Skip to content
Publicly Available Published by De Gruyter October 4, 2019

Changes in inflammatory plasma proteins from patients with chronic pain associated with treatment in an interdisciplinary multimodal rehabilitation program – an explorative multivariate pilot study

  • Björn Gerdle EMAIL logo , Emmanuel Bäckryd , Torkel Falkenberg , Erik Lundström and Bijar Ghafouri

Abstract

It has been suggested that alterations in inflammation molecules maintain chronic pain although little is known about how these factors influence homeostatic and inflammatory events in common chronic pain conditions. Nonpharmacological interventions might be associated with alterations in inflammation markers in blood. This study of patients with chronic pain investigates whether an interdisciplinary multimodal rehabilitation program (IMMRP) was associated with significant alterations in the plasma pattern of 68 cytokines/chemokines 1 year after rehabilitation and whether such changes were associated with clinical changes. Blood samples and self-reports of pain, psychological distress, and physical activity of 25 complex chronic pain patients were collected pre-IMMRP and at 12-month follow-up. Analyses of inflammatory proteins (cytokines/chemokines/growth factors) were performed directly in plasma using the multiplex immunoassay technology Meso Scale Discovery. This explorative pilot study found that 12 substances, mainly pro-inflammatory, decreased after IMMRP. In two other relatively small IMMRP studies, four of these proinflammatory markers were also associated with decreases. The pattern of cytokines/chemokines pre-IMMRP was associated with changes in psychological distress but not with pain or physical activity. The present study cannot impute cause and effect. These results together with the results of the two previous IMMRP studies suggest that there is a need for larger and more strictly controlled studies of IMMRP with respect to inflammatory markers in blood. Such studies need to consider responders/non-responders, additional therapies, involved pain mechanisms and diagnoses. This and the two other studies open up for developing biologically measurable outcomes from plasma. Such biomarkers will be an important tool for further development of IMMRP and possibly other treatments for patients w ith chronic pain.

1 Introduction

One-fifth of the European population has moderate to severe chronic pain [1]. The prevalence of local chronic pain conditions is high – e.g. chronic neck-shoulder pain CNSP is 8% and non-neuropathic low back pain CLBP is 19.5% [2], [3], [4]. These conditions are associated with psychological distress, low health, sick leave, and high socioeconomic costs [5]. CNSP and CLBP can gradually become more easily triggered and spread to most of the body (i.e. chronic wide spread pain [CWP]) with a 5–10% prevalence [6]. According to the ACR criteria, fibromyalgia (FM) has a prevalence of 2–4% and is a subgroup of CWP with generalised hyperalgesia [7], [8].

Few mechanism-based diagnoses exist for chronic pain; one exception is neuropathic pain. Typically, diagnoses according to ICD10 (International Statistical Classification of Diseases and Related Health Problems – Tenth Revision) are based on duration and anatomical locations such as CLBP. Hence, a certain clinical diagnosis may mechanistically combine different phenotypes [9]. The lack of blood biomarkers makes diagnostic procedures problematic [10].

Several decades ago, the above chronic pain conditions were considered to be of peripheral origin. Imaging techniques have challenged the peripheral origin hypothesis and provide evidence for altered central (CNS) nociceptive/pain processing and morphology [11], [12], [13], [14], [15], [16], [17]. Therefore, some researchers have characterised these chronic pain conditions as central pain conditions [18], [19].

Understanding of the relative roles of peripheral and central factors is fundamental for developing treatments. The CNS alterations may be driven by peripheral nociception generators to produce the clinical presentations [20], [21]. This suggestion is indirectly supported by observations that CNS alterations in CLBP or chronic hip osteoarthritis are normalised after effective peripheral treatment – e.g. facet joint injections or surgery [22], [23], [24]. Studies have uncovered direct support for a peripheral muscle involvement in chronic pain conditions such as increased muscle levels of serotonin, glutamate, pyruvate, and lactate in CNSP and CWP/FM and lowered concentrations of ATP and PCr in FM [25], [26], [27], [28]. However, only a few molecules have been investigated and it is unclear if important pathophysiological mechanisms have been targeted.

Both the immune system and the pain signalling system acutely and highly coordinate response to threats [29], [30]. After tissue injury, tissue substances are released that can stimulate and sensitise nociceptors. Immune cells such as cytokines, histamine, and serotonin are activated and released [31]. Nociceptors produce as well as respond to cytokines and chemokines [32]. Moreover, antidromic firing of nociceptors releases substance P, calcitonin gene-related peptide (CGRP), glutamate, and neurokinin A [32], [33], [34], all contributors to inflammation. Local inflammation can range from mild homeostatic reactions to full-scale inflammation [35]. Cytokines in the periphery and in the CNS show bi-directional relationships [36], [37], [38], [39]. Information on peripheral inflammatory activity is transmitted to the CNS resulting in, for example, sickness behaviour, decreased endogenous pain inhibition, and neuro-inflammation [39], [40], [41], [42].

It has been suggested that alterations in inflammation molecules maintain chronic pain. Although more than 200 cytokines and chemokines have been described, little is known about how these factors influence homeostatic and inflammatory events in common chronic pain conditions [43]. Human studies have generally been restricted to a few mainly pro-inflammatory molecules [44]. The literature is not consistent for CWP/FM with respect to muscle and blood [45], [46]. A review of CLBP vaguely concluded that “classic” pro-inflammatory cytokines (IL-6, TNF-α, IL-8, and IL-1β) are often elevated [10]. Hence, a heterogenous picture exists. In amputees with and without chronic residual limb pain, 11 of 37 inflammatory markers in blood showed significant differences [47]. In a study using a panel of 92 cytokines/chemokines, we found that 11 substances in plasma significantly differentiated between CWP/FM and controls [48].

When choosing treatments, chronic pain patients are largely managed using a trial-and-error approach [49]. Different activated neurobiological mechanisms such as the extent of peripheral biochemical alterations in the patients may explain the small to moderate effect sizes of common non-pharmacological interventions [50], [51], [52], [53]. Interdisciplinary multimodal pain rehabilitation programs (IMMRP) are bio-psycho-social interventions that continue over several weeks with common goals in combination with individualised goals of the patient [54], [55], [56], [57]. Swedish IMMRPs include pain education, physical exercise, return to work strategies, and cognitive behavioural therapy (CBT) coordinated by a team [54], [55], [56], [57]. Systematic reviews (SRs) conclude higher efficacy of IMMRP compared with single-treatment or treatment-as-usual programs in chronic pain patients [55], [57], [58], [59], [60], [61], [62]. Wang et al. investigated whether the serum level of cytokine IL-8 changed after IMMRP in FM and reported a significant decrease in IL-8 [63]. Very recently Hysing et al. from a group of patients with mixed severe chronic pain conditions reported that participation in IMMRP was associated with significant reductions in seven inflammatory biomarkers [64]. In addition, two components of IMMRP – physical exercise and CBT – might be associated with alterations in peripheral inflammation markers [65], [66], [67], [68], [69].

This explorative pilot study investigates whether IMMRP is associated with significant alterations in the plasma pattern of 68 cytokines and chemokines 12 months after IMMRP and whether such changes are associated with changes in clinical aspects.

2 Subjects and methods

2.1 Study design

This longitudinal study investigates chronic pain patients before IMMRP and at a 12-month follow-up.

2.2 Subjects

The subjects were referred to the Pain and Rehabilitation Centre, University Hospital, Linköping, Sweden by other healthcare services (mainly primary healthcare) for complex chronic non-malignant pain conditions. The only exclusion criterium was not being of working age (18–67 years).

The Pain and Rehabilitation Centre is associated with the Swedish Quality Registry for Pain Rehabilitation (SQRP), which is recognized by the Swedish Association of Local Authorities and Regions. All relevant clinical departments/centres within specialist care throughout Sweden deliver data to SQRP [70]. The SQRP is based on questionnaires (i.e. PROM data); a detailed description of the SQRP is reported elsewhere [71]. The patients complete the SQRP questionnaires during their first visit to the Pain and Rehabilitation Centre. During their first visit, all the patients were given a medical examination (see below). After the patients provided informed consent to participate in the study and signed a consent form, which was in accordance with the Declaration of Helsinki, they committed to answer selected questions from SQRP immediately after (post-IMMRP) as well as at a 6-month and 12-month follow-up (12-m FU). In the present explorative pilot study, we compared data from the pre-IMMRP and the 12-m FU. In addition to the questionnaires in the SQRP, three blood samples were drawn from the patients. After final recruiting, there were 25 patients (17 women and eight men) with a mean age (SD) of 47.3 (10.5).

The study was approved by the Regional Ethics Committee in Stockholm (Dnr: 2014/953-31/1).

2.3 Clinical examination

All patients were given a standard routine clinical examination performed by a MD. The examination consisted of routine neurological examination, including registrations of systolic and diastolic blood pressures and auscultation of heart and lungs. All subjects had their weight (kg) and height (m) registered and these data were used to calculate their body mass index (BMI; kg/m2). Each patient’s diagnosis was determined using International Statistical Classification of Diseases and Related Health Problems – Tenth Revision, Swedish version (ICD10-SE). The different diagnoses of the patients can be seen in Table 1.

Table 1:

Diagnoses (ICD-10-SE) of the 25 patients.

Diagnose code Denotation Number of patients
M35.7 Hypermobility syndrome 1
M53.1 Cervicobrachial syndrome 2
M54.4 Lumbago with sciatica 2
M54.5 Low back pain 2
M54.6 Pain in thoracic spine 1
M54.8 Other dorsalgia 1
M79.1 Myalgia 3
M79.7 Fibromyalgia 5
R51.9 Headache 1
R52.2A Chronic pain, nociceptive 3
R52.2B Chronic pain, neuropathic 1
R52.2C Other chronic pain 1
R52.9 Pain, unspecified 2

2.4 Patient reported outcome measures (PROM)

We retrieved selected data from the Swedish Quality Registry for Pain Rehabilitation (SQRP) for the patients. The PROM data retrieved from SQRP are briefly described below.

2.4.1 Pain intensity (NRS-7days)

Data collected included average pain intensity the previous week (numeric rating scale; 0=no pain and 10=worst possible pain).

2.4.2 Hospital Anxiety and Depression Scale (HADS)

HADS measures anxiety and depression [72] and consists of an anxiety subscale (HADS-A) and a depression subscale (HADS-D). Both subscales have seven items, scoring range between 0 and 21; lower score indicates lower possibility of anxiety or depression. HADS is frequently used in clinical practice as well as in research and has good psychometric characteristics [72], [73]. HADS has the following clinical cut-offs: 0–6, no symptoms; 7–10, probably symptomatic; and ≥11, severely symptomatic [72]. Hospital Anxiety and Depression Scale (HADS) is validated in its Swedish translation [74].

2.4.3 Physical activity and inactivity

The following three indicator items, which were developed by the Swedish National Board of Health and Welfare, were used to register physical activity and inactivity.

FYS1: During a typical week, how much time do you spend a week on physical exercise that makes you breathless, such as running, exercise gymnastics, or ball sports: 0=0–30 min; 1=30–60 min; 2=60–90 min; 3=90–120 min; or 4=>120 min?

FYS2: During a typical week, how much time do you spend a week on everyday exercise such as walking, cycling, or gardening: 0=0–30 min; 1=30–60 min; 2=60–90 min; 3=90–150 min; 4=150–300 min; or 5≥300 min?

FYS3: How long do you sit for a normal day (not including sleep): 0=always, 1=13–15 h; 2=10–12 h; 3=7–9 h; 4=4–6 h, 5=1–3 h; and 6=0 h/never?

2.4.4 IMMRP

Medical assessments and decisions to offer IMMRP were performed by senior physicians, primarily from specialists in rehabilitation medicine or similar specialties, or by specialists in training under the supervision of a senior colleague. Most patients were also assessed by a psychologist, an occupational therapist, and a physiotherapist. The following inclusion criteria for IMMRP were used: (i) disabling chronic pain (on sick leave or experiencing major interference in daily life due to chronic pain); (ii) age between 18 and 67 years; (iii) no further medical investigations needed; (iv) written consent to participate and attend IMMRP; (v) and agreement not to participate in other parallel treatments. General exclusion criteria included severe psychiatric morbidity, abuse of alcohol and/or drugs, diseases that did not allow physical exercise, or presence of red flags. Red flags are clinical indicators of possible serious underlying conditions requiring further medical intervention (i.e. other than IMMRP).

The IMMRP was conducted in groups of six to nine participants for 6 weeks (at least 20 h per week of group-based activities) and was based on CBT principles and included physical exercise, occupational therapy ergonomics (including training in coping strategies), and pain education. In addition, lectures in basic pain science and pain management were offered for both patients as well as for relatives, friends, and colleagues. The program also included work-related advice and support, and individually-tailored sessions with team members were available if necessary. Individual sessions might also be required for a few weeks following the program. The patients were encouraged to take an active part in goal setting.

2.4.5 Blood samples

Blood samples were collected before the IMMRP (pre-IMMRP) and at the 12-month follow-up (12-m FU). The two blood samples were centrifuged for removal of red blood cells and the plasma fraction was transferred to new tubes, aliquoted in small portions, and saved at −86 °C.

2.4.6 Chemical analyses

Analyses of inflammatory proteins (cytokines/chemokines/growth factors) were performed directly in plasma using the multiplex immunoassay technology Meso Scale Discovery (MSD) (Rockville, MD, USA). Using the manufacturer’s protocols for MSD, we analysed up to 68 custom kit-panel inflammatory substances with the electro-chemo-luminescence method. Data were collected and analysed using MESO QUICKPLEX SQ 120 instrument equipped with DISCOVERY WORKBENCH® data analysis software (Meso Scale Diagnostics, Rockville, MD, USA). On the day of analysis, samples were thawed, blinded, and randomly mixed. The lower and upper limits of detection (LLOD-ULOD), expressed in pg/mL, were different for each substance (Supplementary Table S1).

2.5 Statistics

Statistical analyses were made using IBM SPSS (version 24.0; IBM Corporation, Armonk, NY, USA) and SIMCA-P+ (version 15.0; Sartorius Stedim Biotech, Umeå, Sweden) and p≤0.05 was used as level of significance in all analyses. Data are presented as mean±one standard deviation (±1SD) together with median. Non-parametric tests were used for comparisons within the patient group (i.e. Friedman’s Two-way Analysis of Variance by Ranks) for clinical variables and inflammatory substances over time. Spearman’s rank correlation test was used for bivariate correlation analysis. Effect sizes (ES; Cohen’s d) for within group analysis were computed using a calculator when appropriate (https://webpower.psychstat.org/models/means01/effectsize.php).

Traditional univariate statistical methods can quantify level changes of individual substances but disregard interrelationships and thereby ignore system-wide aspects. Moreover, traditional statistical methods (e.g. multiple and logistic regression) have obvious problems handling data sets with more variables than subjects (i.e. short and broad data sets). Therefore, we used advanced multivariate data analysis by projection (MVDA) using SIMCA-P+. When applying MVDA, we followed the recommendations concerning proteomics data presented by Wheelock and Wheelock [75]. Variables were mean centred and scaled for unified variance (UV-scaling). An unsupervised principal component analysis (PCA) was used to detect whether moderate or strong outliers existed. No multivariate outlier was detected. PCA displays systematic variation in the data matrix. All variables were log transformed before the statistical analyses if data were skewed. A cross validation technique was used to identify nontrivial components (p). Variables loading on the same component p were correlated, and variables with high loadings but with opposing signs were negatively correlated. Obtained components are per definition not correlated and are arranged in decreasing order with respect to explained variation. The loading plot reports the multivariate relationships between variables. A corresponding plot reporting the relationships between subjects (i.e. t-scores) can also be used (score plot) and each subject obtained a score (t) for each of the significant components.

In order to reduce the risk for false positive results when investigating possible changes in the panel of 68 inflammatory biomarkers in relatively few subjects, a PCA was made using data from pre-IMMRP and from the 12-month FU. That is, each patient had two values for each substance. If a significant PCA will be obtained the t-scores for each component and each patient are selected for the two time points. If significant differences in t-scores were found this indicated that differences in the inflammatory panel existed over time in a multivariate perspective. Only then it will be justified to identify substances associated with significant changes using traditional non-parametric tests (i.e. Friedman’s Two-way Analysis of Variance by Ranks).

Orthogonal Partial least squares (OPLS) regression was used when regressing the clinical variables using the inflammatory markers as regressors (X-variables). Regressors with regression coefficients with a jack-knifed 95% confidence interval not including 0 and the variable of importance (VIP) value exceeding 1 were considered important. The OPLS analyses were made in two steps. First, from the analysis all the proteins, we selected proteins with VIP>1.3 combined with the jack-knifed confidence intervals in the coefficients plot not including zero. Second, these proteins were used in a new regression, which is presented in the results. The tables also present p(corr) for each significant protein: the loading of each variable scaled as a correlation coefficient and thus standardising the range from −1 to +1; p(corr) is stable during iterative variable selection and comparable between models. An absolute p(corr)>0.4–0.5 is generally considered significant [75]. R2 describes the goodness of fit – the fraction of sum of squares of all the variables explained by a principal component. Q2 describes the goodness of prediction – the fraction of the total variation of the variables that can be predicted by a principal component using cross validation methods. R2 should not be considerably greater than Q2; if R2 is substantially greater than Q2 (a difference>0.3) [76], the robustness of the model is poor, implying overfitting [75]. Moreover, Analysis of Variance of Cross-Validated predictive residuals (CV-ANOVA), which is a SIMCA-P+ diagnostic tool for assessing model reliability, was also computed. CV-ANOVA provides a familiar p-value metric for the model [75].

3 Results

3.1 Clinical variables

Blood pressures, BMI, and PROM data at the two time points are shown in Table 2; no significant changes occurred over time in these variables according to omnibus testing (p-values: 0.088–0.959).

Table 2:

Clinical variables and PROM data before IMMRP (pre-IMMRP) and at 12-month follow-up (12-m FU).

Time points
Pre-IMMRP
12-m FU
Statistics
Variables Mean SD Median Mean SD Median p-Value
Blood pressure diastolic (mmHg) 82.8 11.1 80.0 80.2 8.5 77.5 0.088
Blood pressure systolic (mmHg) 127.2 18.6 120.0 121.2 15.8 115.0 0.145
BMI (kg/m2) 26.9 3.5 25.2 26.9 4.0 24.6 0.627
NRS-7days 6.7 1.4 7.0 6.2 2.2 6.5 0.353
HADS-A 7.8 5.3 7.5 8.8 6.2 5.0 0.864
HADS-D 7.6 3.9 8.5 8.1 4.8 6.0 0.876
FYS1 1.8 1.5 1.5 2.1 1.8 1.5 0.959
FYS2 2.6 1.3 2.5 2.6 1.6 2.0 0.751
FYS3 3.6 1.0 4.0 3.4 1.5 4.0 0.334
  1. Mean, SD and Median are reported. Furthest to the right are the statistical comparisons between the two time points. IMMRP=interdisciplinary multimodal pain rehabilitation program; BMI=body mass index; NRS-7days=average pain intensity previous week according to a numeric rating scale; HADS=Hospital Anxiety and Depression Scale; HADS-A=the anxiety subscale of HADS; HADS-D=the depression subscale of HADS; FYS1=amount of heavy physical exercise (see Methods for details); FYS2=amount of every day exercise (see methods for details); FYS3=amount of sitting (see Methods for details).

When scrutinising the individual patterns over time (pre-IMMRP vs. 12-m FU), 11 patients reported decreased pain intensity, seven patients reported no changes, and seven reported deterioration. HADS revealed similar symptom patterns for anxiety and depression: for anxiety, seven improved, two were unchanged, and eight worsened; and for depression, seven improved, one was unchanged, and nine worsened. Both these variables had missing data for eight patients.

3.2 Inflammatory substances

In total, 60 of 68 proteins were detected in at least 50% of the samples in each group and were included in the statistical analysis; data pre-IMMRP and 12-m FU are presented in Supplementary Table S2. To investigate possible effects of IMMRP on the panel of 60 inflammatory biomarkers, a PCA was made using data from pre-IMMRP and from the 12-month FU. That is, each patient had two values for each substance. A significant PCA with three components was obtained (R2 cumulative=0.41, Q2 cumulative=0.20). From this analysis, we obtained the t-scores for each component and each patient for the two time points. These results were compared with respect to the two time points. The scores of the first and third components differed significantly between pre-IMMRP and the 12-month follow-up (t1 pre-IMMRP vs. t1 12-m FU: −0.40±4.18 vs. 0.28±3.57, p=0.042; t2 pre-IMMRP vs. t2 12-m FU: −0.16±2.76 vs. 0.21±2.40, p=0.201; t3 pre-IMMRP vs. t3 12-m FU: −0.33±2.00 vs. 0.48±1.79, p=0.019). Hence, in a multivariate perspective differences in the inflammatory panel existed over time. These results justified trying to identify substances associated with significant changes. In this explorative pilot study, we performed Wilcoxon signed rank tests to identify substances that differed between pre-IMMRP and the 12-m FU (Table 3). Twelve substances showed significant alterations (i.e. pre-IMMRP vs. 12-m FU); IL-8 showed a borderline significant alteration (p=0.054) (Table 3). All substances including IL-8 showed decreases from pre-IMMRP to 12-m FU except Eotaxin-3/CCL26, which increased significantly.

Table 3:

Substances with significant changes between before IMMRP (pre-IMMRP) and the 12-month follow-up (12-m FU).

Time points
Pre-IMMRP
12-m FU
Statistics
ES
Substances Mean SD Median Mean SD Median p-Value Cohen’s d
ENA-78/CXCL5 66.71 48.22 40.58 30.92 23.94 24.23 <0.001 0.86
IL-7 0.69 0.52 0.54 0.40 0.37 0.29 <0.001 0.78
TARC/CCL17 19.03 11.59 17.48 13.13 5.03 13.96 <0.001 0.68
Gro-α/CXCL1 24.31 10.04 25.40 17.82 9.2 14.10 0.001 0.62
IL-16 100.37 40.52 89.68 90.09 30.98 79.58 0.016 0.51
SDF-1α/CXCL12 617.51 239.78 588.94 552.48 199.21 484.90 0.028 0.49
VEGF-A 9.06 6.31 7.10 7.05 4.03 6.29 0.030 0.47
IP-10/CXCL10 139.48 142.23 110.76 97.18 63.29 77.71 0.011 0.42
IL-8 1.5 0.65 1.38 1.31 0.49 1.32 0.054 0.39
IL-33 2.34 4.27 0.80 0.94 1.28 0.23 0.030 0.37
Eotaxin-2/CCL24 519.19 406.93 474.74 485.85 404.74 389.84 0.035 0.35
Eotaxin-3/CCL26 11.51 21.59 3.72 18.7 37.05 10.58 0.023 −0.35
TPO/MGDF 153.93 57.75 148.66 148.37 55.77 138.71 0.021 0.17
  1. Mean, SD and Median are reported. Furthest to the right statistical are the comparisons between the two time points and the within-group effect sizes (Cohen’s d). IMMRP=interdisciplinary multimodal pain rehabilitation program.

We found no significant relationships between the inflammatory substances pre-IMMRP and changes in NRS-7days, BMI, blood pressure, and FYS1-3. However, the pattern of inflammatory substances pre-IMMRP correlated with changes in HADS-A and HADS-D (Table 4), but the number of subjects was reduced (n=17) in these analyses due to missing data in HADS at 12-m FU. When these analyses were re-run using the substances with VIP>1.3, significant regressions were obtained for the changes in HADS-A (R2=0.75, Q2=0.63, CV-ANOVA p-value<0.001) and in HADS-D (R2=0.57, Q2=0.39, CV-ANOVA p-value=0.032).

Table 4:

OPLS regressions of changes in HAD-A and HAD-D, respectively using the pre-IMMRP values of the inflammatory substances as regressors (X-variables).

Changes in HADS-A
Changes in HADS-D
Variables VIP p(corr) Variables VIP p(corr)
IL-15 2.85 0.89 Fractalkine/CX3CL1 2.38 0.65
SDF-1α/CXCL12 2.47 0.77 MCP-4/CCL13 2.17 0.59
IL-10 2.38 0.75 MDC/CCL22 1.99 0.54
IL-4 2.33 0.73 MIP-3β 1.88 0.51
MCP-4/CCL13 2.07 0.65 SDF-1α/CXCL12 1.68 0.46
MDC/CCL22 1.63 0.51 IL-15 1.63 0.44
IL-22 1.62 0.51 IL-23 1.61 −0.44
IL-12p70 1.56 0.49 IL-10 1.58 0.43
TNF-β 1.56 0.49 IL-17E/IL-25 1.50 −0.41
IL-17F 1.40 −0.44 IP-10/CXCL10 1.43 0.39
MIF 1.34 −0.42 IL-17F 1.41 −0.38
TPO/MGDF 1.29 0.40 IL-2Ra 1.31 0.36
Fractalkine/CX3CL1 1.08 0.34 EPO 1.29 −0.35
CTACK/CCL27 1.05 0.33 IL-7 1.28 0.35
IFN-γ 1.00 0.31 IL-4 1.28 0.35
TARC/CCL17 1.12 0.30
MCP-3/CCL7 1.09 0.30
R2 0.96 R2 0.99
Q2 0.37 Q2 0.41
CV-ANOVA p-Value 0.49 CV-ANOVA p-value 0.89
n 17 n 17
  1. Only substances with VIP>1.0 are shown. Substances in bold type (i.e. VIP≥1.30) were used in a second regression (see text). For each protein with VIP>1.0 (VIP>1 is considered significant), VIP, and p(corr), a positive sign indicates that improvements in HADS were associated with high pre-IMMRP levels for the cytokines/chemokines The three bottom rows report R2, Q2, CV-ANOVA (p-value), and number of subjects (n). HADS=Hospital Anxiety and Depression Scale; HADS-A=the anxiety subscale of HADS; HADS-D=the depression subscale of HADS.

We found no significant relationships between changes in NRS-7d, HADS-A, BMI, and the changes in the 13 substances shown in Table 3. However, a PCA including changes in the clinical variables and changes in the 13 substances resulted in one significant component, indicating positive correlations between changes in FYS3 and changes in TARC/CCL17, ENA-78/CXCL5, IL-7, and IL-16. Using Spearman’s rank correlation tests, we found the relationship between changes in FYS3 and TARC/CCL17 was significant (ρ=−0.45); that is, improvements in FYS3 were associated with decreases in TARC/CCL17. The PCA also indicated positive correlations between changes in HAD-D and Gro-α/CXCL1, IL-7, SDF-1α/CXCL12, and Eotaxin-3/CCL26, but no significant correlations were obtained in bivariate correlation analyses (i.e. Spearman’s rank correlation tests). It was not possible to significantly regress (OPLS) changes in FYS3 or HAD-D using the 13 substances in Table 3 as regressors.

4 Discussion

4.1 Major findings

This explorative pilot study has two major findings:

  1. Participation in IMMRP was associated with significant changes in 12 substances: the majority of these were pro-inflammatory and showed decreases from pre-IMMRP to 12-m FU.

  2. The pattern of cytokines and chemokines pre-IMMRP was associated with changes in HADS-A and HADS-D.

There were statistically significant decreases in 12 substances at the 12-m FU. The majority of these substances were pro-inflammatory and decreased over time except Eotaxin-3/CCL26, which increased. In addition, substances characterised as growth factors (TPO/MGDF) and associated with angiogenesis (VEGF-A and IP-10/CXCL10) showed significant decreases.

4.2 Other relevant studies

We are aware of only two other studies investigating if participating in IMMRP is associated with changes in inflammatory mediators. Wang et al. investigating whether one cytokine (IL-8) in serum changed as a result of a three-week in-patient IMMRP for FM patients, found a significant decrease [63]. In a recent study, Hysing et al. investigated the plasma inflammatory pattern before and 12 months after an IMMRP over 6 months (n=28) in a group of mixed severe chronic pain conditions using a panel of 92 inflammatory substances [64]. They reported a significant decrease in overall inflammatory activity and found significant alterations in seven proteins at 12-month follow-up. Although these studies are small, their findings generally agree with respect to the effects of IMMRP on inflammatory substances in blood. However, these results from more or less explicitly stated pilot studies need to be confirmed in larger and better controlled IMMRP studies. In addition, for the individual components of IMMRP there is some support for effects on such biomarkers. Hence, a SR of non-pharmacological interventions in FM concluded that exercise interventions might act as anti-inflammatory treatment especially with respect to pro-inflammatory cytokines [69]. Other studies report that components of IMMRP (e.g. physical exercise, CBT, and mindfulness) might be associated with reductions in peripheral inflammation markers [64], [65], [66], [67], [68], [69].

4.3 IL-8

In this study, the biomarkers exhibiting changes possibly associated with IMMRP partially agree with the two earlier IMMRP studies [63], [64]. Hence, in our study, IL-8 tended to decrease (p=0.054), a finding that is in agreement with Wang et al.’s study of FM patients [63]. Our study had no controls, but increased IL-8 levels were found in FM [63], [77], in patients with lumbar radicular pain of severe intensity compared to patients with low pain intensity [78], in patients with sciatica [79], and in CRPS patients [80]. However, a recent review notes a lack of consensus among the literature concerning IL-8 levels in FM patients [81]. In contrast, lower levels in sera were found in patients with lumbar disc herniation [82]. Presently, it remains unclear whether it is appropriate to decrease IL-8 generally in patients with chronic pain conditions or whether this approach is restricted to certain diagnoses. Hysing et al. investigating a group of mixed chronic pain patients, found no significant change in IL-8 associated with participation in IMMRP. However, IL-8 levels decreased in FM patients after 4 months of a pool-aquatic programming [83] and after 6 months of analgesic treatment in CRPS 1 patients [84]. Hence, most studies indicate that IL-8 can be lowered using common interventions for chronic pain.

4.4 ENA-78/CXCL5, Gro-α/CXCL1, and IL-7

In agreement with the study by Hysing et al. [64], we found that three pro-inflammatory substances (ENA-78/CXCL5, Gro-α/CXCL1, and IL-7) had decreased significantly after IMMRP.

The presence of the protein ENA78/CXCL5, a pro-inflammatory substance, together with other pro-inflammatory cytokines associated with BMI possibly indicate low grade inflammation [85]. Furthermore, increased levels were found in prostatic secretions in chronic prostatitis [86]. In the present study, this substance had the highest effect size (0.86; i.e. moderate) of the substances with significant alterations (Table 3).

Some studies report increased blood levels of IL-7 in CRPS patients, in FM patients, and in a group of severe chronic pain patients [64], [77], [80], [87]. We found a significant decrease in the pro-inflammatory IL-7 over time with an effect size of 0.78 (second highest; i.e. moderate). This result agrees with Hysing et al. who also found a significant decrease associated with IMMRP.

The significant decrease in Gro-α/CXCL1 was associated with the fourth highest effect size (0.62). Gro-α/CXCL1 is a pro-inflammatory substance. Increased levels in blood have been found in patients with lumbar radicular pain of severe intensity compared to patients with low pain intensity [78], patients with FM [77], and a mixed group of patients with severe chronic pain [64].

4.5 Other pro-inflammatory substances

Moreover, we found that other pro-inflammatory substances were altered at the 12-m FU: TARC/CCL17, IL-16, SDF-1α/CXCL12, IP-10/CXCL10, IL-33, Eotaxin-2, and Eotaxin-3/CCL26.

TARC/CCL17 is elevated in many inflammatory conditions and in synovial fluid in OA [88]. In an animal model of OA, TARC/CCL17 blockade was associated with decreased pain [88]. In the present study, this substance was associated with a moderate effect size (0.68) among the significantly altered biomarkers, but we found no associations between changes in TARC/CCL17 (or the other substances reported in Table 3) and pain intensity.

IL-16 plays a crucial role in the inflammatory process as it acts as a chemoattractant for peripheral immune cells and has been linked to various inflammatory diseases such as asthma, Crohn’s disease, and rheumatoid arthritis [89]. Elevated plasma level has been found in acute myocardial infarction [89]. IL-16 had a moderate effect size (0.51) in the present study.

Recent animal studies indicate that SDF-1α/CXCL12 mediates the transition from acute pain to tonic pain state and thus contributes to the development and maintenance of persistent pain [90]. In the peripheral and central nervous systems, both SDF-1α/CXCL12 and their receptor chemokine C-X-C motif receptor 4 (CXCR4) are expressed in various kinds of nociceptive structures. This CXCL12/CXCR4 axis possesses pronociceptive properties involving both neuronal and glial mechanisms [91]. In intact animals, a single intrathecal injection of SDF-1α/CXCL12 produced transient mechanical allodynia [91].

For IP-10/CXCL10, a pro-inflammatory substance involved in angiogenesis, we found a significant decrease. Increased IP10/CXCL10 level was found in patients with lumbar radicular pain of severe intensity [78], in FM patients [92], and in untreated early rheumatoid arthritis patients [93].

IL-33, which boosts inflammation and pain [94], signals through the ST2 receptor and promotes inflammation by activating downstream pathways culminating in the production of pro-inflammatory mediators such as IL-1β, TNF-α, and IL-6 in an NF-κB-dependent manner [95]. IL-33 appears to worsen diseases by potentiating inflammatory and nociceptive stimuli [94]. However, significantly lower serum levels in FM have been reported [46]. We are not aware of any other study of relevant chronic pain conditions reporting decreases associated with participation in IMMRP or interventions part of IMMRP.

Elevated blood levels of Eotaxin-1/CCL11 and/or Eotaxin-2/CCL24 have been found in FM patients [92], [96], [97]. Moreover, Eotaxin-1/CCL11 is increased in patients with lumbar radicular pain of severe intensity and in primary dysmenorrhea [78], [98]. In the present study, Eotaxin-2/CCL24 decreased while Eotaxin-3/CCL26 increased (Table 4). We have no explanation for the different patterns and the results must be confirmed in larger studies.

4.6 Factors associated with growth and angiogenesis

In addition, we found significant decreases for one growth factor and one factor associated with angiogenesis.

TPO/MGDF is a humoral growth factor and involved in the regulation of platelet production. Elevated TPO/MGDF levels have been reported in clinical conditions such as haematological diseases usually associated with thrombocytopenia, in critical diseases such as acute coronary syndromes and sepsis, and in smoking [99]. Infections and inflammation can trigger a rise in platelet count (i.e. reactive thrombocytosis), which has been linked to increased TPO/MGDF production [100]. Hence, the decreases in several inflammatory substances including TPO/MGDF may also explain reduction in number of platelets (not shown, data will be published elsewhere).

VEGF-A increases healing, regeneration, and revascularisation and it is increased in patients with lumbar radicular pain of severe intensity compared to patients with low pain intensity [78]. A decrease in this substance in blood was observed in patients with disc herniation after epidural steroid injection [101]. The decrease in the present study may indicate more favourable peripheral circumstances possibly due to the physical exercise component of IMMRP.

4.7 Clinical variables vs. inflammatory mediators

On the group level, we observed no significant changes in the clinical variables (pain intensity, psychological distress, and physical activity). However, some patients reported improvements and others reported no change or deteriorations. Such mixed results have been found also in considerably larger studies of IMMRP [102]. Most available evidence according to SRs suggests that IMMRP is effective (see Introduction) although these effects are small to moderate. We found significant relationships between the pattern of pro-inflammatory substances pre-IMMRP and changes in HADS-A and HADS-D at 12-m FU (Table 4). Aspects of psychological distress are important outcomes of IMMRP according to the outcome domains presented by the Initiative on Methods, Measurement, and Pain Assessment in Clinical Trials (IMMPACT) [103], [104] and the Validation and Application of a Patient-relevant Core Set of Outcome Domains to assess Multimodal PAIN Therapy (VAPAIN) [105] initiatives. A drawback with the analyses of changes in the two HADS subscales was missing data for a minor but substantial part of the patients; therefore, our results need to be confirmed in larger studies. However, both pro-inflammatory (IL-15, fractalkine/CX3CL1, and SDF-1α/CXCL12) and anti-inflammatory (IL-10, IL-4, IL-22, and IL-23) substances pre-IMMRP were associated with improvements with respect to anxiety and depression.

It was not possible to multivarietly correlate the inflammatory pattern pre-IMMRP with changes in pain intensity. Wang and colleagues found that pain intensity decreased at a six-month follow-up [63]. Before IMMRP, no correlation between IL-8 and pain intensity was found, but at the six-month follow-up they found negative correlations between pain intensity and IL-8 levels. Hysing et al. did not present any correlations between inflammatory biomarkers and clinical outcomes, but their patients had overall significant improvements associated with IMMRP [64]. In future larger and better controlled studies, it will be important to investigate changes in clinically important outcomes of IMMRP in relation to the pattern and changes of inflammatory biomarkers. To gain a complete understanding and interpretation of such analyses it will be important to include control groups.

4.8 Strengths and limitations

A major strength of this study is that it adds information about possible alterations in inflammatory proteins associated with participation in IMMRP; a field with only few studies. Our results together with the results of the two earlier IMMRP studies suggest that larger and more strictly controlled studies are needed. Another strength is that this study consists of real patients referred to the Pain and Rehabilitation Centre at a university hospital and not primarily recruited for a research project. However, the diagnoses of the patients contributing to this explorative pilot study as well as the recent study by Hysing et al. [64] were a mix of nociceptive, neuropathic, and nociplastic pain conditions of patients suffering from complex chronic pain. Hence, the involved pain mechanisms may differ and studies of more homogenous groups of chronic patients are warranted. These differences make it problematic to generalise the results to a specific group or diagnosis of chronic pain conditions. Our study also had a low number of patients and a lack of balance with respect to gender/sex, which could bias the results. A strength is that we used MVDA to determine whether changes on a general level occurred in the pattern of blood biomarkers; based on this result, we performed the analyses comparing pre-IMMRP and 12-m FU data. This approach diminishes but does not eliminate the risk for false positive results. Against our results can be argued that factors other than participating in IMMRP per se such as different amount of individual treatments in addition to IMMRP, change in pharmacological treatments and smoking habits may be responsible for the significant changes in the inflammatory mediators found at the 12-m FU. Other important limitations were that we only investigated a few of the immense number of inflammatory markers, we used subjective measurements, and we did not include healthy controls (for normalised concentrations of substances).

4.9 Conclusions and Implications

Patients with chronic pain conditions participating in IMMRP for 6 weeks had significantly lower concentrations of 12 mainly pro-inflammatory substances at the 12-month follow-up. There were some indications that the pattern of biochemical biomarkers pre-IMMRP correlated with changes in psychological distress variables. It is important to note that the present study cannot impute cause and effect. These results together with the results of the two previous IMMRP studies suggest that there is a need for larger and more strictly controlled studies of IMMRP with respect to inflammatory markers in blood. Such studies need to consider responders/non-responders, additional therapies, involved pain mechanisms and diagnoses.

The present and the two previous studies taken together open up for developing biologically measurable outcomes from plasma. Such biomarkers will be an important tool for further development of IMMRP and possibly other treatments for patients with chronic pain.


Corresponding author: Professor Björn Gerdle, MD, PhD, Pain and Rehabilitation Centre, and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden, Phone: +46763927191

  1. Authors’ statements

  2. Research funding: This study was supported by grants from the Swedish Research Council, County Council of Östergötland (Research-ALF), the Magnus Bergvall Foundation, the Åke Wiberg Foundation, and AFA Insurance. AFA Insurance, a commercial founder, is owned by Sweden’s labour market parties: The Confederation of Swedish Enterprise, the Swedish Trade Union Confederation (LO), and The Council for Negotiation and Co-operation (PTK). These parties insure employees in the private sector, municipalities, and county councils. AFA Insurance does not seek to generate a profit, which implies that no dividends are paid to shareholders. The sponsors of the study had no role in study design, data collection, data analysis, data interpretation, writing of the report, or the decision to submit for publication. The authors had full access to all the data in the study and had final responsibility for the decision to submit for publication.

  3. Conflict of interest: The authors report no conflicts of interest.

  4. Informed consent: All participants received written information about the study and gave their written consent.

  5. Ethical approval: The study was conducted in accordance with the Helsinki Declaration and Good Clinical Practice and approved by the Regional Ethics Committee in Stockholm (Dnr: 2014/953-31/1).

  6. Author contributions: Conceptualization, Björn Gerdle, Emmanuel Bäckryd, Torkel Falkenberg and Bijar Ghafouri; Data curation, Erik Lundström and Bijar Ghafouri; Formal analysis, Björn Gerdle, Erik Lundström and Bijar Ghafouri; Writing – original draft, Björn Gerdle; Writing – review & editing, Björn Gerdle, Emmanuel Bäckryd, Torkel Falkenberg, Erik Lundström and Bijar Ghafouri.

  7. Data availability statement: The datasets generated and/or analysed in this study are not publicly available as the Ethical Review Board has not approved the public availability of these data.

References

[1] Breivik H, Collett B, Ventafridda V, Cohen R, Gallacher D. Survey of chronic pain in Europe: prevalence, impact on daily life, and treatment. Eur J Pain 2006;10:287–333.10.1016/j.ejpain.2005.06.009Search in Google Scholar PubMed

[2] Lidgren L. Preface: neck pain and the decade of the Bone and Joint 2000–2010. Spine 2008;33:S1–2.10.1097/BRS.0b013e3181643f14Search in Google Scholar

[3] Meucci RD, Fassa AG, Faria NM. Prevalence of chronic low back pain: systematic review. Rev Saude Publica 2015;49:1.10.1590/S0034-8910.2015049005874Search in Google Scholar PubMed PubMed Central

[4] Fejer R, Kyvik KO, Hartvigsen J. The prevalence of neck pain in the world population: a systematic critical review of the literature. Eur Spine J 2006;15:834–48.10.1007/s00586-004-0864-4Search in Google Scholar PubMed PubMed Central

[5] Bergman S. Chronic musculoskeletal pain: a multifactorial process. Lund: Lund University, 2001.Search in Google Scholar

[6] Gerdle B, Bjork J, Coster L, Henriksson K, Henriksson C, Bengtsson A. Prevalence of widespread pain and associations with work status: a population study. BMC Musculoskelet Disord 2008;9:102.10.1186/1471-2474-9-102Search in Google Scholar PubMed PubMed Central

[7] Coster L, Kendall S, Gerdle B, Henriksson C, Henriksson KG, Bengtsson A. Chronic widespread musculoskeletal pain – A comparison of those who meet criteria for fibromyalgia and those who do not. Eur J Pain 2008;12:600–10.10.1016/j.ejpain.2007.10.001Search in Google Scholar PubMed

[8] Henriksson CM, Liedberg GM, Gerdle B. Women with fibromyalgia: work and rehabilitation. Disabil Rehabil 2005;27:685–94.10.1080/09638280400009089Search in Google Scholar PubMed

[9] Zanin M, Chorbev I, Stres B, Stalidzans E, Vera J, Tieri P, Castiglione F, Groen D, Zheng H, Baumbach J, Schmid JA, Basilio J, Klimek P, Debeljak N, Rozman D, Schmidt H. Community effort endorsing multiscale modelling, multiscale data science and multiscale computing for systems medicine. Brief Bioinform 2019;20:1057–62.10.1093/bib/bbx160Search in Google Scholar PubMed PubMed Central

[10] Khan AN, Jacobsen HE, Khan J, Filippi CG, Levine M, Lehman RA Jr, Riew KD, Lenke LG, Chahine NO. Inflammatory biomarkers of low back pain and disc degeneration: a review. Ann N Y Acad Sci 2017;1410:68–84.10.1111/nyas.13551Search in Google Scholar PubMed PubMed Central

[11] Petersel D, Dror V, Cheung R. Central amplification and fibromyalgia: disorder of pain processing. J Neurosci Res 2011;89:29–34.Search in Google Scholar

[12] Smith H, Harris R, Clauw D. Fibromyalgia: an afferent processing disorder leading to a complex pain generalized syndrome. Pain Physician 2011;14:E217–45.10.36076/ppj.2011/14/E217Search in Google Scholar

[13] Henry DE, Chiodo AE, Yang W. Central nervous system reorganization in a variety of chronic pain states: a review. PM R 2011;3:1116–25.10.1016/j.pmrj.2011.05.018Search in Google Scholar PubMed

[14] Nakamura Y, Nojiri K, Yoshihara H, Takahata T, Honda-Takahashi K, Kubo S, Sakatsume K, Kato H, Maruta T, Honda T. Significant differences of brain blood flow in patients with chronic low back pain and acute low back pain detected by brain SPECT. J Orthop Sci 2014;19:384–9.10.1007/s00776-014-0534-2Search in Google Scholar PubMed

[15] Ung H, Brown JE, Johnson KA, Younger J, Hush J, Mackey S. Multivariate classification of structural MRI data detects chronic low back pain. Cereb Cortex 2014;24:1037–44.10.1093/cercor/bhs378Search in Google Scholar PubMed PubMed Central

[16] Hong JY, Labus JS, Jiang Z, Ashe-Mcnalley C, Dinov I, Gupta A, Shi Y, Stains J, Heendeniya N, Smith SR, Tillisch K, Mayer EA. Regional neuroplastic brain changes in patients with chronic inflammatory and non-inflammatory visceral pain. PLoS One 2014;9:e84564.10.1371/journal.pone.0084564Search in Google Scholar PubMed PubMed Central

[17] Moayedi M, Weissman-Fogel I, Salomons TV, Crawley AP, Goldberg MB, Freeman BV, Tenenbaum HC, Davis KD. Abnormal gray matter aging in chronic pain patients. Brain Res 2012;1456:82–93.10.1016/j.brainres.2012.03.040Search in Google Scholar PubMed

[18] Petersel DL, Dror V, Cheung R. Central amplification and fibromyalgia: disorder of pain processing. J Neurosci Res 2011;89:29–34.10.1002/jnr.22512Search in Google Scholar PubMed

[19] Phillips K, Clauw DJ. Central pain mechanisms in the rheumatic diseases: future directions. Arthritis Rheum 2013;65:291–302.10.1002/art.37739Search in Google Scholar PubMed PubMed Central

[20] Bennett RM. Emerging concepts in the neurobiology of chronic pain: evidence of abnormal sensory processing in fibromyalgia. Mayo Clin Proc 1999;74:385–98.10.4065/74.4.385Search in Google Scholar PubMed

[21] Staud R. The role of peripheral input for chronic pain syndromes like fibromyalgia syndrome. J Musculoskelet Pain 2008;16:67–74.10.1080/10582450801960339Search in Google Scholar

[22] Seminowicz DA, Wideman TH, Naso L, Hatami-Khoroushahi Z, Fallatah S, Ware MA, Jarzem P, Bushnell MC, Shir Y, Ouellet JA, Stone LS. Effective treatment of chronic low back pain in humans reverses abnormal brain anatomy and function. J Neurosci 2011;31:7540–50.10.1523/JNEUROSCI.5280-10.2011Search in Google Scholar PubMed PubMed Central

[23] Rodriguez-Raecke R, Niemeier A, Ihle K, Ruether W, May A. Brain gray matter decrease in chronic pain is the consequence and not the cause of pain. J Neurosci 2009;29:13746–50.10.1523/JNEUROSCI.3687-09.2009Search in Google Scholar PubMed PubMed Central

[24] Rodriguez-Raecke R, Niemeier A, Ihle K, Ruether W, May A. Structural brain changes in chronic pain reflect probably neither damage nor atrophy. PLoS One 2013;8:e54475.10.1371/journal.pone.0054475Search in Google Scholar PubMed PubMed Central

[25] Gerdle B, Ghafouri B, Ernberg M, Larsson B. Chronic musculoskeletal pain: review of mechanisms and biochemical biomarkers as assessed by the microdialysis technique. J Pain Res 2014;7:313–26.10.2147/JPR.S59144Search in Google Scholar PubMed PubMed Central

[26] Gerdle B, Larsson B. Potential muscle biomarkers of chronic myalgia in humans – a systematic review of microdialysis studies. In: Khan T, editor. Biomarker.Rijeka, Croatia: INTECH open Access publisher, 2012.10.5772/38822Search in Google Scholar

[27] Gerdle B, Forsgren M, Bengtsson A, Dahlqvist Leinhard O, Sören B, Karlsson A, Brandejsky V, Lund E, Lundberg P. Decreased muscle concentrations of ATP and PCR in the quadriceps muscle of fibromyalgia patients – a 31P MRS study. Eur J Pain 2013;17:1205–15.10.1002/j.1532-2149.2013.00284.xSearch in Google Scholar PubMed

[28] Park JH, Phothimat P, Oates CT, Hernanz-Schulman M, Olsen NJ. Use of P-31 magnetic resonance spectroscopy to detect metabolic abnormalities in muscles of patients with fibromyalgia. Arthritis Rheum 1998;41:406–13.10.1002/1529-0131(199803)41:3<406::AID-ART5>3.0.CO;2-LSearch in Google Scholar

[29] McMahon S, La Russa F, Bennett D. Crosstalk between the nociceptive and immune systems in host defence and disease. Nat Rev Neurosci 2015;16:389–402.10.1038/nrn3946Search in Google Scholar

[30] Wang T, Ma C. Peripheral nociceptors as immune sensors in the development of pain and itch. Adv Exp Med Biol 2016;904:77–85.10.1007/978-94-017-7537-3_6Search in Google Scholar

[31] Loyd DR, Henry MA, Hargreaves KM. Serotonergic neuromodulation of peripheral nociceptors. Semin Cell Dev Biol 2013;24:51–7.10.1016/j.semcdb.2012.09.002Search in Google Scholar

[32] Ji RR, Chamessian A, Zhang YQ. Pain regulation by non-neuronal cells and inflammation. Science 2016;354:572–7.10.1126/science.aaf8924Search in Google Scholar

[33] Carlton SM. Nociceptive primary afferents: they have a mind of their own. J Physiol 2014;592:3403–11.10.1113/jphysiol.2013.269654Search in Google Scholar PubMed PubMed Central

[34] Littlejohn G. Neurogenic neuroinflammation in fibromyalgia and complex regional pain syndrome. Nat Rev Rheumatol 2015;11:639–48.10.1038/nrrheum.2015.100Search in Google Scholar PubMed

[35] Xanthos DN, Sandkuhler J. Neurogenic neuroinflammation: inflammatory CNS reactions in response to neuronal activity. Nat Rev Neurosci 2014;15:43–53.10.1038/nrn3617Search in Google Scholar PubMed

[36] Iwata M, Ota KT, Duman RS. The inflammasome: pathways linking psychological stress, depression, and systemic illnesses. Brain Behav Immun 2013;31:105–14.10.1016/j.bbi.2012.12.008Search in Google Scholar PubMed PubMed Central

[37] Verma V, Sheikh Z, Ahmed AS. Nociception and role of immune system in pain. Acta Neurol Belg 2015;115:213–20.10.1007/s13760-014-0411-ySearch in Google Scholar PubMed

[38] Pavlov VA, Tracey KJ. Neural circuitry and immunity. Immunol Res 2015;63:38–57.10.1007/s12026-015-8718-1Search in Google Scholar PubMed PubMed Central

[39] Eisenberger NI, Moieni M, Inagaki TK, Muscatell KA, Irwin MR. In sickness and in health: the co-regulation of inflammation and social behavior. Neuropsychopharmacology 2017;42:242–53.10.1038/npp.2016.141Search in Google Scholar PubMed PubMed Central

[40] Karshikoff B, Jensen KB, Kosek E, Kalpouzos G, Soop A, Ingvar M, Olgart Hoglund C, Lekander M, Axelsson J. Why sickness hurts: a central mechanism for pain induced by peripheral inflammation. Brain Behav Immun 2016;57:38–46.10.1016/j.bbi.2016.04.001Search in Google Scholar PubMed

[41] Lampa J, Westman M, Kadetoff D, Agreus AN, Le Maitre E, Gillis-Haegerstrand C, Andersson M, Khademi M, Corr M, Christianson CA, Delaney A, Yaksh TL, Kosek E, Svensson CI. Peripheral inflammatory disease associated with centrally activated IL-1 system in humans and mice. Proc Natl Acad Sci USA 2012;109:12728–33.10.1073/pnas.1118748109Search in Google Scholar PubMed PubMed Central

[42] Kempuraj D, Thangavel R, Selvakumar GP, Zaheer S, Ahmed ME, Raikwar SP, Zahoor H, Saeed D, Natteru PA, Iyer S, Zaheer A. Brain and Peripheral Atypical Inflammatory Mediators Potentiate Neuroinflammation and Neurodegeneration. Front Cell Neurosci 2017;11:216.10.3389/fncel.2017.00216Search in Google Scholar PubMed PubMed Central

[43] Turner MD, Nedjai B, Hurst T, Pennington DJ. Cytokines and chemokines: at the crossroads of cell signalling and inflammatory disease. Biochim Biophys Acta 2014;1843:2563–82.10.1016/j.bbamcr.2014.05.014Search in Google Scholar PubMed

[44] Antunes-Martins A, Perkins JR, Lees J, Hildebrandt T, Orengo C, Bennett DL. Systems biology approaches to finding novel pain mediators. Wiley Interdiscip Rev Syst Biol Med 2013;5:11–35.10.1002/wsbm.1192Search in Google Scholar PubMed

[45] Ernberg M, Christidis N, Ghafouri B, Bileviciute-Ljungar I, Lofgren M, Larsson A, Palstam A, Bjersing J, Mannerkorpi K, Kosek E, Gerdle B. Effects of 15 weeks of resistance exercise on pro-inflammatory cytokine levels in the vastus lateralis muscle of patients with fibromyalgia. Arthritis Res Ther 2016;18:137.10.1186/s13075-016-1041-ySearch in Google Scholar PubMed PubMed Central

[46] Tsilioni I, Russell IJ, Stewart JM, Gleason RM, Theoharides TC. Neuropeptides CRH, SP, HK-1, and inflammatory cytokines IL-6 and TNF are increased in serum of patients with fibromyalgia syndrome, implicating mast cells. J Pharmacol Exp Ther 2016;356:664–72.10.1124/jpet.115.230060Search in Google Scholar PubMed PubMed Central

[47] Chamessian A, Van de Ven T, Buchheit T, Hsia HL, McDuffie M, Gamazon ER, Walsh C, Bruehl S, Buckenmaier C 3rd, Shaw A. Differential expression of systemic inflammatory mediators in amputees with chronic residual limb pain. Pain 2017;158:68–74.10.1097/j.pain.0000000000000728Search in Google Scholar PubMed PubMed Central

[48] Gerdle B, Ghafouri B, Ghafouri N, Bäckryd E, Gordh T. Signs of ongoing inflammation in female patients with chronic widespread pain – a multivariate, explorative, cross-sectional study of blood samples. Medicine 2017;96:9(e6130).10.1097/MD.0000000000006130Search in Google Scholar PubMed PubMed Central

[49] Vardeh D, Mannion RJ, Woolf CJ. Toward a mechanism-based approach to pain diagnosis. J Pain 2016;17(9 Suppl):T50–69.10.1016/j.jpain.2016.03.001Search in Google Scholar PubMed PubMed Central

[50] Sommer C, Leinders M, Uceyler N. Inflammation in the pathophysiology of neuropathic pain. Pain 2018;159:595–602.10.1097/j.pain.0000000000001122Search in Google Scholar PubMed

[51] Turk DC, Wilson HD, Cahana A. Treatment of chronic non-cancer pain. Lancet 2011;377:2226–35.10.1016/S0140-6736(11)60402-9Search in Google Scholar PubMed

[52] Hauser W, Walitt B, Fitzcharles MA, Sommer C. Review of pharmacological therapies in fibromyalgia syndrome. Arthritis Res Ther 2014;16:201.10.1186/ar4441Search in Google Scholar PubMed PubMed Central

[53] Nijs J, Kosek E, Van Oosterwijck J, Meeus M. Dysfunctional endogenous analgesia during exercise in patients with chronic pain: to exercise or not to exercise? Pain Physician 2012;15(3 Suppl):ES205–13.10.36076/ppj.2012/15/ES205Search in Google Scholar

[54] Bennett M, Closs S. Methodological issues in nonpharamacological trials for chronic pain. Pain Clinical Updates 2010;18:1–6.Search in Google Scholar

[55] SBU. Methods for treatment of chronic pain a systematic review of the literature. In: Swedish: Metoder för behandling av långvarig smärta: en systematisk litteraturöversikt. Stockholm: SBU – Swedish Council on Health Technology Assessment, 2006;177:1–2.Search in Google Scholar

[56] SBU. Rehabilitation of chronic pain [In Swedish: Rehabilitering vid långvarig smärta. En systematisk litteraturöversikt]. In: SBU-rapport. Stockholm: SBU – Swedish Council on Health Technology Assessment, 2010.Search in Google Scholar

[57] Scascighini L, Toma V, Dober-Spielmann S, Sprott H. Multidisciplinary treatment for chronic pain: a systematic review of interventions and outcomes. Rheumatology 2008;47:670–8.10.1093/rheumatology/ken021Search in Google Scholar PubMed

[58] Weiner SS, Nordin M. Prevention and management of chronic back pain. Best Pract Res Clin Rheumatol 2010;24:267–79.10.1016/j.berh.2009.12.001Search in Google Scholar PubMed

[59] Kamper SJ, Apeldoorn AT, Chiarotto A, Smeets RJ, Ostelo RW, Guzman J, van Tulder MW. Multidisciplinary biopsychosocial rehabilitation for chronic low back pain: cochrane systematic review and meta-analysis. Br Med J 2015;350:h444.10.1136/bmj.h444Search in Google Scholar PubMed PubMed Central

[60] Norlund A, Ropponen A, Alexanderson K. Multidisciplinary interventions: review of studies of return to work after rehabilitation for low back pain. J Rehabil Med 2009; 41:115–21.10.2340/16501977-0297Search in Google Scholar PubMed

[61] Busch H, Bodin L, Bergstrom G, Jensen IB. Patterns of sickness absence a decade after pain-related multidisciplinary rehabilitation. Pain 2011;152:1727–33.10.1016/j.pain.2011.02.004Search in Google Scholar PubMed

[62] Jensen IB, Busch H, Bodin L, Hagberg J, Nygren A, Bergstrom G. Cost effectiveness of two rehabilitation programmes for neck and back pain patients: a seven year follow-up. Pain 2009;142:202–8.10.1016/j.pain.2008.12.015Search in Google Scholar PubMed

[63] Wang H, Buchner M, Moser MT, Daniel V, Schiltenwolf M. The role of IL-8 in patients with fibromyalgia: a prospective longitudinal study of 6 months. Clin J Pain 2009;25:1–4.10.1097/AJP.0b013e31817e13a3Search in Google Scholar PubMed

[64] Hysing EB, Smith L, Thulin M, Karlsten R, Bothelius K, Gordh T. Detection of systemic inflammation in severely impaired chronic pain patients and effects of a multimodal pain rehabilitation program. Scand J Pain 2019;19:235–44.10.1515/sjpain-2018-0340Search in Google Scholar PubMed

[65] Lasselin J, Kemani MK, Kanstrup M, Olsson GL, Axelsson J, Andreasson A, Lekander M, Wicksell RK. Low-grade inflammation may moderate the effect of behavioral treatment for chronic pain in adults. J Behav Med 2016;39:916–24.10.1007/s10865-016-9769-zSearch in Google Scholar PubMed PubMed Central

[66] Teixeira AA, Lira FS, Pimentel GD, Oliveira de Souza C, Batatinha H, Biondo LA, Yamashita AS, Junior EA, Neto JC. Aerobic exercise modulates the free fatty acids and inflammatory response during obesity and cancer cachexia. Crit Rev Eukaryot Gene Expr 2016;26:187–98.10.1615/CritRevEukaryotGeneExpr.2016016490Search in Google Scholar PubMed

[67] Kawi J, Lukkahatai N, Inouye J, Thomason D, Connelly K. Effects of exercise on select biomarkers and associated outcomes in chronic pain conditions: systematic review. Biol Res Nurs 2016;18:147–59.10.1177/1099800415599252Search in Google Scholar PubMed

[68] Andrade A, Vilarino GT, Sieczkowska SM, Coimbra DR, Steffens RAK, Vietta GG. Acute effects of physical exercises on the inflammatory markers of patients with fibromyalgia syndrome: a systematic review. J Neuroimmunol 2018;316:40–9.10.1016/j.jneuroim.2017.12.007Search in Google Scholar PubMed

[69] Sanada K, Diez MA, Valero MS, Perez-Yus MC, Demarzo MM, Garcia-Toro M, Garcia-Campayo J. Effects of non-pharmacological interventions on inflammatory biomarker expression in patients with fibromyalgia: a systematic review. Arthritis Res Ther 2015;17:272.10.1186/s13075-015-0789-9Search in Google Scholar PubMed PubMed Central

[70] Bromley Milton M, Borsbo B, Rovner G, Lundgren-Nilsson A, Stibrant-Sunnerhagen K, Gerdle B. Is pain intensity really that important to assess in chronic pain patients? A study based on the Swedish quality registry for pain rehabilitation (SQRP). PLoS One 2013;8:e65483.10.1371/journal.pone.0065483Search in Google Scholar PubMed PubMed Central

[71] Gerdle B, Molander P, Stenberg G, Stalnacke BM, Enthoven P. Weak outcome predictors of multimodal rehabilitation at one-year follow-up in patients with chronic pain-a practice based evidence study from two SQRP centres. BMC Musculoskelet Disord 2016;17:490.10.1186/s12891-016-1346-7Search in Google Scholar PubMed PubMed Central

[72] Zigmond AS, Snaith RP. The hospital anxiety and depression scale. Acta Psychiatr Scand 1983;67:361–70.10.1111/j.1600-0447.1983.tb09716.xSearch in Google Scholar PubMed

[73] Bjelland I, Dahl AA, Haug TT, Neckelmann D. The validity of the hospital anxiety and depression scale. An updated literature review. J Psychosom Res 2002;52:69–77.10.1016/S0022-3999(01)00296-3Search in Google Scholar PubMed

[74] Lisspers J, Nygren A, Soderman E. Hospital Anxiety and Depression Scale (HAD): some psychometric data for a Swedish sample. Acta Psychiatr Scand 1997;96:281–6.10.1111/j.1600-0447.1997.tb10164.xSearch in Google Scholar PubMed

[75] Wheelock AM, Wheelock CE. Trials and tribulations of omics data analysis: assessing quality of SIMCA-based multivariate models using examples from pulmonary medicine. Mol Biosyst 2013;9:2589–96.10.1039/c3mb70194hSearch in Google Scholar PubMed

[76] Eriksson L, Byrne T, Johansson E, Trygg J, Vikström C. Multi- and megavariate data analysis: basic principles and applications, 3rd revised ed. Malmö: MKS Umetrics AB, 2013.Search in Google Scholar

[77] Backryd E, Tanum L, Lind AL, Larsson A, Gordh T. Evidence of both systemic inflammation and neuroinflammation in fibromyalgia patients, as assessed by a multiplex protein panel applied to the cerebrospinal fluid and to plasma. J Pain Res 2017;10:515–25.10.2147/JPR.S128508Search in Google Scholar PubMed PubMed Central

[78] Moen A, Lind AL, Thulin M, Kamali-Moghaddam M, Roe C, Gjerstad J, Gordh T. Inflammatory serum protein profiling of patients with lumbar radicular pain one year after disc herniation. Int J Inflam 2016;2016:3874964.10.1155/2016/3874964Search in Google Scholar PubMed PubMed Central

[79] Wang K, Bao JP, Yang S, Hong X, Liu L, Xie XH, Wu XT. A cohort study comparing the serum levels of pro- or anti-inflammatory cytokines in patients with lumbar radicular pain and healthy subjects. Eur Spine J 2016;25:1428–34.10.1007/s00586-015-4349-4Search in Google Scholar PubMed

[80] Alexander GM, Peterlin BL, Perreault MJ, Grothusen JR, Schwartzman RJ. Changes in plasma cytokines and their soluble receptors in complex regional pain syndrome. J Pain 2012;13:10–20.10.1016/j.jpain.2011.10.003Search in Google Scholar PubMed

[81] Singh L, Kaur A, Bhatti MS, Bhatti R. Possible molecular mediators involved and mechanistic insight into fibromyalgia and associated co-morbidities. Neurochem Res 2019;44:1517–32.10.1007/s11064-019-02805-5Search in Google Scholar PubMed

[82] Palada V, Ahmed AS, Finn A, Berg S, Svensson CI, Kosek E. Characterization of neuroinflammation and periphery-to-CNS inflammatory cross-talk in patients with disc herniation and degenerative disc disease. Brain Behav Immun 2019;75:60–71.10.1016/j.bbi.2018.09.010Search in Google Scholar PubMed

[83] Ortega E, Garcia JJ, Bote ME, Martin-Cordero L, Escalante Y, Saavedra JM, Northoff H, Giraldo E. Exercise in fibromyalgia and related inflammatory disorders: known effects and unknown chances. Exerc Immunol Rev 2009;15:42–65.Search in Google Scholar

[84] Lenz M, Uceyler N, Frettloh J, Hoffken O, Krumova EK, Lissek S, Reinersmann A, Sommer C, Stude P, Waaga-Gasser AM, Tegenthoff M, Maier C. Local cytokine changes in complex regional pain syndrome type I (CRPS I) resolve after 6 months. Pain 2013;154:2142–9.10.1016/j.pain.2013.06.039Search in Google Scholar PubMed

[85] Kitahara CM, Trabert B, Katki HA, Chaturvedi AK, Kemp TJ, Pinto LA, Moore SC, Purdue MP, Wentzensen N, Hildesheim A, Shiels MS. Body mass index, physical activity, and serum markers of inflammation, immunity, and insulin resistance. Cancer Epidemiol Biomarkers Prev 2014;23:2840–9.10.1158/1055-9965.EPI-14-0699-TSearch in Google Scholar PubMed PubMed Central

[86] Hochreiter WW, Nadler RB, Koch AE, Campbell PL, Ludwig M, Weidner W, Schaeffer AJ. Evaluation of the cytokines interleukin 8 and epithelial neutrophil activating peptide 78 as indicators of inflammation in prostatic secretions. Urology 2000;56:1025–9.10.1016/S0090-4295(00)00844-XSearch in Google Scholar

[87] Sturgill J, McGee E, Menzies V. Unique cytokine signature in the plasma of patients with fibromyalgia. J Immunol Res 2014;2014:938576.10.1155/2014/938576Search in Google Scholar PubMed PubMed Central

[88] Lee MC, Saleh R, Achuthan A, Fleetwood AJ, Forster I, Hamilton JA, Cook AD. CCL17 blockade as a therapy for osteoarthritis pain and disease. Arthritis Res Ther 2018;20:62.10.1186/s13075-018-1560-9Search in Google Scholar PubMed PubMed Central

[89] Schernthaner C, Paar V, Wernly B, Pistulli R, Rohm I, Jung C, Figulla HR, Yilmaz A, Cadamuro J, Haschke-Becher E, Schulze PC, Hoppe UC, Lichtenauer M, Kretzschmar D. Elevated plasma levels of interleukin-16 in patients with acute myocardial infarction. Medicine (Baltimore) 2017;96:e8396.10.1097/MD.0000000000008396Search in Google Scholar PubMed PubMed Central

[90] Yang F, Sun W, Luo WJ, Yang Y, Yang F, Wang XL, Chen J. SDF1-CXCR4 Signaling Contributes to the Transition from Acute to Chronic Pain State. Mol Neurobiol 2017;54:2763–75.10.1007/s12035-016-9875-5Search in Google Scholar PubMed

[91] Luo X, Wang X, Xia Z, Chung SK, Cheung CW. CXCL12/CXCR4 axis: an emerging neuromodulator in pathological pain. Rev Neurosci 2016;27:83–92.10.1515/revneuro-2015-0016Search in Google Scholar PubMed

[92] Zhang Z, Cherryholmes G, Mao A, Marek C, Longmate J, Kalos M, Amand RP, Shively JE. High plasma levels of MCP-1 and eotaxin provide evidence for an immunological basis of fibromyalgia. Exp Biol Med (Maywood) 2008;233:1171–80.10.3181/0712-RM-328Search in Google Scholar PubMed

[93] Pandya JM, Lundell AC, Andersson K, Nordstrom I, Theander E, Rudin A. Blood chemokine profile in untreated early rheumatoid arthritis: CXCL10 as a disease activity marker. Arthritis Res Ther 2017;19:20.10.1186/s13075-017-1224-1Search in Google Scholar PubMed PubMed Central

[94] Fattori V, Borghi S, Verri WJ. IL-33/ST2 signaling boosts inflammation and pain. Proc Natl Acad Sci USA 2017;114:E10034–5.10.1073/pnas.1716120114Search in Google Scholar PubMed PubMed Central

[95] Fattori V, Hohmann MSN, Rossaneis AC, Manchope MF, Alves-Filho JC, Cunha TM, Cunha FQ, Verri WA, Jr. Targeting IL-33/ST2 signaling: regulation of immune function and analgesia. Expert opinion on therapeutic targets 2017;21:1141–52.10.1080/14728222.2017.1398734Search in Google Scholar PubMed

[96] Garcia JJ, Cidoncha A, Bote ME, Hinchado MD, Ortega E. Altered profile of chemokines in fibromyalgia patients. Ann Clin Biochem 2014;51(Pt 5):576–81.10.1177/0004563213506413Search in Google Scholar PubMed

[97] Furer V, Hazan E, Mor A, Segal M, Katav A, Aloush V, Elkayam O, George J, Ablin JN. Elevated Levels of Eotaxin-2 in Serum of Fibromyalgia Patients. Pain Res Manag 2018;2018:7257681.10.1155/2018/7257681Search in Google Scholar PubMed PubMed Central

[98] Gul E, Celik Kavak E. Eotaxin levels in patients with primary dysmenorrhea. J Pain Res 2018;11:611–3.10.2147/JPR.S146603Search in Google Scholar PubMed PubMed Central

[99] Lupia E, Goffi A, Bosco O, Montrucchio G. Thrombopoietin as biomarker and mediator of cardiovascular damage in critical diseases. Mediators Inflamm 2012;2012:390892.10.1155/2012/390892Search in Google Scholar PubMed PubMed Central

[100] Pluthero FG, Kahr WH. Platelet production: new players in the field. Blood 2016;127:797–9.10.1182/blood-2016-01-687707Search in Google Scholar PubMed

[101] Weber KT, Satoh S, Alipui DO, Virojanapa J, Levine M, Sison C, Quraishi S, Bloom O, Chahine NO. Exploratory study for identifying systemic biomarkers that correlate with pain response in patients with intervertebral disc disorders. Immunol Res 2015;63:170–80.10.1007/s12026-015-8709-2Search in Google Scholar PubMed PubMed Central

[102] Gerdle B, Akerblom S, Brodda Jansen G, Enthoven P, Ernberg M, Dong HJ, Stalnacke BM, Ang BO, Boersma K. Who benefits from multimodal rehabilitation – an exploration of pain, psychological distress, and life impacts in over 35,000 chronic pain patients identified in the Swedish Quality Registry for Pain Rehabilitation. J Pain Res 2019;12:891–908.10.2147/JPR.S190003Search in Google Scholar PubMed PubMed Central

[103] Dworkin RH, Turk DC, Farrar JT, Haythornthwaite JA, Jensen MP, Katz NP, Kerns RD, Stucki G, Allen RR, Bellamy N, Carr DB, Chandler J, Cowan P, Dionne R, Galer BS, Hertz S, Jadad AR, Kramer LD, Manning DC, Martin S, et al. Core outcome measures for chronic pain clinical trials: IMMPACT recommendations. Pain 2005;113:9–19.10.1016/j.pain.2004.09.012Search in Google Scholar PubMed

[104] Turk DC, Dworkin RH, Allen RR, Bellamy N, Brandenburg N, Carr DB, Cleeland C, Dionne R, Farrar JT, Galer BS, Hewitt DJ, Jadad AR, Katz NP, Kramer LD, Manning DC, McCormick CG, McDermott MP, McGrath P, Quessy S, Rappaport BA,et al. Core outcome domains for chronic pain clinical trials: IMMPACT recommendations. Pain 2003;106:337–45.10.1016/j.pain.2003.08.001Search in Google Scholar PubMed

[105] Kaiser U, Kopkow C, Deckert S, Neustadt K, Jacobi L, Cameron P, De Angelis V, Apfelbacher C, Arnold B, Birch J, Bjarnegard A, Christiansen S, Williams CdCA, Gossrau G, Heinks A, Huppe M, Kiers H, Kleinert U, Martelletti P, McCracken L, et al. Developing a core outcome-domain set to assessing effectiveness of interdisciplinary multimodal pain therapy: the VAPAIN consensus statement on core outcome-domains. Pain 2017;159:673–83.10.1097/j.pain.0000000000001129Search in Google Scholar PubMed


Supplementary Material

The online version of this article offers supplementary material (https://doi.org/10.1515/sjpain-2019-0088).


Received: 2019-06-26
Revised: 2019-08-30
Accepted: 2019-09-10
Published Online: 2019-10-04
Published in Print: 2019-12-18

©2020 Scandinavian Association for the Study of Pain. Published by Walter de Gruyter GmbH, Berlin/Boston. All rights reserved.

Downloaded on 20.5.2024 from https://www.degruyter.com/document/doi/10.1515/sjpain-2019-0088/html
Scroll to top button