Expert Consensus Open Access
Copyright ©The Author(s) 2021. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Clin Cases. Mar 26, 2021; 9(9): 2110-2122
Published online Mar 26, 2021. doi: 10.12998/wjcc.v9.i9.2110
Expert consensus of the Chinese Association for the Study of Pain on pain treatment with the transdermal patch
Ke Ma, Department of Algology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
Wei Jiang, Department of Anesthesiology, Third Medical Center of People’s Liberation Army General Hospital, Beijing 100039, China
Yun-Xia Wang, Department of Algology, The Third People’s Hospital of Hubei Province, Hubei Zhongshan Hospital, Wuhan 430033, Hubei Province, China
Lin Wang, Department of Algology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, China
Yan Lv, Department of Algology, Xijing Hospital, Air Force Medical University, Xi'an 710032, Shaanxi Province, China
Jin-Feng Liu, Department of Algology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
Rong-Guo Liu, Department of Algology, Fujian Provincial Hospital, Fuzhou 350001, Fujian Province, China
Hui Liu, Department of Algology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
Li-Zu Xiao, Department of Algology, Shenzhen Sixth People’s Hospital (Nanshan Hospital), Shenzhen 518000, Guangdong Province, China
Dong-Ping Du, Department of Algology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiaotong University, Shanghai 200233, China
Li-Juan Lu, Department of Algology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu Province, China
Xiao-Qiu Yang, Department of Algology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
Ling-Jie Xia, Department of Algology, Henan Provincial People’s Hospital, Zhengzhou 450000, Henan Province, China
Dong Huang, Department of Algology, The Third Xiangya Hospital of Central South University, Changsha 410013, Hunan Province, China
Zhi-Jian Fu, Department of Algology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, Shandong Province, China
Bao-Gan Peng, Department of Orthopedics, The Third Medical Center, General Hospital of the Chinese People’s Liberation Army, Beijing 100039, China
Yan-Qing Liu, Department of Algology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
ORCID number: Ke Ma (0000-0002-5603-9321); Wei Jiang (0000-0002-0680-2456); Yun-Xia Wang (0000-0003-2994-8471); Lin Wang (0000-0001-5438-0932); Yan Lv (0000-0002-2868-2352); Jin-Feng Liu (0000-0002-3459-3962); Rong-Guo Liu (0000-0001-5186-3541); Hui Liu (0000-0002-9599-6853); Li-Zu Xiao (0000-0001-8395-6072); Dong-Ping Du (0000-0002-4730-594X); Li-Juan Lu (0000-0002-6571-8529); Xiao-Qiu Yang (0000-0003-3108-1029); Ling-Jie Xia (0000-0002-4162-1036); Dong Huang (0000-0002-4880-2438); Zhi-Jian Fu (0000-0002-9387-9768); Bao-Gan Peng (0000-0002-7767-9424); Yan-Qing Liu (0000-0002-7347-0789).
Author contributions: All authors contributed equally to this work; Liu YQ and Ma K are considered as the co-corresponding authors.
Conflict-of-interest statement: The authors declare no conflict of interests for this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Ke Ma, PhD, Chief Doctor, Department of Algology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Yangpu District, Shanghai 200092, China. marke72@163.com
Received: November 30, 2020
Peer-review started: November 30, 2020
First decision: January 29, 2021
Revised: February 14, 2021
Accepted: March 9, 2021
Article in press: March 9, 2021
Published online: March 26, 2021

Abstract

Chronic pain lasting more than 3 mo, or even several years can lead to disability. Treating chronic pain safely and effectively is a critical challenge faced by clinicians. Because administration of analgesics through oral, intravenous or intramuscular routes is not satisfactory, research toward percutaneous delivery has gained interest. The transdermal patch is one such percutaneous delivery system that can deliver drugs through the skin and capillaries at a certain rate to achieve a systemic or local therapeutic effect in the affected area. It has many advantages including ease of administration and hepatic first pass metabolism avoidance as well as controlling drug delivery, which reduces the dose frequency and side effects. If not required, then the patch can be removed from the skin immediately. The scopolamine patch was the first transdermal patch to be approved for the treatment of motion sickness by the Food and Drug Administration in 1979. From then on, the transdermal patch has been widely used to treat many diseases. To date, no guidelines or consensus are available on the use of analgesic drugs through transdermal delivery. The pain branch of the Chinese Medical Association, after meeting and discussing with experts and based on clinical evidence, developed a consensus for promoting and regulating standard use of transdermal patches containing analgesic drugs.

Key Words: Transdermal drug delivery systems, Pain, Transdermal patches, Topical, Nonsteroidal anti-inflammatory drugs, Analgesics

Core Tip: Currently no international guidelines or consensus are available on the treatment of pain using transdermal patches. With the help of experts and based on recent clinical evidence, the pain branch of the Chinese Medical Association formulated China’s expert consensus on “transdermal pain treatment in China,” while considering China’s national conditions with a view to regulate and promote the standardized use of the transdermal patch containing analgesic drugs.



INTRODUCTION

According to the International Association for the Study on Pain, “Pain is an unpleasant sensory and emotional experience associated with actual or potential tissue damage, or described in terms of such damage.” Chronic pain refers to pain lasting or recurring for more than 3 mo[1]. Chronic low back pain is one of the leading causes of disability in the Chinese population[2,3]. Hence, treating chronic pain safely and effectively is one of the important clinical problems. In recent years, the research and development of new drugs for chronic pain via oral, intravenous and intramuscular delivery are not satisfactory, but transdermal delivery of analgesic drugs has made great progress[4].

Currently, no international guidelines or consensus are available on the use of transdermal analgesic drugs. Therefore, this article summarizes China’s expert consensus on “transdermal pain treatment in China,” with a view to regulate and promote the standardized use of the transdermal patch containing analgesic drugs. The consensus was formulated by the pain branch of the Chinese Medical Association with the help of experts and using recent clinical evidence.

OVERVIEW OF TRANSDERMAL DRUG DELIVERY SYSTEM

In the transdermal drug delivery system (TDDS), the drug enters systemic circulation through the skin and capillaries at a certain rate to achieve a systemic or local therapeutic effect[5,6]. TDDS in the broad sense includes all topically administered formulations such as ointment, patch, cataplasm, aerosol, coating, etc., whereas TDDS in the narrow sense refers to the transdermal patch[7]. The history of transdermal drug delivery dates back thousands of years. In ancient China, leaves and grass stalks were used to smear the skin and wounds. More than 1500 prescriptions are available in the treatise on external treatment of emergency Guangsheng in the Ming and Qing dynasties[8]. In the modern era, the scopolamine patch for the treatment of motion sickness was the first transdermal patch to be approved by the Food and Drug Administration (FDA) in 1979. From then on, the transdermal patch has been widely used to treat many diseases[9].

CLINICAL CHARACTERISTICS OF TDDS
Advantages of TDDS

Compared with other routes of administration, transdermal administration has many advantages, as summarized in Table 1[10,11].

Table 1 Advantages of transdermal drug delivery system administration.
Advantages
Simple administration and improved patient compliance
Avoids hepatic first pass metabolism
Avoids direct interaction of drugs with food or other drugs in the gastrointestinal tract, which may affect drug absorption
Helps in controlled drug delivery and reduces frequency of dosing
Reduces dosage and side effects
Can be removed from the skin surface immediately
Has physical form, characteristics and identification marks so that it can be easily and quickly identified in an emergency (such as when the patient is unresponsive, unconscious or comatose)
Absorption and factors influencing skin absorption

Traditionally, transdermal drugs are absorbed by passive diffusion. The stratum corneum, the outermost layer of the skin, is the main rate-limiting barrier for transdermal drug transport. Factors affecting transdermal absorption include physicochemical properties (molecular weight, solubility, partition coefficient and dissociation constant, PKA), carrier properties and skin conditions (Table 2)[10,12]. The rate and extent of drug absorption through skin can be calculated using the following formula:

Table 2 Factors influencing drug percutaneous absorption[10,12].
Influencing factor
Effect on transdermal drug absorption
Drug concentrationGenerally, the amount of drug absorbed per unit area per unit time increases with the increase in TDDS drug concentration
Drug distribution coefficientDrugs with both water-soluble and fat-soluble properties can be effectively absorbed through the skin. The water-soluble properties of drugs determine the concentration of the drug at the absorption site and the partition coefficient affects the rate of drug transport at the absorption site
Drug molecular weightThe ideal relative molecular weight for transdermal administration is 400 Da or less
Carrier factorThe main effects of carriers on percutaneous absorption include solubility of drugs in carriers and change of drug distribution coefficient by carrier
Site of application and timeThe larger the application area (TDDS) and the longer the application time, the more the drugs are absorbed
Skin conditionsHydration of skin helps increase percutaneous absorption. TDDS can form a closed water barrier with evaporating sweat to increase the hydration degree of the skin. It can be applied to the thin cuticle, with better absorption through the skin. When the skin is damaged, the drug will directly enter the subcutaneous tissue and capillaries, which may affect the properties of TDDS

Log P = −2.7 + 0.71 × log Ko/w − 0.0061 × M

Where, Ko/w = oil-water partition coefficient and M = molecular weight[9-11].

Systemic and local effects of TDDS

In TDDS for systemic delivery, the drug from TDDS gets transported to the subcutaneous capillaries through the skin without accumulating in the dermis. Once the drug reaches systemic circulation, it exhibits its therapeutic action. Examples of such TDDS include fentanyl transdermal patch, buprenorphine transdermal patch and scopolamine transdermal patch[10,13].

In TDDS, the drug is transported to the subcutaneous tissue through the skin and then to the deeper tissue to exert local action. Some of the TDDS for local action include nonsteroidal anti-inflammatory drugs (NSAIDs) transdermal patch, capsaicin patch and lidocaine patch[10,13].

Structure and development of the transdermal patch

According to the characteristics of its dosage form, the transdermal patch can be roughly divided into three generations (Table 3). The first generation of transdermal patch is the most representative transdermal patch[14].

Table 3 Development of transdermal patch[14].
Classification
Characteristic
First-generation transdermal patchThe drug should have suitable properties (highly potency, low molecular weight and lipophilic) to solve the problem of low oral bioavailability, to reduce the frequency of drug administration or to achieve stable drug administration
Second-generation transdermal patchThis generation of patch can promote and improve the percutaneous absorption of small molecule drugs by means of a chemical penetration enhancer, ion introduction or ultrasound
Third-generation transdermal patchThese patches help to promote percutaneous absorption of macromolecules, including therapeutic proteins and vaccines

The structure of a TDDS includes four layers: (1) an impermeable backing layer to protect the system from invasion of external substances and to prevent the loss of drugs or evaporation of skin moisture; (2) a drug storage or framework system to store and release drugs; (3) a liner that protects the patch during storage, which has to be removed prior to use; and (4) an adhesive layer to keep the patch in contact with the skin[10]. The adhesive layer can be divided into peripheral type and surface type. Peripheral type refers to applying a circle of pressure-sensitive adhesive on the periphery of the TDDS drug part. In surface type, TDDS is completely covered with finger-sensitive adhesive coating. Of the two types, the surface type of adhesive layer is the most common[10].

Only two types of TDDS were available before 1990: reservoir type and matrix type. After 1990, the adhesive dispersion type was introduced. In the adhesive dispersion type, the drug is dispersed in the adhesive layer itself; this helps to reduce the layers to two or three[15]. Since 1999, the FDA has not approved a reservoir type because of the risk of uncontrolled drug release from the reservoir. Most of the existing patches in the United States market (72%) are of the adhesive dispersion type[16].

Complications with application of patch

Dose adjustment is the main challenge faced in the use of a transdermal patch because only fixed dosages are commercially available[17]. In theory, an alternative option to reduce the dose is cutting the patch. However, cutting the patch may result in altering the structure of the patch, which may result in altering drug release, especially controlled release, and the quality of the adhesive layer[18] Cutting the microdrug reservoir of the patch can damage the microparticles of the drug, which leads to inaccuracy of dose evaluation[17,18]. A comparative study on the use of the clonidine patch (Catapres TTS) in sections and as a whole reported difficulty in predicting absorption degree and rate of drug release after cutting the patch, and the incidence of abnormal (too high or too low) blood concentration increased significantly with sections of patch. For most analgesic drugs, it is recommended to refer to the relevant instructions provided by the manufacturer[19].

Application site of patch: transdermal analgesic drugs can be applied at two application sites. Opioid transdermal patch is generally fixed on the chest, abdomen or upper arm because of its systemic effect. Topical patches with local effects such as topical NSAID patches, 8% capsaicin patch, 5% lidocaine patch and other patches are generally applied to the pain site[17,18].

COMMON CLINICAL TDDS
Transdermal NSAIDs

NSAIDs act by inhibiting prostaglandin synthesis and reduce persistent hyperalgesia by inhibiting cyclooxygenase activity to play an analgesic and anti-inflammatory role[20,21]. Compared with NSAIDs administered systemically, topical administration of NSAIDs reach therapeutic concentrations at the pain/inflammation site while maintaining low serum levels and potentially minimizing adverse effects related to high systemic absorption[20-22].

The advantages of topical NSAID application depend on the ability of the drug to penetrate the skin and reach the site of action. Different NSAIDs have different penetration abilities. Table 4 summarizes the pharmacokinetic properties of different topical NSAIDs[23-29]. According to studies reported, for a drug to be formulated as a TDDS, it should have ideal properties such as a partition coefficient (log P) of 1 to 4, molecular weight of < 400 Da, lipophilic, highly potent, low melting point and short half-life[6,30,31]. The peak plasma concentrations of NSAIDs vary greatly, but their concentrations in synovial fluid were much more stable than those in the plasma[32]. The plasma concentration of NSAIDs through topical delivery is about 1% to 10% of that of systemic administration. However, whether the concentration of NSAIDs in the local tissue of the application site is higher than that of systemic administration remains uncertain[32]. Most studies show that the concentration of NSAIDs in the deep tissue of the application site (such as the skeletal muscle and synovium) is equivalent to that of systemic administration. In addition, topical administration of NSAIDs in elderly patients may result in increased plasma concentration, which may be due to reduced drug clearance and thin skin[31,32].

Table 4 Pharmacokinetic characteristics of topical nonsteroidal anti-inflammatory drugs patches[23-29].

Loxoprofen sodium
Ketoprofen
Diclofenac sodium
Flurbiprofen
Indometacin
Ibuprofen
Log P value1.972.944.313.814.423.51
Cmax in ng/mL61.20891.360.8143.0027.00556.00
Tmax in h82.307.6016.9020.0016.0014.40
T1/2 in h7.8NANA13.9011.55NA

Topical NSAIDs can be used in different diseases such as acute sprain/strain, low back pain, chronic musculoskeletal pain and neuropathic pain[13,21,22,31-33]. A review showed similar efficacy with topical and oral NSAIDs in the treatment of chronic skeletal muscle pain[34,35]. When NSAIDs alone are not effective in treating moderate to severe pain or multisite pain, combination therapy with other analgesic drugs helps in effective treatment. When a combination of topical and oral NSAIDs is used, attention should be paid to avoid an overdose[36-40].

NSAIDs for topical application can be in the form of ointment, gel, gel paste and patch. Some studies compared topical NSAID patches with ointments and gelatin formulations. The results showed that the permeability of patches was better than that of gelatin and ointments. Compared with ointments, patches had better adherence[41-44].

Adverse effects with topical NSAIDs can be cutaneous and systemic adverse reactions. The incidence of cutaneous adverse reactions is about 1% to 2%, which include erythema, pruritus, irritation, fever or burning sensation and contact dermatitis. Most of the cutaneous adverse reactions were reported to be mild and disappeared after drug withdrawal[45-47].

Transdermal opioids

The commonly used opioid transdermal patches in China are the fentanyl transdermal patch and buprenorphine transdermal patch[13]. Table 5 represents the pharmacology of opioid transdermal patches[48-52].

Table 5 Pharmacology of fentanyl and buprenorphine transdermal patches[48-52].

Fentanyl transdermal patch
Buprenorphine transdermal patch
AbsorptionBioavailability, 92%; Plasma protein binding, 79%-87%; Cmax, 2.6 μg/L; Effective time, 12.7-16.6 h; Peak time 38.1 h; AUC, 117 μg/L; H (0-72 h)Bioavailability, 50%; Plasma protein binding, 96%; Cmax, 305 pg/mL; Onset time 21 h; Peak time, about 60 h; AUC, 20228 pg/mL
MetabolismMetabolized by CYP3A4 in the liver, and the metabolites are basically inactiveMetabolized by CYP3A4 in the liver
EliminationThe half-life of the transdermal patch is about 17 h (13-22 h)The half-life of the transdermal patch is 25.3 h
Mechanismμ opioid receptor agonistμ opioid receptor partial agonist, δ opioid receptor agonist, weak κ opioid receptor antagonist, ORL-1 agonist
IndicationModerate to severe chronic pain and intractable pain treated with opioid analgesicsChronic pain beyond the control of nonopioid analgesics
Dosage form specification2.1-, 4.2-, 8.4- and 12.6-mg paste; Four specifications, lasting for 72 h5-, 10- and 20-mg paste. Each paste is used for 7 d
Adverse reactions, > 10%Nausea, headache, constipation, dry mouth, drowsiness, fuzzy consciousness, powerlessness, sweatingErythema, pruritus, nausea
Fentanyl transdermal patch

Fentanyl is a strong opioid analgesic. Its analgesic intensity is about 100 times that of morphine, and its transdermal penetration ability is 43 times that of morphine. Because of its ideal properties such as small molecular weight (337 Da), highly lipophilic properties and no biotransformation in the process of transdermal penetration, it was the first analgesic drug formulated as a TDDS[49]. The fentanyl transdermal patch was approved by the FDA in 1990 for the treatment of chronic pain[13]. Other than chronic pain, it is also used for neuropathic pain and cancer pain[50].

Buprenorphine transdermal patch

Buprenorphine, a semisynthetic derivative of dimethylmorphine, is a partial agonist of the μ opioid receptor and antagonist of the κ receptor. The buprenorphine transdermal patch was approved by the FDA in 2010 for pain treatment. It is used for moderate to severe cancer pain, skeletal muscle pain, neuropathological pai, and visceral pain. As with fentanyl, buprenorphine is also not recommended in the treatment of acute and breakthrough pain[4,13,51,52].

Compared with fentanyl and other opioids, buprenorphine transdermal patch has lower neurotoxicity, especially in the elderly or patients with Alzheimer’s disease. Another significant advantage of the transdermal buprenorphine patch is that no dosage adjustment is needed in patients with renal insufficiency[13].

Opioid transdermal patches can significantly reduce pain and gastrointestinal related adverse effects such as nausea, vomiting and constipation and reduce the proportion of patients discontinuing treatment due to adverse reactions. However, in the treatment of chronic low back pain and knee and hip arthritis opioids (including opioid transdermal patches) were reported to have no significant difference in relieving pain and improving body function, while more adverse reactions were reported when compared with NSAIDs[53-57].

Capsaicin transdermal patch

Capsaicin is a selective transient receptor potential vanilloid receptor, subtype 1 agonist that activates the nociceptive sensory nerve fibers (C- and Ad-fibers) of transient receptor potential vanilloid receptor, subtype 1 in the skin, resulting in enhanced sensitivity to stimuli, burning sensation and erythema. Exposure to a single high dose or repeated exposure to low dose of capsaicin can lead to the nonfunctioning of nociceptors[13,58].

Studies reported absorption of 1% capsaicin into the epidermis and dermis after an hour of patch application. The absorption of capsaicin is directly proportional to the surface area and time of application. Skin temperature was also found to have an influence on the absorption of capsaicin. The results of a population pharmacokinetics study showed that peak plasma concentration (1.38 ng/mL) of an 8% capsaicin patch was attained after 1.46 h. Moreover, capsaicin has a high protein-binding capacity (93%-94%). After absorption, it is mainly metabolized in the liver by P450, and the elimination half-life is 1.64 h[59,60]. The transdermal patch available is 8% capsaicin at a dosing frequency of one patch per day. Capsaicin is used for the treatment of neuropathic pain. Better results were reported with capsaicin than placebo in the treatment of post herpetic neuralgia and diabetic peripheral neuralgia[61,62].

Lidocaine transdermal patch

Lidocaine is a voltage-gated sodium channel blocker (mainly Nav1.7 and 1.8). It can reduce the ectopic negative charge, increase the threshold of peripheral ectopic discharge and reduce the pain transmission by stabilizing the membrane potential of neurons on abnormal excitation of Aδ and C fibers to have an analgesic effect. In addition, the lidocaine patch also has an analgesic effect on pain from injurious sources[13,63].

Transdermal lidocaine majorly penetrates local tissues through the skin, and very little is absorbed into systemic circulation (about 3% ± 2%); hence, adverse reactions related to systemic administration can be avoided. Patients with severe impairment of heart, kidney and liver function should be cautious with lidocaine use. It is contraindicated in pregnant women and patients allergic to local anesthetics[13,64,65].

The lidocaine patch is generally well tolerated even after long term use. The most common adverse events include erythema, pruritus, rash, burning sensation, dermatitis, edema and other skin reactions. The lidocaine patch is used in the treatment of neuropathic pain, but there is no high-quality evidence to prove its clinical efficacy[64-69].

Other transdermal patches

The ketamine transdermal patch and dextromethorphan transdermal patch are common nonbarbiturate anesthetics. They are peripheral N-methyl-D-aspartate receptor inhibitors and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor inhibitors. They also inhibit voltage-gated Na+ and K+ channels[63].

Ketamine transdermal patch (25 mg/24 h) is used to relieve postoperative pain. The 4% dextromethorphan hydrochloride is formulated with 4% lidocaine and 10% trolamine salicylate (Permavan) and used to treat pain, but its efficacy and safety have not been determined. At present, neither of the two transdermal patches have been approved for marketing[70,71].

The bupivacaine transdermal patch is also under development. Compared with the lidocaine patch for 12 h, the bupivacaine patch can be applied once every 3 d and can be used for post herpetic neuralgia treatment. The United States bupivacaine transdermal patch (Eladur) is under investigation[71].

Rotigotine and amitriptyline are commonly used in the treatment of neuropathic pain. At present, the rotigotine transdermal patch (listed in Europe and the United States in 2007 and in China in 2018) and amitriptyline transdermal patch are available. The rotigotine transdermal patch can improve chronic pain in Parkinson’s disease[72]. However, neither of the drugs has been approved for pain treatment[73].

Clonidine, a α2 adrenergic receptor agonist and imidazoline receptor agonist, is used in antihypertension, acute and chronic pain management and sedation. However, the clonidine transdermal patch is currently approved only in the treatment of hypertension. The analgesic effect of the clonidine patch is related to the α2 receptor in the skin and the imidazoline receptor in the peripheral nerve endings, but clonidine use in the treatment of neuropathic pain is insufficient[71,74-77].

CONCLUSION

Based on expert opinions and careful assessment of the existing evidence, the classification and definition of grade evidence were summarized in Tables 6 and 7, while the consensus recommended by experts was summarized in Table 8. This consensus can help clinicians to use transdermal patches in pain management more effectively.

Table 6 Quality classification and definition of grade evidence.
Quality level
Definition
High (a)Very sure that the true effect value is close to the effect value estimation
Medium (b)There is a moderate degree of confidence in the value of effect; the real effect value may be close to the estimated value, but there is still a possibility that the two are not the same
Low (c)There is limited confidence in the effect estimates; the true effect values may not be the same as the effect estimates
Very low (d)There is little confidence in the estimated effect; the true effect value may be quite different from the effect estimate
Table 7 Grade recommended strength classification and definition.
Recommended strength
Explanation
Expression method
Expression method
Strong recommendations to support the use of an interventionThe advantages of the intervention measures outweigh the disadvantagesRecommended1
Weak recommendations to support the use of an interventionInterventions may have more advantages than disadvantagesRecommended use2
Weak recommendations against the use of an interventionInterventions may do more harm than good or the relationship between the advantages and disadvantages is not clearNot recommended2
Strong recommendations against the use of an interventionThe disadvantages of the intervention measures are obviously greater than the advantagesNot recommended1
Table 8 Consensus statement of Chinese experts on pain treatment with transdermal patch.
Consensus opinion
Recommended strength level of evidence
The effect of the transdermal patch in pain treatment is clear. It has the advantages of reducing adverse drug reactions and improving patient compliance1A
NSAID transdermal patch is effective in the treatment of chronic skeletal muscle pain with few side effects, which is recommended as the first choice for the treatment of chronic musculoskeletal pain1A
NSAIDs can be used as a combination therapy for neuropathic pain2C
When the efficacy of transdermal NSAIDs alone is not good enough, which can be combined with analgesic drugs of another administration route, such as oral NSAIDs2B
Opioid transdermal patch is effective in the treatment of chronic pain, but it should not be used as the initial treatment for chronic pain due to addiction and adverse reactions1B
Opioid transdermal patch should not be used in the treatment of acute or breakthrough pain1A
When other first-line treatment drugs are ineffective, 8% capsaicin patch can be considered for chronic pain related to peripheral neuropathic pain1B
When other first-line treatment drugs are ineffective, 5% lidocaine patch can be considered for chronic pain related to peripheral neuropathic pain2B
ACKNOWLEDGEMENTS

The authors would like to thank Daiichi-Sankyo Co., Ltd for the medical support of the manuscript.

Footnotes

Manuscript source: Unsolicited manuscript

Specialty type: Pharmacology and pharmacy

Country/Territory of origin: China

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Liang XY S-Editor: Zhang H L-Editor: Filipodia P-Editor: Ma YJ

References
1.  Lu Y, Cheng J, Fan B, Liu Y, Yu S, Zhang D, Fu Z, Song X, Yi X, Cheng Z, Liu X, Fu K, Ma K, Huang D, Yang X, Xiao L, Feng Z, Jin Y, Dong Z, Han J. ICD-11 Chinese version of chronic pain classification. Zhongguo Tengtong Yixue Zazhi. 2018;24:801-805.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Yang G, Wang Y, Zeng Y, Gao GF, Liang X, Zhou M, Wan X, Yu S, Jiang Y, Naghavi M, Vos T, Wang H, Lopez AD, Murray CJ. Rapid health transition in China, 1990-2010: findings from the Global Burden of Disease Study 2010. Lancet. 2013;381:1987-2015.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1375]  [Cited by in F6Publishing: 1328]  [Article Influence: 120.7]  [Reference Citation Analysis (0)]
3.  Dong WL, Li YC, Liu SW, Jiang YY, Mao F, Qi L, Zeng XY, Zhou MG. [The disease burden for low back pain in China, 1990 and 2013]. Zhonghua Yu Fang Yi Xue Za Zhi. 2017;51:132-136.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 4]  [Reference Citation Analysis (0)]
4.  Pergolizzi JV Jr, Coluzzi F, Taylor R Jr. Transdermal buprenorphine for moderate chronic noncancer pain syndromes. Expert Rev Neurother. 2018;18:359-369.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 10]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
5.  Ma HD, Wang L, Guo DY. Research progress of transdermal drug delivery system and new carriers. Shaanxi Zhongyi. 2017;38:1319-1320.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Fathima SA, Begum S, Fatima SS. Transdermal Drug Delivery System. Int J Pharm Clin Res. 2017;9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
7.  Zhang XH, Wang H, Hou HM. Progress of Composition and Properties of Pressure Sensitive Adhesive in Transdermal Drug Delivery System. Zhongguo Yaoxue Zazhi. 2008;39:767-772.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Gu ZT, Shen Y. History Brief of Herbal Transdermal Drug Delivery. Zhonghua Zhongyiyao Xue Kan. 2008;26:1771-1773.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Prausnitz MR, Mitragotri S, Langer R. Current status and future potential of transdermal drug delivery. Nat Rev Drug Discov. 2004;3:115-124.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 914]  [Cited by in F6Publishing: 803]  [Article Influence: 40.2]  [Reference Citation Analysis (0)]
10.  Allen L  Ansel Pharmaceutical Dosage System. 9th ed. Science Press; 2012.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Bolognia JL, Schaffer JV, Cerroni L.   Dermatology. 4th ed. Elsevier; 2018.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Pastore MN, Kalia YN, Horstmann M, Roberts MS. Transdermal patches: history, development and pharmacology. Br J Pharmacol. 2015;172:2179-2209.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 335]  [Cited by in F6Publishing: 256]  [Article Influence: 28.4]  [Reference Citation Analysis (0)]
13.  Leppert W, Malec-Milewska M, Zajaczkowska R, Wordliczek J. Transdermal and Topical Drug Administration in the Treatment of Pain. Molecules. 2018;23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 76]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
14.  Prausnitz MR, Langer R. Transdermal drug delivery. Nat Biotechnol. 2008;26:1261-1268.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1858]  [Cited by in F6Publishing: 1771]  [Article Influence: 118.1]  [Reference Citation Analysis (0)]
15.  Perumal O, Murthy SN, Kalia YN. Turning theory into practice: the development of modern transdermal drug delivery systems and future trends. Skin Pharmacol Physiol. 2013;26:331-342.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 30]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
16.  Zhong H, Chan G, Hu Y, Hu H, Ouyang D. A Comprehensive Map of FDA-Approved Pharmaceutical Products. Pharmaceutics. 2018;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 177]  [Cited by in F6Publishing: 159]  [Article Influence: 26.5]  [Reference Citation Analysis (0)]
17.  Durand C, Alhammad A, Willett KC. Practical considerations for optimal transdermal drug delivery. Am J Health Syst Pharm. 2012;69:116-124.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 35]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
18.  Ball AM, Smith KM. Optimizing transdermal drug therapy. Am J Health Syst Pharm. 2008;65:1337-1346.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 34]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
19.  Zuppa AF, Tejani SM, Cullen EJ Jr, Nadkarni VM. Plasma Concentrations Following Application of Whole versus Cut Transdermal Clonidine Patches To Critically Ill Children. J Pediatr Pharmacol Ther. 2004;9:43-48.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 6]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
20.  Kawabata A. Prostaglandin E2 and pain--an update. Biol Pharm Bull. 2011;34:1170-1173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 206]  [Cited by in F6Publishing: 216]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
21.  Rafanan BS Jr, Valdecañas BF, Lim BP, Malairungsakul A, Tassanawipas W, Shiyi C, Tse LF, Luong TK. Consensus recommendations for managing osteoarthritic pain with topical NSAIDs in Asia-Pacific. Pain Manag. 2018;8:115-128.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 18]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
22.  McPherson ML, Cimino NM. Topical NSAID formulations. Pain Med. 2013;14 Suppl 1:S35-S39.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 62]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
23.  Sawamura R, Sakurai H, Wada N, Nishiya Y, Honda T, Kazui M, Kurihara A, Shinagawa A, Izumi T. Bioactivation of loxoprofen to a pharmacologically active metabolite and its disposition kinetics in human skin. Biopharm Drug Dispos. 2015;36:352-363.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
24.  Wang M, Li L, Xie J, Sun Y, Ling G, He Z. Transdermal Adhesive Patches Loaded with Ketoprofen Evaluated by Dynamic Detection of Percutaneous Absorption. AAPS PharmSciTech. 2017;18:2141-2148.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
25.  Drago S, Imboden R, Schlatter P, Buylaert M, Krähenbühl S, Drewe J. Pharmacokinetics of Transdermal Etofenamate and Diclofenac in Healthy Volunteers. Basic Clin Pharmacol Toxicol. 2017;121:423-429.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
26.  Lewis F, Connolly MP, Bhatt A. A Pharmacokinetic Study of an Ibuprofen Topical Patch in Healthy Male and Female Adult Volunteers. Clin Pharmacol Drug Dev. 2018;7:684-691.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
27.  Ma J, Gao Y, Sun Y, Ding D, Zhang Q, Sun B, Wang M, Sun J, He Z. Tissue distribution and dermal drug determination of indomethacin transdermal-absorption patches. Drug Deliv Transl Res. 2017;7:617-624.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
28.  Taburet AM, Singlas E, Glass RC, Thomas F, Leutenegger E. Pharmacokinetic comparison of oral and local action transcutaneous flurbiprofen in healthy volunteers. J Clin Pharm Ther. 1995;20:101-107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 26]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
29.  Yano T, Nakagawa A, Tsuji M, Noda K. Skin permeability of various non-steroidal anti-inflammatory drugs in man. Life Sci. 1986;39:1043-1050.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 158]  [Cited by in F6Publishing: 168]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
30.  Benson HAE, Grice JE, Mohammed Y, Namjoshi S, Roberts MS. Topical and Transdermal Drug Delivery: From Simple Potions to Smart Technologies. Curr Drug Deliv. 2019;16:444-460.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 138]  [Article Influence: 27.6]  [Reference Citation Analysis (0)]
31.  Chandrashekar NS, Shobha Rani RH. Physicochemical and pharmacokinetic parameters in drug selection and loading for transdermal drug delivery. Indian J Pharm Sci. 2008;70:94-96.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 50]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
32.  Vaile JH, Davis P. Topical NSAIDs for musculoskeletal conditions. A review of the literature. Drugs. 1998;56:783-799.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 98]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
33.  Coderre TJ. Topical drug therapeutics for neuropathic pain. Expert Opin Pharmacother. 2018;19:1211-1220.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 12]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
34.  Rannou F, Pelletier JP, Martel-Pelletier J. Efficacy and safety of topical NSAIDs in the management of osteoarthritis: Evidence from real-life setting trials and surveys. Semin Arthritis Rheum. 2016;45:S18-S21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 116]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
35.  Derry S, Conaghan P, Da Silva JA, Wiffen PJ, Moore RA. Topical NSAIDs for chronic musculoskeletal pain in adults. Cochrane Database Syst Rev. 2016;4:CD007400.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 84]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
36.  Lin TC, Solomon DH, Tedeschi SK, Yoshida K, Kao Yang YH. Comparative Risk of Cardiovascular Outcomes Between Topical and Oral Nonselective NSAIDs in Taiwanese Patients With Rheumatoid Arthritis. J Am Heart Assoc. 2017;6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
37.  Honvo G, Leclercq V, Geerinck A, Thomas T, Veronese N, Charles A, Rabenda V, Beaudart C, Cooper C, Reginster JY, Bruyère O. Safety of Topical Non-steroidal Anti-Inflammatory Drugs in Osteoarthritis: Outcomes of a Systematic Review and Meta-Analysis. Drugs Aging. 2019;36:45-64.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 45]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
38.  Zeng C, Wei J, Persson MSM, Sarmanova A, Doherty M, Xie D, Wang Y, Li X, Li J, Long H, Lei G, Zhang W. Relative efficacy and safety of topical non-steroidal anti-inflammatory drugs for osteoarthritis: a systematic review and network meta-analysis of randomised controlled trials and observational studies. Br J Sports Med. 2018;52:642-650.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 113]  [Article Influence: 18.8]  [Reference Citation Analysis (0)]
39.  Chinese Medical Association Sports Medical Association;  Chinese Expert Committee on the Treatment of External NSAIDs Pain. Chinese expert consensus on the treatment of musculoskeletal pain with topical NSAIDs. Zhongguo Yixue Qianyan Zazhi. 2016;8:24-27.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Chinese Medical Doctor Association Pain Physician Branch National Key Clinical Specialist·China-Japan Hospital Pain Specialist Medical Consortium;  Beijing Pain Treatment Quality Control and Improvement Center. Expert consensus on drug treatment of chronic musculoskeletal pain (2018). Zhongguo Tengtong Yixue Zazhi. 2018;24:881-887.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Mazières B. Topical ketoprofen patch. Drugs R D. 2005;6:337-344.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 67]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
42.  Allegrini A, Nuzzo L, Pavone D, Tavella-Scaringi A, Giangreco D, Bucci M, Toniato E, Mezzetti A, Martinotti S, Comuzio S, Di Grigoli M, Bonani S. Efficacy and safety of piroxicam patch versus piroxicam cream in patients with lumbar osteoarthritis. A randomized, placebo-controlled study. Arzneimittelforschung. 2009;59:403-409.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 8]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
43.  Mayba JN, Gooderham MJ. A Guide to Topical Vehicle Formulations. J Cutan Med Surg. 2018;22:207-212.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 34]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
44.  Sugibayashi K  Skin Permeation and Disposition of Therapeutic and Cosmeceutical Compounds. Japan: Springer; 2017.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Romita P, Foti C, Calogiuri G, Cantore S, Ballini A, Dipalma G, Inchingolo F. Contact dermatitis due to transdermal therapeutic systems: a clinical update. Acta Biomed. 2018;90:5-10.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Bruyère O, Honvo G, Veronese N, Arden NK, Branco J, Curtis EM, Al-Daghri NM, Herrero-Beaumont G, Martel-Pelletier J, Pelletier JP, Rannou F, Rizzoli R, Roth R, Uebelhart D, Cooper C, Reginster JY. An updated algorithm recommendation for the management of knee osteoarthritis from the European Society for Clinical and Economic Aspects of Osteoporosis, Osteoarthritis and Musculoskeletal Diseases (ESCEO). Semin Arthritis Rheum. 2019;49:337-350.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 368]  [Cited by in F6Publishing: 247]  [Article Influence: 49.4]  [Reference Citation Analysis (0)]
47.  Bannuru RR, Osani MC, Vaysbrot EE, Arden NK, Bennell K, Bierma-Zeinstra SMA, Kraus VB, Lohmander LS, Abbott JH, Bhandari M, Blanco FJ, Espinosa R, Haugen IK, Lin J, Mandl LA, Moilanen E, Nakamura N, Snyder-Mackler L, Trojian T, Underwood M, McAlindon TE. OARSI guidelines for the non-surgical management of knee, hip, and polyarticular osteoarthritis. Osteoarthritis Cartilage. 2019;27:1578-1589.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1191]  [Cited by in F6Publishing: 1451]  [Article Influence: 290.2]  [Reference Citation Analysis (0)]
48.  Jeal W, Benfield P. Transdermal fentanyl. A review of its pharmacological properties and therapeutic efficacy in pain control. Drugs. 1997;53:109-138.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 176]  [Cited by in F6Publishing: 161]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
49.  Muijsers RB, Wagstaff AJ. Transdermal fentanyl: an updated review of its pharmacological properties and therapeutic efficacy in chronic cancer pain control. Drugs. 2001;61:2289-2307.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 179]  [Cited by in F6Publishing: 189]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
50.  US Food and Drug Administration  Duralgesic® prescribing information. June 24, 2019. [cited 30 November 2020]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/Label/2018/019813s075s076Lbl.pdf.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Evans HC, Easthope SE. Transdermal buprenorphine. Drugs. 2003;63:1999-2010; discussion 2011.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 100]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
52.  Pergolizzi JV Jr, Mercadante S, Echaburu AV, Van den Eynden B, Fragoso RM, Mordarski S, Lybaert W, Beniak J, Orońska A, Slama O;  Euromed Communications meeting. The role of transdermal buprenorphine in the treatment of cancer pain: an expert panel consensus. Curr Med Res Opin. 2009;25:1517-1528.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 48]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
53.  US Food and Drug Administration  BUTRANS® prescribing information. [cited 30 November 2020]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/Label/2018/021306s032s034Lbl.pdf.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Wolff RF, Aune D, Truyers C, Hernandez AV, Misso K, Riemsma R, Kleijnen J. Systematic review of efficacy and safety of buprenorphine versus fentanyl or morphine in patients with chronic moderate to severe pain. Curr Med Res Opin. 2012;28:833-845.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 51]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
55.  Wolff RF, Reid K, di Nisio M, Aune D, Truyers C, Hernandez AV, Misso K, Riemsma R, Kleijnen J. Systematic review of adverse events of buprenorphine patch versus fentanyl patch in patients with chronic moderate-to-severe pain. Pain Manag. 2012;2:351-362.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
56.  da Costa BR, Nüesch E, Kasteler R, Husni E, Welch V, Rutjes AW, Jüni P. Oral or transdermal opioids for osteoarthritis of the knee or hip. Cochrane Database Syst Rev. 2014: CD003115.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 88]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
57.  Busse JW, Wang L, Kamaleldin M, Craigie S, Riva JJ, Montoya L, Mulla SM, Lopes LC, Vogel N, Chen E, Kirmayr K, De Oliveira K, Olivieri L, Kaushal A, Chaparro LE, Oyberman I, Agarwal A, Couban R, Tsoi L, Lam T, Vandvik PO, Hsu S, Bala MM, Schandelmaier S, Scheidecker A, Ebrahim S, Ashoorion V, Rehman Y, Hong PJ, Ross S, Johnston BC, Kunz R, Sun X, Buckley N, Sessler DI, Guyatt GH. Opioids for Chronic Noncancer Pain: A Systematic Review and Meta-analysis. JAMA. 2018;320:2448-2460.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 358]  [Cited by in F6Publishing: 397]  [Article Influence: 66.2]  [Reference Citation Analysis (0)]
58.  Krebs EE, Gravely A, Nugent S, Jensen AC, DeRonne B, Goldsmith ES, Kroenke K, Bair MJ, Noorbaloochi S. Effect of Opioid vs Nonopioid Medications on Pain-Related Function in Patients With Chronic Back Pain or Hip or Knee Osteoarthritis Pain: The SPACE Randomized Clinical Trial. JAMA. 2018;319:872-882.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 635]  [Cited by in F6Publishing: 575]  [Article Influence: 95.8]  [Reference Citation Analysis (0)]
59.  Burness CB, McCormack PL. Capsaicin 8 % Patch: A Review in Peripheral Neuropathic Pain. Drugs. 2016;76:123-134.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 37]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
60.  Anand P, Bley K. Topical capsaicin for pain management: therapeutic potential and mechanisms of action of the new high-concentration capsaicin 8% patch. Br J Anaesth. 2011;107:490-502.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 476]  [Cited by in F6Publishing: 402]  [Article Influence: 30.9]  [Reference Citation Analysis (0)]
61.  Blair HA. Capsaicin 8% Dermal Patch: A Review in Peripheral Neuropathic Pain. Drugs. 2018;78:1489-1500.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 46]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
62.  Derry S, Rice AS, Cole P, Tan T, Moore RA. Topical capsaicin (high concentration) for chronic neuropathic pain in adults. Cochrane Database Syst Rev. 2017;1:CD007393.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 80]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
63.  Persson MSM, Stocks J, Walsh DA, Doherty M, Zhang W. The relative efficacy of topical non-steroidal anti-inflammatory drugs and capsaicin in osteoarthritis: a network meta-analysis of randomised controlled trials. Osteoarthritis Cartilage. 2018;26:1575-1582.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 42]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
64.  Yang XD, Fang PF, Xiang DX, Yang YY. Topical treatments for diabetic neuropathic pain. Exp Ther Med. 2019;17:1963-1976.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 11]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
65.  Mick G, Correa-Illanes G. Topical pain management with the 5% lidocaine medicated plaster--a review. Curr Med Res Opin. 2012;28:937-951.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 57]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
66.  de León-Casasola OA, Mayoral V. The topical 5% lidocaine medicated plaster in localized neuropathic pain: a reappraisal of the clinical evidence. J Pain Res. 2016;9:67-79.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 24]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
67.  Sabatowski R, Hans G, Tacken I, Kapanadze S, Buchheister B, Baron R. Safety and efficacy outcomes of long-term treatment up to 4 years with 5% lidocaine medicated plaster in patients with post-herpetic neuralgia. Curr Med Res Opin. 2012;28:1337-1346.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 28]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
68.  Hans G, Sabatowski R, Binder A, Boesl I, Rogers P, Baron R. Efficacy and tolerability of a 5% lidocaine medicated plaster for the topical treatment of post-herpetic neuralgia: results of a long-term study. Curr Med Res Opin. 2009;25:1295-1305.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 73]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
69.  Derry S, Wiffen PJ, Moore RA, Quinlan J. Topical lidocaine for neuropathic pain in adults. Cochrane Database Syst Rev.  2014: CD010958.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 73]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
70.  Finnerup NB, Attal N, Haroutounian S, McNicol E, Baron R, Dworkin RH, Gilron I, Haanpää M, Hansson P, Jensen TS, Kamerman PR, Lund K, Moore A, Raja SN, Rice AS, Rowbotham M, Sena E, Siddall P, Smith BH, Wallace M. Pharmacotherapy for neuropathic pain in adults: a systematic review and meta-analysis. Lancet Neurol. 2015;14:162-173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2249]  [Cited by in F6Publishing: 2197]  [Article Influence: 244.1]  [Reference Citation Analysis (0)]
71.  Azevedo VM, Lauretti GR, Pereira NL, Reis MP. Transdermal ketamine as an adjuvant for postoperative analgesia after abdominal gynecological surgery using lidocaine epidural blockade. Anesth Analg. 2000;91:1479-1482.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 30]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
72.  Pickering G, Martin E, Tiberghien F, Delorme C, Mick G. Localized neuropathic pain: an expert consensus on local treatments. Drug Des Devel Ther. 2017;11:2709-2718.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 46]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
73.  Rascol O, Zesiewicz T, Chaudhuri KR, Asgharnejad M, Surmann E, Dohin E, Nilius S, Bauer L. A Randomized Controlled Exploratory Pilot Study to Evaluate the Effect of Rotigotine Transdermal Patch on Parkinson's Disease-Associated Chronic Pain. J Clin Pharmacol. 2016;56:852-861.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 67]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
74.  Gerner P, Kao G, Srinivasa V, Narang S, Wang GK. Topical amitriptyline in healthy volunteers. Reg Anesth Pain Med. 2003;28:289-293.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 12]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
75.  US Food and Drug Administration  Catapres-TTS® label information. August 10, 2019. [cited 30 November 2020]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/Label/2012/018891s028 Lbl.pdf.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Knezevic NN, Tverdohleb T, Nikibin F, Knezevic I, Candido KD. Management of chronic neuropathic pain with single and compounded topical analgesics. Pain Manag. 2017;7:537-558.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 32]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
77.  Wrzosek A, Woron J, Dobrogowski J, Jakowicka-Wordliczek J, Wordliczek J. Topical clonidine for neuropathic pain. Cochrane Database Syst Rev. 2015;8:CD010967.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 18]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]