-19

85.

Hameed H, Hameed M, Christo PJ. The effect of morphine on glial cells as a potential therapeutic target for pharmacological development of analgesic drugs. Curr Pain Headache Rep 2010;96-104

86.

Rolan P. Methods for targeting glial activation in the treatment of patients with chronic pain. Presented at the Mechanisms and Treatment of Neuropathic Pain Annual Meeting; Snowbird, Utah, USA; 1-3 November 2007

87.

Olmarker K, Nutu M, Storkson R. Changes in spontaneous behavior in rats exposed to experimental disc herniation are blocked by selective TNF-alpha inhibition. Spine 2003;28:1635-42

88.

Olmarker K, Rydevik B. Selective inhibition of tumor necrosis factor-alpha prevents nucleus pulposus-induced thrombus formation, intraneural edema, and reduction of nerve conduction velocity. Possible implications for future pharmacologic treatment strategies of sciatica. Spine 2001;26:863-9

89.

90.

91.

.

Scott LJ. Etanercept: a review of its use in autoimmune inflammatory diseases. Drugs 2014;74:1379-410 Korhonen T, Karppinen J, Malmivaara A, et al. Efficacy of infliximab for disc herniation-induced sciatica: one-year follow-up. Spine 2004;29(19):2115-19 Korhonen T, Karppinen J, Paimela L, et al. The treatment of disc-herniation-induced sciatica with infliximab: one-year follow-up results of FIRST II, a randomized controlled trial. Spine 2006;31(24):2759-66 Clinical trial which failed to show efficacy of TNF-a inhibitors in the treatment of sciatica.

Expert Opin. Emerging Drugs (2014) 20(1)

92.

Genevay S, Atlas SJ, Katz JN. Variation in eligibility criteria from studies of radiculopathy due to a herniated disc and of neurogenic claudication due to lumbar spinal stenosis: a structured literature review. Arthritis Rheum 2010;35(7):803-11

93.

Okoro T, Tafazal S, Longworth S, et al. Tumor necrosis alpha-blocking agent (Etanercept): a triple blind randomized controlled trial of its use in treatment of sciatica. J Spin Disorder Tech 2010;23(1):74-7

94.

Cohen SP, White RL, Kurihara C, et al. Epidural steroids, etanercept, or saline in subacute sciatica: a multicenter, randomized trial. Ann Intern Med 2012;156(8):551-9

95.

Kume K, Amano S, Yamada S. The efficacy and safety of caudal epidural injection with the TNF-alpha antagonist, etanercept, in patients with disc-herniation-incduced sciatica. Results of a randomized, controlled, 1-month follow-up study. Ann Rheum Dis 2008;67(Suppl II):131

96.

Ohtori S, Miyagi M, Eguchi Y, et al. Epidural administration of spinal nerves with the tumor necrosis factor-alpha inhibitor, etanercept, compared with dexamethasone for treatment of sciatica in patients with lumbar spinal stenosis: a prospective randomized study. Spine 2012;37(6):439-44

97.

Freeman BJ, Ludbrook GL, Hall S, et al. Randomized, double-blind, placebocontrolled, trial of transforaminal epidural etanercept for the treatment of symptomatic lumbar disc herniation. Spine 2013;38(23):1986-94

98.

Marks DM, Shah MJ, Patkar AA, et al. Serotonin-Norepinephrine Reuptake Inhibitors for Pain Control: premise and Promise. Curr Neuropharmacol 2009;7(4):331-6

99.

Verdu B, Decosterd I, Buclin T, et al. Antidepressants for the treatment of chronic pain. Drugs 2008;68(18):2611-32

100. Basile AS, Janowsky A, Golembiowska K, et al. Characterization of the antinociceptive actions of bicifadine in models of acute, persistent and chronic pain. J Pharmacol Exp Ther 2007;321:1208-25 101. Riff D, Huang N, Czobor P, et al. A five-day, multi-center, randomized, placebo-controlled, double-blind, efficacy

21

E. Hsu et al.

and safety study of bicifadine and tramadol versus placebo in the treatment of postoperative bunionectomy pain. J Pain 2005;7(2):S40 102. Krieter PA, et al. Pharmacokinetics, disposition, and metabolism of bicifadine in humans. Drug Metab Dispos 2007;36:252-9

Expert Opin. Emerging Drugs Downloaded from informahealthcare.com by Selcuk Universitesi on 12/24/14 For personal use only.

103. Czobor P, Skolnick P. The secrets of a successful clinical trial: compliance, compliance, compliance. Mol Interv 2011;11(2):107-10 104. Gilron I. Drug discovery for neuropathic pain. In: Simpson DM, McArthur JC, Dworkin RH, editors. Neuropathic pain: mechanisms, diagnosis, and treatment. Oxford University Press; NY, USA: 2012. p. 38-57 105. Nicotra L, Loram LC, Watkins LR, et al. Toll-like receptors in chronic pain. Exp Neurol 2012;234(2):316-29 106. Liu T, Gao Y, Ji R. Emerging role of Toll-like receptors in the control of pain and itch. Neurosci Bull 2012;28(2):131-44 107. Connolly DJ, O’Neill LA. New developments in Toll-like receptor targeted therapeutics. Curr Opin Pharmacol 2012;12(4):510-18 108. Pasternak GW. Opiate pharmacology and relief of pain. J Clin Oncol 2014;32(16):1655-61 109. Przewłocki R, Przewłocka B. Opioids in chronic pain. Eur J Pharmacol 2001;429(1):79-91 110. Steiner DJ, Sitar S, Wen W, et al. Efficacy and safety of the seven-day buprenorphine transdermal system in opioid-naı¨ve patients with moderate to severe chronic low back pain: an enriched, randomized, double-blind, placebo-controlled study. J Pain Symptom Manage 2011;42:903-17 111. Yarlas A, Miller K, Wen W, et al. A randomized, placebo-controlled study of the impact of the 7-day buprenorphine transdermal system on health-related quality of life in opioidnaı¨ve patients with moderate-to-severe chronic low back pain. J Pain 2013;14:14-23 . A clinical trial demonstrating efficacy of buprenorphine in the treatment of CLBP.

22

112. Kamibayashi T, Maze M. Clinical uses of alpha2-adrenergic agonists. Anesthesiology 2000;93(5):1345-9 113. Szallasi A, Blumberg PM. Vanilloid (Capsaicin) receptors and mechanisms. Pharmacol Rev 1999;51:159-212 114. Szallasi A, Blumberg PM. [3H] resiniferatoxin binding by the vanilloid receptor species-related differences, effects of temperature and sulhydryl reagents. Naunyn Schmiedebergs Arch Pharmacol 1993;347:84-91 115. Caterina MJ, Schumacher MA, Tominaga M, et al. The capsaicin receptor: a heat-activated ion channel in the pain pathway. Nature 1997;389:816-24 116. Hayes P, Meadows HJ, Gunthorpe MJ, et al. Cloning and functional expression of a human orthologue of rat vanilloid receptor-1. Pain 2000;88:205-15 117. Steenland HW, Ko SW, Wu LJ, et al. Hot receptors in the brain. Mol Pain 2006;2:34 118. Cortright DN, Szallasi A. Biochemical pharmacology of the vanilloid receptor TPRV1. An update. Eur J Biochem 2004;271:1814-19 119. Gunthorpe MJ, Benham CD, Randall A, et al. The diversity in the vanilloid (TRPV) receptor family of ion channels. Trends Pharmacol Sci 2002;23:183-91 120. Starowicz K, Nigam S, Di Marzo V, et al. Biochemistry and pharmacology of endovanilloids. Pharmacol Ther 2007;114:13-33 121. Holzer P. TRPV1 and the gut: from a tasty receptor for a painful vanilloid to a key player in hyperalgesia. Eur J Pharmacol 2004;500:231-4 122. Ji RR, Samad TA, Jin SX, et al. p38 MAPK activation by NGF in primary sensory neurons after inflammation increases TRPV1 levels and maintains heat hyperalgesia. Neuron 2002;36:57-68 123. Keeble J, Russell F, Curtis B, et al. Involvement of transient receptor potential vanilloid 1 in the vascular and hyperalgesic components of joint inflammation. Arthritis Rheum 2005;52:3248-56 124. Szolcsanyi J. Forty years in capsaicin research for sensory pharmacology and physiology. Neuropeptides 2004;32:377-84

Expert Opin. Emerging Drugs (2014) 20(1)

125. Caterina MJ. Transient receptor potential ion channels as participants in thermosensation and thermoregulation. Am J Physiol Regul Integr Comp Physiol 2000;292:R64-76 126. Davis JB, Gray J, Gunthorpe MJ, et al. Vanilloid receptor-1 is essential for inflammatory thermal hyperalgesia. Nature 2000;405:183-7 127. Menendez L, Juarez L, Garcia E, et al. Analgesic effects of capasazepine and resiniferatoxin on bone cancer pain in mice. Neurosci Lett 2006;393:70-3 128. Wong GY, Gavva NR. Therapeutic potential of vanilloid receptor TRPV1 agonists and antagonists as analgesics: recent advances and setbacks. Brain Res Rev 2009;60(1):267-77 129. Knotkova H, Pappagallo M, Szallasi A, et al. Capsaicin (TRPV1 Agonist) therapy for pain relief: farewell or revival? Clin J Pain 2008;24:142-54 130. Cantillon M, Vause E, Sykes D, et al. Safety, tolerability and efficacy of intraoperative ALGRX 4975 in a randomized, double-blind, placebocontrolled study of subjects undergoing bunionectomy. J Pain 2005;6:S48 131. Diamond E, Richards P, Miller T. ALGRX 4975 reduces pain of intermetatarsal neuroma: preliminary results from a randomized, double-blind, placebo-controlled, phase II multicenter clinical trial. J Pain 2006;7:S41 132. Richards P, Vasko G, Stasko I, et al. ALGRX 4975 reduces pain of acute lateral epicondylitis: preliminary results from a randomized, double-blind, placebo-controlled, phase II multicenter clinical trial. J Pain 2006;7:S3 133. Cantillon M, Vause E, Sykes D, et al. Safety, tolerability and efficacy of ALGRX 4975 in Osteoarthritis (OA) of the knee. J Pain 2005;6:S39 134. Cantillon M, Vause E, Sykes D, et al. Preliminary safety, tolerability and efficacy of ALGRX 4975 in osteoarthritis (OA) of the knee. J Pain 2005;6:S39 135. Chen TY, Corcos J, Camel M, et al. Prospective, randomized, double-blind study of safety and tolerability of intravesical resiniferatoxin (RTX) in interstitial cystitis (IC). Int Urogynecol J Pelvic Floor Dysfunct 2005;16:293-7

Expert opinion on emerging drugs: chronic low back pain

136.

Expert Opin. Emerging Drugs Downloaded from informahealthcare.com by Selcuk Universitesi on 12/24/14 For personal use only.

137.

138.

139.

140.

141.

142.

143.

144.

145.

Payne CK, Mosbaugh PG, Forrest JB, et al. Intravesical resiniferatoxin for the treatment of interstitial cystitis: a randomized, double-blind, placebo controlled trial. J Urol 2005;173:1590-4 Honore P, Wismer CT, Mikusa J, et al. A-425619 [1-isoquinolin-5-yl-3-(trifluromethyl-benzyl)-urea], a novel transient receptor potential receptor type V1 receptor antagonist, relieves pathophysiological pain associated with inflammation and tissue injury in rats. J Pharmacol Exp Ther 2005;314:410-21 Gavva NR, Tamir R, Qu Y, et al. AMG 9810 [(E)-3-(4-t-butylphenyl)-N-(2,3dihydrobenzo[b][1,4] dioxin-6-yl) acrylamide], a novel vanilloid receptor (TPRV1) antagonist with antihyperalgesic properties. J Pharmacol Exp Ther 2005;313:474-84 Rami HK, Thompson M, Stemp G, et al. Discovery of SB-705498: a potent, selective, orally bioavailable TRPV1 antagonist suitable for clinical development. Bioorg Med Chem Lett 2006;16:3287-91 Walker KM, Urban L, Medhurst SJ, et al. The VR1 antagonist capsazepine reverses mechanical hyperalgesia in models of inflammatory and neuropathic pain. J Pharmacol Exp Ther 2003;304:56-62 Pomonis JD, Harrison JE, Mark L, et al. N-(4-tertiarybutylphenyl)-4-(3chlorophyridin-2-yl)ttrahydropyrazine-1 (2H)-carbox-amide (BCTC), a novel, orally effective vanilloid receptor 1 antagonist with analgesic properties: II. In vivo characterization in rat models of inflammatory and neuropathic pain. J Pharmacol Exp Ther 2003;306:387-93

data from 3 phase 1 trials. J Clin Pharmacol 2012;52(7):1028-41 146.

147.

148.

Rowbotham MC, Nothaft W, Duan WR, et al. Oral and cutaneous thermosensory profile of selective TRPV1 inhibition by ABT-102 in a randomized healthy volunteer trial. Pain 2011;152(2):1192-200 Nash MS, McIntyre P, Groarke A, et al. BCTP, a classic polymodal inhibitor of TRPV1 with a reduced liability for hyperthermia, is analgesic and ameliorates visceral hypersensitivity. J Pharmacol Exp Ther 2012;342(2):389-98 Watabiki T, Kiso T, Kuramochi T, et al. Amerljoiration of neuropathic pain by novel transient receptor potential vanilloid 1 antagonist AS1928370 in rats without hyperthermic effect. J Pharmacol Exp Ther 2011;336(3):743-50

149.

Benzon H, Raja SN, Fishman SM, et al. Essentials of pain medicine. Elsevier; Philadelphia: 2011

150.

Jeftinija S, Jeftnija K, Liu F, et al. Excitatory amino acids are released from rat primary afferent neurons in vitro. Neurosci Lett 1991;125:191-4

151.

Cumberbatch MJ, Chizh BA, Headley PM. AMPA receptors have an equal role in spinal nociceptive and nonnociceptive transmission. Neuroreport 1994;5:877-80

152.

King AE, Lopez-Garcia JA. Excitatory amino acide receptor mediated neurotransmission from cutaneous afferents in rat dorsal horn in vitro. J Physiol 1993;472:443-57

153.

Gunthrope MJ, Chizh BA. Clinical development of TRPV1 antagonists: targeting a pivotal point in the pain pathway. Drug Discov Today 2009;14:56-67

Chizh BA, Headley PM. NMDA antagonists and neuropathic pain - multiple drug targets and multiple uses. Curr Pharm Des 2005;11(23):2977-94

154.

Trevisani M, Gatti R. TRPV1 antagonists as analgesic agents. Open Pain J 2013;6(Suppl 1):108-18

Laube B, Kuhse J, Betz H. Evidence for a tetrameric structure of recombinant NMDA receptors. J Neurosci 1998;18:2954-61

155.

Gavva NR, Treanor JJ, Garami A, et al. Pharmacological blockade of the vanilloid receptor TRPV1 elicits marked hyperthermia in humans. Pain 2008;136:202-10

Childers WE, Reinhardt BB. N-methyld-aspartate antagonists and neuropathic pain: the search for relief. J Med Chem 2007;50(11):2557-62

156.

Jones MW, McClean M, Parsons CG, et al. The in vivo relevance of the varied channel-blocking properties of uncompetitive NMDA antagonists: tests on spinal neurons. Neuropharmacology 2001;41:50-61

Othman AA, Nothaft W, Awni WM, et al. Pharmacokinetics of the TRPV1 antagonist ABT-102 in healthy human volunteers: population analysis of

Expert Opin. Emerging Drugs (2014) 20(1)

157. McQuay HJ, Carroll D, Jadad AR, et al. Dextromethorphan for the treatment of neuropathic pain: a double-blind randomized controlled crossover trial with integral n-of-1 design. Pain 1994;59:127-33 158. Santangelo RM, Acker TM, Zimmerman SS, et al. Novel NMDA receptor modulators: an update. Expert Opin Ther Patents 2012;22(11):1337-52 159. Nikolajsen L, Gotrup H, Kristensen AG, et al. Memantine (a N-methyl-Daspartate receptor antagonist) in the treatment of neuropathic pain after amputation or surgery: a randomized double-blinded, cross-over study. Anesth Analg 2000;91:960-6 160. Sang CN, Booher S, Gilron I, et al. Dextromethorphan and memantine in painful diabetic neuropathy and postherpetic neuralgia: efficacy and doseresponse trials. Anesthesiology 2002;96:1053-61 161. Nelson KA, Park KM, Robinovitz E, et al. High-dose oral dextromethorphan versus placebo in painful diabetic neuropathy and postherpetic neuralgia. Neurology 1997;48:1212-18 162. Parsons CG. NMDA receptors as targets for drug action in neuropathic pain. Eur J Pharmacol 2001;429:71-8 163. Chizh BA, Headley PM, Tzschentke TM. NMDA receptor antagonists as analgesics: focus on the NR2B subtype. Trends Pharmacol Sci 2001;22:636-42 164. Boyce S, Wyatt A, Webb JK, et al. Selective NMDA NR2B antagonists induce antinociception without motor dysfunction: correlation with restricted localization of NR2B subunit in dorsal horn. Neuropharmacology 1999;38:611-38 165. Sang CN, Weaver JJ, Jinga L, et al. The NR2B subunit-selective NMDA receptor antagonist, CP-101-606, reduces spontaneous pain intensity in patients with central and peripheral neuropathic pain. Society of Neuroscience Abstract; 2003. 814.9 166. Danysz W, Parsons AC. Glycine and Nmethyl-D-aspartate receptors: physiological significance and possible therapeutic applications. Pharmacol Rev 1998;50:597-664 167. Wallace MS, Rowbotham MC, Katz NP, et al. A randomized, double-blind,

23

E. Hsu et al.

placebo controlled trial of a glycine antagonist in neuropathic pain. Neurology 2002;59:1694-700 168. Sulayman DD, Yang Y, Waxman J. The NaV1.7 sodium channel: from molecule to man. Nat Rev Neurosci 2013;14:49-62

Expert Opin. Emerging Drugs Downloaded from informahealthcare.com by Selcuk Universitesi on 12/24/14 For personal use only.

169. Fischer TZ, Gilmore ES, Estacion M, et al. A novel Nav1.7 mutation producing carbamazepine-responsive erythromelalgia. Ann Neurol 2009;65:733-41 170. PROPANE STUDY: Probing the Role of Sodium Channels in Painful Neuropathies Brussels, Belgium: European Commission Research & Innovation. 2014. Available from: http://ec.europa. eu/research/health/medical-research/brainresearch/projects/propane-study_en.html [Last accessed 1 October 2014] 171. Hughes JP, Chessell I, Malamut R, et al. Understanding chronic inflammatory and neuropathic pain. Ann NY Acad Sci 2012;1255:30-44 . An international prospective cohort study that aims to enable genomically targeted pain pharmacotherapies. 172. McCleane G. Does gabapentin have an analgesic effect on background movement and referred pain? A randomised, doubleblind, placebo controlled study. Pain Clinic 2001;13:103-7 173. Yildirim K, Sisecioglu M, Karatay S, et al. The effectiveness of gabapentin in patients with chronic radiculopathy. Pain Clinic 2003;15:213-18 174. Romano CL, et al. Pregabalin, celecoxib, and their combination for treatment of chronic low-back pain. J Orthop Traumatol 2009;10(4):185-91 175. Baron R, Kern U, Mu¨ller M, et al. Effectiveness and tolerability of a moderate dose of tapentadol prolonged release for managing severe, chronic low back pain with a neuropathic component: an open-label continuation arm of a randomized phase 3b study. Pain Pract 2014; Epub ahead of print . A clinical trial that found no difference between tapentadol and tapentadol with gabapentin for mixed neuropathic/nociceptive CLBP. 176. Yaksh T. Calcium channels as therapeutic targets in neuropathic pain. J Pain 2006;7(1):S13-30

24

177. Maier C, Gockel HH, Gruhn K, et al. Increased risk of suicide under intrathecal ziconotide tredatment? -- A warning". Pain 2010;152(1):235-7 178. Waxman SG, Zamponi GW. Regulating excitability of peripheral afferents: emerging ion channel targets. Nat Neurosci 2014;17(2):153-63 179. Aley KO, McCarter G, Levine JD, et al. Nitric oxide signaling in pain and nociceptor sensitization in the rat. J Neurosci 1998;18:7008-14 180. Jain NK, Patil CS, Singh A, et al. Sildenafil, a phosphodiesterase-5 inhibitor, enhances the antinociceptive effect of morphine. Pharmacology 2003;67(3):150-6 181. Rocha FA, Silva FS Jr, Leite AC, et al. Tadalafil analgesia in experimental arthritis involves suppression of intraarticular TNF release. Br J Pharmacol 2011;164(2b):828-35 182. Huang LJ, Yoon MH, Choi JI, et al. Effect of sildenafil on neuropathic pain and hemodynamics in rats. Yonsei Med J 2010;51(1):82-7 183. Yoon MH, Park KD, Lee HG, et al. Additive antinociception between intrathecal sildenafil and morphine in the rat formalin test. J Korean Med Sci 2008;23(6):1033-8 184. Fink DJ, Wechuck J, Mata M, et al. Gene therapy for pain: results of a phase I clinical trial. Ann Neurol 2011;70:207-12 185. Mamet J, Klukinov M, Yaksh T, et al. Single intrathecal administration of the transcription factor decoy AYX1 prevents acute and chronic pain after incisional, inflammatory, or neuropathic injury. Pain 2014;155(2):322-33 186. Hao S, Mata M, Glorioso JC, et al. Gene transfer to interfere with TNFalpha signaling in neuropathic pain. Gene Ther 2007;14:1010-16 187. Ebert AD, Yu J, Rose FF Jr, et al. Induced pluripotent stem cells from a spinal muscular atrophy patient. Nature 2008;457(7227):277-80 188. Karussis D, Karageorgiou C, Vaknin-Dembinsky A, et al. Safety and immunological effects of mesenchymal stem cell transplantation in patients with multiple sclerosis and amyotrophic lateral sclerosis. Arch Neurol 2010;67(10):1187-94

Expert Opin. Emerging Drugs (2014) 20(1)

189. Braz JM, Sharif-Naeini R, Vogt D, et al. Forebrain GABAergic neuron precursors integrate into adult spinal cord and reduce injury-induced neuropathic pain. Neuron 2012;74(4):663-75 190. Treatment of Degenerative Disc Disease With Allogenic Mesenchymal Stem Cells (MSV) (Disc_allo). National Institutes of Health/ClinicalTrials.gov, Washington, DC; 2014. Available from: http://clinicaltrials. gov/ct2/show/NCT01860417?term=stem +cell+AND+pain&no_unk=Y&rank=2 [Last accessed 1 October 2014] 191. Safety and Preliminary Efficacy Study of Mesenchymal Precursor Cells (MPCs) in Subjects With Lumbar Back Pain. National Institutes of Health/ ClinicalTrials.gov, Washington, DC; 2014. Available from: http://clinicaltrials. gov/ct2/show/NCT01290367?term=stem +cell+AND+pain&no_unk=Y&rank=3 [Last accessed 1 October 2014] 192. Dworkin RH, Thakur R, Griesing T, et al. Randomized clinical trials of pregabalin for neuropathic pain: methods, results, and implications. Prog Neurother Neuropsychopharm 2008;3:167-87 193. Katz N. Methodological issues in clinical trials of opioids for chronic pain. Neurology 2005;65:S32-49 194. T€olle T, Freynhagen R, Versavel M, et al. Pregabalin for relief of neuropathic pain associated with diabetic neuropathy: a randomized, double-blind study. Eur J Pain 2008;12:203-13 195. XenoPort, Inc. Phase II results for GSK1838262 (XP13512) reported for neuropathic pain associated with diabetic peripheral neuropathy. 2009. Available from: http://phx. corporate-ir.net/phoenix.zhtml? c=187883&p=irolnewsArticle&ID=1280577&highlight= 196. Dworkin RH, Turk DC, Peirce-Sandner S, et al. Considerations for improving assay sensitivity in chronic pain clinical trials: IMMPACT recommendations. Pain 2012;153:1148-58 . This study describes an international effort to improve assay sensitivity in the design of clinical trials for pain therapeutics. 197. International Conference on Harmonisation. E10: Choice of control groups and related issues in clinical trials.

Expert opinion on emerging drugs: chronic low back pain

2001. Available from: http://www.fda. gov.ezproxy.welch.jhmi.edu/downloads/ RegulatoryInformation/Guidances/ ucm125912.pdf [Accessed 23 August 2014] 198.

Expert Opin. Emerging Drugs Downloaded from informahealthcare.com by Selcuk Universitesi on 12/24/14 For personal use only.

199.

200.

201.

202.

203.

Wasan AD, Davar G, Jamison R. The association between negative affect and opioid analgesia in patients with discogenic low back pain. Pain 2005;117:450-61 Kearney PM, Baigent C, Godwin J, et al. Do selective cyclo-oxygenase-2 inhibitors and traditional non-steroidal antiinflammatory drugs increase the risk of atherothrombosis? Meta-analysis of randomised trials. BMJ 2006;332:1302-8 Moore RA, Derry S, Makinson GT, et al. Tolerability and adverse events in clinical trials of celecoxib in osteoarthritis and rheumatoid arthritis: systematic review and meta-analysis of information from company clinical trial reports. Arthritis Res Ther 2005;7:R644-65 Silverstein FE, Faich G, Goldstein JL, et al. Gastrointestinal toxicity with celecoxib vs nonsteroidal antiinflammatory drugs for osteoarthritis and rheumatoid arthritis: the CLASS study: a randomized controlled trial. Celecoxib Long-term Arthritis Safety Study. JAMA 2000;284:1247-5 Chou R, Peterson K, Helfand M. Comparative efficacy and safety of skeletal muscle relaxants for spasticity and musculoskeletal conditions: a systematic review. J Pain Symptom Manage 2004;28:140-75 Salerno SM, Browning R, Jackson JL. The effect of antidepressant treatment on chronic back pain: a meta-analysis. Arch Intern Med 2002;162:19-24

204.

Khoromi S, Patsalides A, Parada S, et al. Topiramate in chronic lumbar radicular pain. J Pain 2005;6:829-36

205.

Roy A. How the FDA Stifles New Cures, Part II: 90% of Clinical Trial Costs are Incurred in Phase III. Available from: http://www.forbes.com/sites/ theapothecary/2012/04/25/how-the-fda-

stifles-new-cures-part-ii-90-of-clinicaltrial-costs-are-incurred-in-phase-iii/ Last accessed 1 October 2014] 206.

Woolf CJ. Overcoming obstacles to developing new analgesics. Nat Med 2010;16(11):1241-7

207.

Faraone SV, Biederman J, Spencer TJ, Wilens TE. The drug-placebo response curve: a new method for assessing drug effects in clinical trials. J Clin Psychopharmacol 2000;20:673-9. Abstract

208.

Cohen J. Statistical power analysis for the behavioral sciences. 2nd edition. Erlbaum; Hillsdale, NJ: 1988

209.

Artus M, van der Windt D, Jordan KP, et al. The clinical course of low back pain: a meta-analysis comparing outcomes in randomised clinical trials (RCTs) and observational studies. BMC Musculoskelet Disord 2014;15(68):1-16

oxycodone DETERx: an extendedrelease, abuse-deterrent formulation. J Opioid Manag 2014;10(4):233-46 216. Available from: http://www. collegiumpharma.com/productspipeline 217. Colucci SV, Perrino PJ, Shram M, et al. Abuse potential of intravenous oxycodone/naloxone solution in nondependent recreational drug users. Clin Drug Investig 2014;34(6):421-9 218. Available from: http://www.medscape. com/viewarticle/824882 219. Available from: http://www.pfizer.com/ news/press-release/press-releasedetail/ alo_02_demonstrates_significant_ difference_in_pain_scores_in_chronic_ low_back_pain_patients_and_lower_ abuse_potential_compared_to_ immediate_release_oxycodone_ in_recreational_opioid_users 220. Available from: http://qrxpharma.com/go. cfm?do=Page.View&pid=4

210.

Available from: http://www.sec.gov/ Archives/edgar/data/1588972/ 000119312513409707/d605505ds1.htm

221. Available from: http://www.teikokuusa. com/research.html

211.

Available from: http://phx.corporate-ir. net/phoenix.zhtml?c=121590&p=irolnewsArticle&ID=1549072&highlight

Affiliation

212.

Company presentation, Durect. 2014. Slide 39 Available from: http:// phx.corporate-ir.net/External.File? item=UGFyZW50SUQ9MTM0MTA2fENoaWxkSUQ9LTF8VHlwZT0z&t=1

213.

Available from: http://phx.corporate-ir. net/phoenix.zhtml?c=251847&p=irolnewsArticle&ID=1963337&highlight= mnk-155

214.

Available from: http://www. collegiumpharma.com/press-releases/128august-6-2014-collegium-pharmaceuticalannounces-positive-top-line-data-fromphase-iii-study-of-oxycodone-deterxr-itsabuse-deterrent-extended-release-productfor-chronic-pain

215.

Kopecky EA, Fleming AB, Noonan PK, et al. Impact of physical manipulation on in vitro and in vivo release profiles of

Expert Opin. Emerging Drugs (2014) 20(1)

Eugene Hsu1 MD MBA, Sunberri Murphy2 DO, David Chang2 DO & Steven P Cohen†3 MD † Author for correspondence 1 Fellow in Anesthesiology and Perioperative Medicine, Oregon Health and Science University School of Medicine, Portland, OR, USA 2 Resident in Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA 3 Professor of Anesthesiology and Physical Medicine and Rehabilitation, Johns Hopkins School of Medicine and Uniformed Services University of the Health Sciences, 550 North Broadway, Suite 301, Baltimore, MD 21205, USA Tel: +1 410 955 1822; Fax: +1 410 614 7592; E-mail: [email protected]

25

Expert opinion on emerging drugs: chronic low back pain.

It is difficult to overestimate the personal and socioeconomic impact of chronic low back pain (CLBP). It is the leading cause of years lost to disabi...
714KB Sizes 0 Downloads 10 Views