Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Jun 19.
Published in final edited form as: J Pain. 2022 Sep 15;24(1):84–97. doi: 10.1016/j.jpain.2022.08.007

Perturbations in Neuroinflammatory Pathways Are Associated With a Worst Pain Profile in Oncology Patients Receiving Chemotherapy

Joosun Shin *, Kord M Kober *,#, Carolyn Harris *, Kate Oppegaard *, Alejandra Calvo-Schimmel *, Steven M Paul *, Bruce A Cooper *, Adam Olshen , Vasuda Dokiparthi *, Yvette P Conley , Marilyn Hammer §, Jon D Levine , Christine Miaskowski *,†,#
PMCID: PMC11186595  NIHMSID: NIHMS1998748  PMID: 36115520

Abstract

Unrelieved pain occurs in 55% of cancer patients. Identification of molecular mechanisms for pain may provide insights into therapeutic targets. Purpose was to evaluate for perturbations in neuroinflammatory pathways between oncology patients with and without severe pain. Worst pain severity was rated using a 0 to 10 numeric rating scale six times over two cycles of chemotherapy. Latent profile analysis was used to identify subgroups of patients with distinct pain profiles. Pathway impact analyses were performed in two independent samples using gene expression data obtained from RNA sequencing (n = 192) and microarray (n = 197) technologies. Fisher’s combined probability test was used to identify significantly perturbed pathways between None versus the Severe pain classes. In the RNA sequencing and microarray samples, 62.5% and 56.3% of patients were in the Severe pain class, respectively. Nine perturbed pathways were related to neuroinflammatory mechanisms (i.e., retrograde endocannabinoid signaling, gamma-aminobutyric acid synapse, glutamatergic synapse, Janus kinase-signal transducer and activator of transcription signaling, phagosome, complement and coagulation cascades, cytokine-cytokine receptor interaction, chemokine signaling, calcium signaling). First study to identify perturbations in neuroinflammatory pathways associated with severe pain in oncology outpatients. Findings suggest that complex neuroimmune interactions are involved in the maintenance of chronic pain conditions.

Perspective:

In this study that compared oncology patients with none versus severe pain, nine perturbed neuroinflammatory pathways were identified. Findings suggest that complex neuroimmune interactions are involved in the maintenance of persistent pain conditions.

Keywords: Cancer, chemotherapy, cytokines, gene expression, gamma amino butyric acid, glutamine, neuro-immune interactions, neuroinflammation


Approximately 70% of patients experience moderate to severe pain during chemotherapy.56,64 Pain can be related to the cancer itself, associated with treatment (e.g., mucositis, peripheral neuropathy), or be related to other chronic conditions (e.g., back pain, osteoarthritis). In our previous study of patients undergoing chemotherapy,64 of the 926 patients evaluated, 20.8% reported only non-cancer pain, 37.7% reported only cancer pain, and 41.5% reported both types of pain. In the context of the opioid epidemic, recent evidence suggests that the undertreatment of pain in oncology patients remains a significant clinical problem.43 One of the gaps in effective management of multiple pain problems in oncology patients is an incomplete understanding of the mechanisms that underlie chronic pain. While direct neuronal activation is involved in the development and maintenance of chronic pain,22 emerging evidence suggests a role for neuroinflammation.14,32,49 The bidirectional communication between the immune and the nervous systems may provide opportunities to develop more targeted interventions for pain.27

Neuroinflammation plays a fundamental role in mediating neuronal plasticity.32 As part of this process, activation of cytokines results in peripheral and central sensitization and the development of chronic pain,28 including chronic cancer and non-cancer pain.71 As noted in one review,86 the transition from acute to chronic pain involves prolonged innate and adaptive immune signaling that induces maladaptive neuronal plasticity within the peripheral and central nervous systems.32,71 However, while in a study of breast cancer survivors,52 we identified perturbations in neuroinflammatory pathways associated with chemotherapy-induced peripheral neuropathy (CIPN), no studies have evaluated for these types of perturbations in oncology patients with severe pain during chemotherapy. Therefore, the purpose of this study, using the results of a previous latent profile analysis (LPA) that identified four classes of patients with distinct pain profiles (i.e., None, Mild, Moderate, Severe),74 was to use an extreme phenotype approach, to evaluate for differentially perturbed pathways associated with neuroinflammation between the None and the Severe pain classes.

Methods

Patients and Settings

This study is part of a larger, longitudinal study of the symptom experience of oncology outpatients receiving chemotherapy.60,75 Eligible patients were ≥18 years of age; had a diagnosis of breast, gastrointestinal, gynecological, or lung cancer; had received chemotherapy within the preceding four weeks; were scheduled to receive at least two additional cycles of chemotherapy; were able to read, write, and understand English; and gave written informed consent. Patients were recruited from two Comprehensive Cancer Centers, one Veteran’s Affairs hospital, and 4 community-based oncology programs.

Study Procedures

The study was approved by the Institutional Review Board at each of the study sites. Of the 2234 patients approached, 1343 consented to participate (60.1% response rate). The major reason for refusal was being overwhelmed with their cancer treatment. Eligible patients were approached in the infusion unit during their first or second cycle of chemotherapy by a member of the research team to discuss study participation and obtain written informed consent. Blood for ribonucleic acid (RNA) isolation was collected at the enrollment assessment. Medical records were reviewed for disease and treatment information. For this study, a total of 717 patients provided a blood sample for the analyses (Supplemental Figure 1). Of these 717 patients, 357 patients had their samples processed using RNA sequencing (i.e., RNA-seq sample) and 360 patients had their samples processed using microarray (i.e., microarray sample) technologies.

Instruments

Demographic and Clinical Characteristics

Demographic information was obtained using a self-report questionnaire. Functional status was assessed using the Karnofsky Performance Status (KPS) scale.37 The occurrence, treatment, and functional impact of 13 common medical conditions were assessed using the Self-Administered Comorbidity Questionnaire (SCQ).70 Alcohol consumption, behaviors, and associated problems were measured using the Alcohol Use Disorders Identification test (AUDIT).5 The toxicity of each patient’s chemotherapy regimen was rated using the MAX2 index.20,79 Medical records were reviewed for disease and treatment information.

Pain Measure

Worst pain severity was assessed using the Brief Pain Inventory (BPI).16 Patients were asked to indicate whether they were generally bothered by pain (yes/no). If they were generally bothered by pain, patients rated their worst pain severity in the past 24 hours using a 0 (no pain) to 10 (worst pain imaginable) numeric rating scale (NRS). Additional information was collected on causes of pain, as well as its duration, locations, and interference.

Data Analysis

Latent Profile Analysis

In our previous analysis,74 LPA was used to identify unobserved subgroups of patients (i.e., latent classes) with distinct worst pain profiles over the six assessments, using the patients’ ratings of worst pain severity. Before performing the LPA, patients who reported the occurrence of pain for ≤1 of the six assessments were identified and label as the “None” class (n = 371, 28.4%). Then, the LPA was performed on the remaining 934 patients using MPlus Version 8.4.58 Estimation was carried out with full information maximum likelihood with standard error and a Chi square test that are robust to non-normality and non-independence of observations (“estimator=MLR”). Model fit was evaluated to identify the solution that best characterized the observed latent class structure with the Bayesian Information Criterion,36 Vuong-Lo-Mendell-Rubin likelihood ratio test, entropy, and latent class percentages that were large enough to be reliable.58 Missing data were accommodated for with the use of the Expectation-Maximization (EM) algorithm.57 Three latent classes were identified based on clinically meaningful cutoff scores. For the current analysis, using an extreme phenotype approach, an evaluation of differentially perturbed pathways between patients in the None and Severe pain classes was performed.

Imputation Process

Missing data for demographic and clinical characteristics were imputed by the k-nearest-neighbors method, with k=9. For continuous variables the Euclidean distance was used to find the nearest neighbors. The imputed value was the weighted average of the nearest neighbors, with each weight originally exp(−dist(x,j)), after which the weights were scaled to one. For categorical variables, distance was 0 if the predictor and the neighbor had the same value and 1 if they did not. The imputed value was the mode of the nearest neighbors.

Demographic and Clinical Data

Demographic and clinical data from the two patient samples (i.e., RNA-seq, microarray) were analyzed separately. Differences in demographic and clinical characteristics between the patients in the None and Severe pain classes were evaluated using parametric and non-parametric tests. Significance corresponded to a p-value of <.05. Characteristics included in the final model were selected using a backwards stepwise logistic regression approach based on the likelihood ratio test (LRT). The area under the curve (AUC) of the receiver operating characteristic (ROC) curves was used to gauge the overall adequacy of the logistic regression model for each sample.17 All these analyses were performed using R (version 4.1).78

Differential Expression and Pathway Impact Analyses (PIA)

Details on the methods of the gene expression and pathway impact analyses are provided in Supplemental File 1. In brief, differential expression was quantified using empirical Bayes models that were implemented separately for each sample (i.e., using edgeR67 for the RNA-seq sample and limma77 for the microarray sample). These analyses were adjusted for demographic and clinical characteristics that were significantly different between the None versus Severe pain classes. In addition, the models included surrogate variables not associated with class memberships to adjust for variations due to unmeasured sources.44,45 Expression loci were annotated with Entrez gene identifiers. Gene symbols were derived from the HUGO Gene Nomenclature Committee resource database.29 The differential expression results were summarized as the log fold-change and p-value for each gene. Only genes that had a common direction of expression (i.e., the same sign for the log fold-change) across the two samples were retained for subsequent analyses. Common genes were matched using gene symbol.

To interpret the results in the context of pain-related mechanisms, we used PIA that included potentially important biological factors (e.g., gene-gene interactions, flow signals in a pathway, pathway topologies), as well as the magnitude (i.e., log fold-change) and P-values from the differential expression analysis for each sample.53 The PIA included the results of the differential expression analyses for all of the genes (i.e., cutoff free) that had a common direction of differential expression to determine probability of pathway perturbations (pPERT) using Pathway Express.18 A total of 225 signaling pathways were defined using the Kyoto Encyclopedia of Genes and Genomes (KEGG) database.3 For each sample, a separate test was performed for each pathway. Then, we used Fisher’s Combined Probability method to combine these test results to obtain a single test (global) of the null hypothesis.24,25 The significance of the combined transcriptome-wide PIA was assessed using a false discovery rate (FDR) of 0.015.19 Finally, we evaluated these results for perturbed neuroinflammatory signaling pathways.

Results

RNA-seq Performance

Of the 357 patients in the RNA-seq sample, 72 were in the None and 120 were in the Severe pain classes. Median library threshold size was 9,042,589 reads. Following the application of quality control filters, 10,881 genes were included in the final analysis. The common dispersion was estimated as 0.26493, yielding a biological coefficient of variation of 0.5147 well within the expected value for clinical samples.41,51

RNA Microarray Performance

Of the 360 patients in the microarray sample, 86 were in the None and 111 were in the Severe pain classes. All of these samples demonstrated good hybridization performance for biotin, background negative, and positive control assays on the arrays. Limma was used for background correction, quantile normalization, and log2 transformation.76 Following quality control filters, 46,542 loci were included in the final analysis.

Differences in Demographic and Clinical Characteristics

Of 192 patients with phenotypic data in the RNA seq sample (Table 1), compared to the None class, the Severe class was more likely to be female; had fewer years of education; a higher body mass index; a lower performance status; a higher number of comorbidities; and a higher comorbidity burden; were more likely to have adult care responsibilities; had a lower annual income, and were more likely to self-report diagnoses of anemia or back pain.

Table 1.

Differences in Demographic and Clinical Characteristics Between Patients in the None Versus the Severe Pain Classes in the RNA-sequencing Sample

Characteristic None 37.5% (n = 72) Severe 62.5% (n = 120) Statistics
Mean (SD) Mean (SD)

Age (years) 57.2 (12.3) 55.2 (12.2) t = 1.09, p = 0.277
Education (years) 16.4 (3.5) 15.2 (2.9) t = 2.53, p = 0.012
Body mass index (kg/m2) 25.5 (5.6) 27.6 (6.7) t =−2.22, p = 0.028
Karnofsky Performance Status score 82.4 (13.3) 72.9 (11.6) t = 5.16, p < 0.001
Number of comorbidities 2.2 (1.6) 3.0 (1.6) t = −3.49, p < 0.001
Self-administered Comorbidity Questionnaire score 5.0 (3.3) 7.2 (4.1) t = −3.94, p < 0.001
Alcohol Use Disorders Identification Test score 2.8 (1.4) 2.7 (1.9) t = 0.29, p = 0.774
Time since diagnosis (years) 2.1 (3.7) 1.2 (2.2) U, p = 0.443
Time since diagnosis (years, median) 0.45 0.44
Number of prior cancer treatments 1.5 (1.3) 1.5 (1.4) t =−0.24, p = 0.811
Number of metastatic sites including lymph node involvement 1.2 (1.2) 1.2 (1.3) t = 0.16, p = 0.871
Number of metastatic sites excluding lymph node involvement 0.7 (1.0) 0.7 (1.1) t =−0.02, p = 0.986
Hemoglobin (g/dL) 11.6 (1.5) 11.3 (1.4) t = 1.19, p = 0.234
Hematocrit (%) 34.7 (4.4) 34.1 (4.0) t = 0.92, p = 0.361
MAX2 score 0.17 (0.08) 0.19 (0.08) t = −1.43, p = 0.155

% (n) % (n)

Gender FE, p = 0.007
 Female 68.1 (49) 85.0 (102)
 Male 31.9 (23) 15.0 (18)
Ethnicity X2 = 3.67; p = 0.300
 White 70.8 (51) 58.3 (70)
 Black 5.6 (4) 10.0 (12)
 Asian or Pacific Islander 11.1 (8) 11.7 (14)
 Hispanic mixed or other 12.5 (9) 20.0 (24)
Married or partnered (% yes) 61.1 (44) 54.2 (65) FE, p = 0.370
Lives alone (% yes) 23.6 (17) 31.7 (38) FE, p = 0.253
Childcare responsibilities (% yes) 26.4 (19) 19.2 (23) FE, p = 0.281
Adult care responsibilities (% yes) 1.4 (1) 11.7 (14) FE, p = 0.011
History of premature birth (% yes) 2.8 (2) 5.8 (7) FE, p = 0.488
Currently employed (% yes) 34.7 (25) 26.7 (32) FE, p = 0.256
Income U, p < 0.001
 <$30,000 11.1 (8) 38.3 (46)
 $30,000 to <$70,000 15.3 (11) 23.3 (28)
 $70,000 to <$100,000 22.2 (16) 15.0 (18)
 ≥$100,000 51.4 (37) 23.3 (28)
Specific comorbidities (% yes)
 Heart disease 4.2 (3) 8.3 (10) FE, p = 0.377
 High blood pressure 29.2 (21) 36.7 (44) FE, p = 0.345
 Lung disease 11.1 (8) 12.5 (15) FE, p = 0.823
 Diabetes 11.1 (8) 15.8 (19) FE, p = 0.400
 Ulcer or stomach disease 2.8 (2) 7.5 (9) FE, p = 0.214
 Kidney disease 1.4 (1) 0.8 (1) FE, p = 1.000
 Liver disease 9.7 (7) 4.2 (5) FE, p = 0.136
 Anemia or blood disease 4.2 (3) 14.2 (17) FE, p = 0.029
 Depression 18.1 (13) 29.2 (35) FE, p = 0.121
 Osteoarthritis 12.5 (9) 17.5 (21) FE, p = 0.416
 Back pain 16.7 (12) 51.7 (62) FE, p < 0.001
 Rheumatoid arthritis 1.4 (1) 6.7 (8) FE, p = 0.157
Exercise on a regular basis (% yes) 66.7 (48) 65.0 (78) FE, p = 0.876
Smoking current or history of (% yes) 38.9 (28) 35.0 (42) FE, p = 0.643
Cancer diagnosis X2 = 7.84, p = 0.049
 Breast 36.1 (26) 40.0 (48) NS
 Gastrointestinal 43.1 (31) 25.0 (30) NS
 Gynecological 11.1 (8) 19.2 (23) NS
 Lung 9.7 (7) 15.8 (19) NS
Type of prior cancer treatment X2 = 0.54, p = 0.909
 No prior treatment 25.0 (18) 26.7 (32)
 Only surgery, CTX, or RT 41.7 (30) 45.0 (54)
 Surgery & CTX, or surgery & RT, or CTX & RT 15.3 (11) 13.3 (16)
 Surgery & CTX & RT 18.1 (13) 15.0 (18)
CTX cycle length X2 = 3.03, p = 0.220
 14 day cycle 47.2 (34) 42.5 (51)
 21 day cycle 40.3 (29) 50.8 (61)
 28 day cycle 12.5 (9) 6.7 (8)
Emetogenicity of CTX X2 = 0.73, p = 0.695
 Minimal/low 15.3 (11) 19.2 (23)
 Moderate 62.5 (45) 62.5 (75)
 High 22.2 (16) 18.3 (22)
Antiemetic regimens X2 = 0.92, p = 0.821
 None 4.2 (3) 2.5 (3)
 Steroid alone or serotonin receptor antagonist alone 18.1 (13) 18.3 (22)
 Serotonin receptor antagonist and steroid 52.8 (38) 49.2 (59)
 NK-1 receptor antagonist and two other antiemetics 25.0 (18) 30.0 (36)

Abbreviations: CTX = chemotherapy; dL = deciliter; FE = Fisher’s exact test; g = grams; kg = kilograms; m2 = meter squared; NK-1 = neurokinin-1; NS = not significant; RNA = ribonucleic acid; RT = radiation therapy; SD = standard deviation; U = Mann-Whitney U test.

Of 197 patients with phenotypic data in the microarray sample (Table 2), compared to the None class, the Severe class had fewer years of education; a lower performance status; a higher number of comorbidities, a higher comorbidity burden, and a higher number of prior cancer treatments; were more likely to be not married or partnered; had lower annual income; were less likely to be employed; were less likely to exercise regularly; were more likely to have a current or previous history of smoking; and were more likely to self-reported diagnoses of anemia, depression, osteoarthritis, or back pain.

Table 2.

Differences in Demographic and Clinical Characteristics Between Patients in the None Versus Severe Pain Classes in the Microarray Sample

Characteristic None 43.7% (n = 86) Severe 56.3% (n= 111) Statistics
Mean (SD) Mean (SD)

Age (years) 57.2 (10.2) 54.3 (12.9) t = 1.67, p = 0.096
Education (years) 16.8 (3.1) 15.8 (2.8) t = 2.37, p = 0.019
Body mass index (kg/m2) 26.3 (6.4) 27.2 (5.8) t =−0.97, p = 0.333
Karnofsky Performance Status score 83.7 (10.6) 74.6 (11.5) t = 5.65, p < 0.001
Number of comorbidities 1.9 (1.1) 3.0 (1.5) t = −5.67, p < 0.001
Self-administered Comorbidity Questionnaire score 4.3 (2.3) 6.9 (3.4) t = −6.10, p < 0.001
Alcohol Use Disorders Identification Test score 3.0 (1.7) 2.8 (2.8) t = 0.56, p = 0.579
Time since diagnosis (years) 1.4 (2.7) 2.3 (3.7) U, p = 0.202
Time since diagnosis (years, median) 0.40 0.45
Number of prior cancer treatments 1.5 (1.5) 2.0 (1.7) t =−2.18, p = 0.030
Number of metastatic sites including lymph node involvement 1.0 (1.2) 1.4 (1.4) t = −1.71, p = 0.089
Number of metastatic sites excluding lymph node involvement 0.6 (1.1) 0.9 (1.2) t = −1.64, p = 0.103
Hemoglobin (g/dL) 11.7 (1.4) 11.6 (1.3) t = 0.52, p = 0.607
Hematocrit (%) 34.9 (4.0) 34.6 (3.7) t = 0.43, p = 0.670
MAX2 score 0.18 (0.09) 0.17 (0.08) t = 1.13, p = 0.261

% (n) % (n)

Gender FE, p = 0.182
 Female 79.1 (68) 86.5 (96)
 Male 20.9 (18) 13.5 (15)
Ethnicity X2 = 1.60, p = 0.659
 White 72.1 (62) 64.0 (71)
 Black 11.6 (5) 9.0 (10)
 Asian or Pacific Islander 5.8 (10) 14.4 (16)
 Hispanic, Mixed, or Other 10.5 (9) 12.6 (14)
Married or partnered (% yes) 80.2 (69) 54.1 (60) FE, p < 0.001
Lives alone (% yes) 15.1 (13) 20.7 (23) FE, p = 0.356
Childcare responsibilities (% yes) 23.3 (20) 27.0 (30) FE, p = 0.622
Adult care responsibilities (% yes) 5.8 (5) 13.5 (15) FE, p = 0.097
History of premature births (% yes) 4.7 (4) 5.4 (6) FE, p = 1.000
Currently employed (% yes) 45.3 (39) 24.3 (27) FE, p = 0.002
Income U, p < 0.001
 <$30,000 12.8 (11) 31.5 (35)
 $30,000 to <$70,000 14.0(12) 26.1 (29)
 $70,000 to <$100,000 16.3 (14) 14.4 (16)
 ≥$100,000 57.0 (49) 27.9 (31)
Specific comorbidities (% yes)
 Heart disease 3.5 (3) 8.1 (9) FE, p = 0.236
 High blood pressure 31.4 (27) 31.5 (35) FE, p = 1.000
 Lung disease 5.8 (5) 12.6 (14) FE, p = 0.145
 Diabetes 5.8 (5) 9.0 (10) FE, p = 0.434
 Ulcer or stomach disease 2.3 (2) 7.2 (8) FE, p = 0.191
 Kidney disease 1.2 (1) 1.8 (2) FE, p = 1.000
 Liver disease 5.8 (5) 6.3 (7) FE, p = 1.000
 Anemia or blood disease 8.1 (7) 22.5 (25) FE, p = 0.007
 Depression 10.5 (9) 34.2 (38) FE, p < 0.001
 Osteoarthritis 4.7 (4) 17.1 (19) FE, p = 0.007
 Back pain 5.8 (5) 44.1 (49) FE, p < 0.001
 Rheumatoid arthritis 1.2 (1) 3.6 (4) FE, p = 0.389
Exercise on a regular basis (% yes) 79.1 (68) 61.3 (68) FE, p = 0.008
Smoking current or history of (% yes) 20.9 (18) 40.5 (45) FE, p = 0.004
Cancer diagnosis X2 = 0.72, p = 0.869
 Breast 41.9 (36) 39.6 (44)
 Gastrointestinal 25.6 (22) 22.5 (25)
 Gynecological 19.8 (17) 24.3 (27)
 Lung 12.8 (11) 13.5 (15)
Type of prior cancer treatment X2 = 5.59, p = 0.133
 No prior treatment 24.4 (21) 17.1 (19)
 Only surgery, CTX, or RT 50.0 (43) 41.4 (46)
 Surgery & CTX, or surgery & RT, or CTX & RT 12.8 (11) 20.7 (23)
 Surgery & CTX & RT 12.8 (11) 20.7 (23)
CTX cycle length X2 = 1.34, p = 0.512
 14 day cycle 36.0 (31) 30.6 (34)
 21 day cycle 59.3 (51) 61.3 (68)
 28 day cycle 4.7 (4) 8.1 (9)
Emetogenicity of CTX X2 = 2.21, p = 0.331
Minimal/low 18.6 (16) 27.0 (30)
Moderate 58.1 (50) 55.0 (61)
High 23.3 (20) 18.0 (20)
Antiemetic regimens X2 = 3.28, p = 0.351
 None 11.6 (10) 8.1 (9)
 Steroid alone or serotonin receptor antagonist alone 24.4 (21) 19.8 (22)
 Serotonin receptor antagonist and steroid 47.7 (41) 45.9 (51)
 NK-1 receptor antagonist and two other antiemetics 16.3 (14) 26.1 (29)

Abbreviations: CTX = chemotherapy; dL = deciliter; FE = Fisher’s exact test; g = grams; kg = kilograms; m2 = meter squared; NK-1 = neurokinin-1; RT = radiation therapy; U = Mann-Whitney U test.

Differences in Pain Characteristics

As summarized in Table 3, no differences were found between the two Severe pain classes in any of the pain characteristics evaluated. In brief, the majority of the patients had both cancer and non-cancer pain; were experiencing chronic pain; had worst pain scores in the severe range; and reported moderate levels of interference.

Table 3.

Differences in Pain Characteristics Between Patients in the RNA-Sequencing Sample Compared to the Microarray Sample

Characteristic RNA-seq(n = 120) Microarray(n =111) Statistics
% (n) % (n)

Sources of pain
 Type of pain X2 = 0.29, p = .866
 Only non-cancer pain 15.8 (18) 14.2 (15)
 Only cancer pain 27.2 (31) 30.2 (32)
 Both cancer and non-cancer pain 57.0 (65) 55.7 (59)
Causes of non-cancer pain
 Headache 33.7 (28) 41.9 (31) FE, p= .324
 Low back pain 57.8 (48) 48.7 (36) FE, p = .266
 Fibromyalgia 4.8 (4) 9.5 (7) FE, p = .351
 Diabetic neuropathy 3.6 (3) 1.4 (1) FE, p = .623
 Arthritis 33.7 (28) 27.0 (20) FE, p = .390

Acute versus Chronic Pain

Length of time with non-cancer pain FE, p = .460
 Less than 3 months 19.4 (14) 17.1 (12)
 ≥3 months 80.6 (58) 82.9 (58)
Length of time with cancer pain FE, p = .829
 Less than 3 months 56.4 (53) 50.0 (45)
 ≥3 months 43.6 (41) 50.0 (45)

Pain Characteristics

Mean (SD) Mean (SD)

Pain intensity
 Now 3.2 (2.6) 2.8 (2.3) t = 1.10, p = .275
 Average 4.4 (2.0) 4.2 (1.8) t = 0.80, p = .422
 Worst 8.1 (1.3) 8.4 (1.5) t = −1.33, p = .185
Pain duration
 Number of days per week in pain 4.1 (2.3) 4.2 (2.2) t =−0.34, p = .731
 Number of hours per day in pain 9.7 (8.3) 10.3 (8.7) t =−0.56, p = .575
Pain locations
 Number of pain locations 11.3 (9.1) 11.5 (9.7) t =−0.14, p = .889
Pain interference
 General activity 4.9 (3.1) 4.9 (2.9) t = 0.04, p = .972
 Mood 4.4 (3.2) 4.8 (2.8) t =−0.79, p = .431
 Walking ability 4.6 (3.3) 4.4 (3.0) t = 0.48, p = .633
 Normal work 5.3 (3.3) 5.1 (2.9) t = 0.48, p = .634
 Relations with other people 3.4 (3.1) 3.5 (3.0) t =−0.33, p = .741
 Sleep 5.1 (3.2) 5.3 (2.9) t =−0.59, p = .556
 Enjoyment of life 5.0 (3.3) 5.0 (3.0) t = −0.11, p = .912
 Sexual activity 4.5 (4.2) 5.4 (4.1) t = −1.51, p = .133
 Mean pain interference score 4.6 (2.7) 4.8 (2.4) t =−0.59, p = .558
Pain frequency % (n) % (n)
 1 to 4 times per month 15.5 (16) 14.9 (15) U, p = .740
 Several times per week 18.5 (19) 17.8 (18)
 Multiple times per day 42.7 (44) 40.6 (41)
 Continuously 23.3 (24) 26.7 (27)

Abbreviations: FE = Fisher’s Exact test; RNA = ribonucleic acid; SD = standard deviation; seq = sequencing, U = Mann Whitney U test.

Logistic Regression Analyses

In the logistic regression analysis for the RNA-seq sample (Table 4), five variables were retained in the final model (i.e., gender, income, adult care responsibilities, KPS score, self-reported diagnosis of back pain) and were used as covariates in the gene expression analysis. Patients who were female, had a lower annual income, had adult care responsibilities, had a lower functional status, and self-reported a diagnosis of back pain were more likely to belong to the Severe class.

Table 4.

Multiple Logistic Regression Analyses Predicting Membership in the Severe Pain Class

RNA seq Sample (n = 192)

Predictors Odds Ratio 95% CI p-value

Gender (male) 0.42 0.18, 0.98 0.047
Income
 <$30,000 1.00
 $30,000 to <$70,000 0.43 0.13, 1.33 0.147
 $70,000 to <$100,000 0.22 0.07, 0.68 0.010
≥$100,000 0.17 0.06, 0.45 <0.001
Adult care responsibilities 10.96 1.75, 218.39 0.033
Karnofsky Performance Status score 0.96 0.93, 0.99 0.005
Self-reported diagnosis of back pain 2.98 1.36, 6.78 0.007
Overall model fit: AUC of the ROC = 0.820

Microarray Sample (n = 197)

Predictors Odds Ratio 95% CI p-value

Married or partnered 0.29 0.12, 0.66 0.003
Exercise on a regular basis 0.38 0.16, 0.87 0.022
Current or history of smoking 2.69 1.19, 6.32 0.018
Karnofsky Performance Status score 0.93 0.90, 0.96 < 0.001
Number of prior cancer treatments 1.37 1.07, 1.79 0.013
Self-reported diagnosis of anemia or blood disease 2.53 0.85, 8.17 0.096
Self-reported diagnosis of depression 3.60 1.34, 10.42 0.011
Self-reported diagnosis if back pain 9.15 3.21, 31.53 <0.001
Overall model fit: AUC of the ROC = 0.883

Abbreviations: AUC = Area under curve; CI = confidence interval; ROC = receiver operating characteristic.

In the logistic regression analysis for the microarray sample (Table 4), eight variables were retained in the final model (i.e., married/partnered, exercise on a regular basis, current or history of smoking, KPS score, number of prior cancer treatments, self-reported diagnoses of anemia, depression, back pain) and were used as covariates in the gene expression analysis. Patients who were not married or partnered did not exercise on a regular basis, had a current or history of smoking, had a lower KPS score, had a higher number of prior cancer treatments, and self-reported a diagnosis of depression or back pain were more likely to belong to the Severe class.

Perturbed Signaling Pathways Associated With Worst Pain Severity

Of the 13 surrogate variables identified for the RNA-seq sample, none were associated with class membership. The final differential expression model for this sample included 13 surrogate variables and five phenotypic characteristics. Of the 15 surrogate variables identified for microarray sample, two were associated with class membership and were excluded from the final model. The final differential expression model for this sample included 13 surrogate variables and eight phenotypic characteristics. For both samples, a total of 3,868 genes were included in the PIA analyses. Using Fisher’s Combined Probability method, across the two samples, 51 KEGG signaling pathways were significantly perturbed at an FDR of 0.015 (see Supplemental File 2). Of these, nine were related to neuroinflammatory mechanisms (Table 5).

Table 5.

Perturbed Neuroinflammatory KEGG Pathways Between Patients in the None Versus the Severe PaSin Classes

Pathway ID Pathway Name Combined Analysis Statistics

Neuroinflammatory Pathways
 hsa04060 Cytokine-cytokine receptor interaction X2 = 19.92, pPert = 0.008
 hsa04062 Chemokine signaling pathway X2 = 21.85, pPert = 0.005
 hsa04145 Phagosome X2 = 20.31, pPert = 0.007
 hsa04610 Complement and coagulation cascades X2 = 19.33, pPert = 0.008
 hsa04630 JAK-STAT signaling pathway X2 = 17.03, pPert = 0.012
Signal Transduction Pathway
 hsa04020 Calcium signaling pathway X2 = 22.84, pPert = 0.005
Neurotransmitter Pathways
 hsa04723 Retrograde endocannabinoid signaling X2 = 17.55, pPert = 0.011
 hsa04724 Glutamatergic synapse X2 = 21.91, pPert = 0.005
 hsa04727 GABAergic synapse X2 = 15.84, pPert = 0.014

Abbreviations: GABA = gamma-aminobutyric acid; JAK-STAT = Janus kinase-signal transducer and activator of transcription; KEGG = Kyoto Encyclopedia of Genes and Genomes; pPert = Combined perturbation P-value using Fisher’s Method adjusted using the Bonferroni method.

Discussion

This study is the first to provide evidence that suggests that perturbations in several neuroinflammatory pathways are associated with severe pain in oncology patients receiving chemotherapy. As noted in one review,63 a growing body of evidence suggests that both neurons and immune cells directly and indirectly detect and respond to painful stimuli and contribute to the initiation and maintenance of chronic pain. Our findings suggest that shared neuroinflammatory mechanisms may contribute to both cancer and non-cancer pain in oncology patients with severe pain. The remainder of this discussion focuses on the nine perturbed pathways identified in this study.

Complement and Coagulation Cascades Pathway

As noted in a recent review,82 complement signaling is important in directing neuronal responses to tissue injury and nerve trauma. After an acute injury, a complex interplay occurs between nociceptive neurons and immune cells to promote healing and facilitate guarding of the site of injury. The complement works by activating immune cells and stimulates these cells to release inflammatory mediators. However, in the setting of chronic pain, persistent or unbalanced signaling of complement factors occurs.82 In fact elevated levels of several key complement factors (e.g., C3a, C5, C5a) were found in patients with rheumatoid arthritis35,40,59 and osteoarthritis.81,87

Phagosome Pathway

While little is known about the interaction between phagocytes and nociceptor signaling, emerging evidence suggests that phagocytes (i.e., macrophage, dendritic cells) can contribute to the development and maintenance of pain.27 For example, macrophages release a variety of immune mediators that bind to receptors on nociceptors. This binding induces neuronal hyperexcitability and hypersensitivity.27 In addition, microglia play an essential role in the initiation of neuroinflammation by releasing the complement components C1q and C3 that induce phagocytosis by binding to neuronal surfaces.9 For example, following high-frequency stimulation-induced spinal long-term potentiation in rats,94 the number of activated microglia in the dorsal and ventral horn increased, which suggests an association between microglial activation, spinal plasticity, and chronic pain hypersensitivity. In addition, in a recent preclinical study,93 blockade of spinal microglia function significantly attenuated neuropathic pain through the inhibition of neuroinflammation.

Cytokine-cytokine Receptor Interaction Pathway

As noted in one review,49 in response to tissue injury, nociceptors induced a number of pro- and anti-inflammatory mediators that directly bind to and activate cytokine receptors. This bidirectional interaction between pain and inflammation leads to hyperexcitability and hypersensitivity of nociceptor neurons (i.e., peripheral sensitization).33 For example, in both preclinical and clinical studies, interleukin (IL)-6, tumor necrosis factor-alpha (TNF-α), and IL-1β appear to be involved in the development and maintenance of pain associated with cancer,21,72 rheumatoid arthritis,1 and peripheral neuropathies.83,92 In contrast, in a recent preclinical study,42 IL-10 attenuated pain hypersensitivity following cisplatin administration, which suggests that IL-10 may decrease neuroinflammation. In another preclinical study,90 IL-4 receptor knockout mice showed upregulation of pro-inflammatory mediators. Furthermore, cytokine signaling in the periphery is transmitted to the central terminals of the nociceptors and the brain.28 As a result, pro-inflammatory cytokines activate microglia that contributes to the development and maintenance of central sensitization.39,93 For example, following sciatic nerve chronic constriction injury, differentially perturbed cytokine-cytokine receptor interaction pathways were found in dorsal horn tissues.13

Chemokine Signaling Pathway

Chemokines are chemotactic cytokines that control the movement of circulating peripheral immune cells (e.g., T lymphocytes, natural killer cells, B cells, dendritic cells) by mediating cell-to-cell communication.34 In addition, they activate G-protein-coupled receptors.10,28 The chemokine (C-X-C motif) ligand-(CXCL) receptor pair serves as a mediator for glia-neuron communication61,95 that when activated leads to persistent hyperexcitability and neuroplasticity in peripheral nociceptors.28 Subsequently, this chemokine signaling alters nociceptive transduction through activation of chemokine receptors in dorsal root ganglia (DRG) cells.47 As noted in one review of rodent models of neuropathic pain,85 upregulation of the expression of C-C motif chemokine ligand 2 (CCL2) and its receptor (CCR2) in DRG neurons was identified. In addition, in another preclinical study of autologous nucleus pulposus-induced pain,95 increases in chemokine CCL2/CCR2 signaling in DRG and spinal cord were associated with the maintenance of lumbar disc herniation-induced pain. Of note, the administration of CCR2 antagonist decrease mechanical allodynia.

Additional preclinical studies provide evidence that chemokines and their receptors play a crucial role in cancer pain,61,62,89 visceral pain,4 and inflammatory pain.11,88 For example, chemokines appear to be involved in the regulation of neuronal excitability, neurotransmitter release, and neuronal survival84 by enhancing the activity of the N-methyl-D-aspartate (NMDA) receptors in dorsal horn neurons.26 In a recent preclinical study of inflammatory pain,12 CXCL1/CXCR2 signaling induced an enhancement of NMDA-induced currents in spinal cord neurons. The authors suggested that CXCL1/CXCR2 drives hyperactivity of NMDA receptors, which in turn mediates persistent inflammatory pain through the induction and maintenance of central sensitization.12

Janus Kinase-signal Transducer and Activator of Transcription (JAK-STAT) Signaling Pathway

As noted in one review on the association between the JAK-STAT signaling pathway and pain,10 this pathway is involved in both pro-and anti-nociceptive mechanisms through numerous inflammatory responses. For example, IL-6 binding to the IL-6 receptor induces the activation of the JAK-STAT transduction pathway.54 Phosphorylated JAK1 and 2 and STAT3 are translocated to the nucleus, leading to the expression of target genes and an increase in the release of proinflammatory cytokines.55,92 In contrast, IL-4 binding to the IL-4 receptor results in the activation of JAKs 1 and 3 and consequently STAT6, which in turn leads to the inhibition of the production of proinflammatory cytokines.10 In fact, a JAK-STAT inhibitor was approved by Food and Drug Administration and is used to treat rheumatoid arthritis.31 Given the positive findings across other studies,2,48,65 the use of a JAK-STAT inhibitor may warrant investigation in oncology patients with severe pain.

Calcium Signaling Pathway

Primary afferent neurons express multiple types of voltage-gated calcium channels (VGCCs), including N- and T-type channels.8 Calcium signaling mediated by these VGCCs is involved in the development and maintenance of chronic pain, including neuropathic80 and inflammatory73 pain, through the induction of a multifaceted cascade of signaling molecules.30 This calcium signaling-related cascade begins with an influx of calcium ions into the post-synaptic neuron through NMDA and a-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptors for glutamate and/or VGCCs.8 Subsequently, modulation of the ion channel pool induces sensitization and hyperexcitability of sensory neurons by releasing glutamate, an excitatory neurotransmitter at central nerve endings.30,50

Of note, several preclinical studies suggest that N-type calcium channel (Cav2.2) knockout mice have attenuation of inflammatory and neuropathic pain.69 Upregulation of N-type calcium channels in rat DRG neurons was associated with neuropathic pain.46,91 In addition, the deletion of the T-type calcium channel genes (e.g., Cav3.2) in rats was associated with major antinociceptive effects.7,15 L-type calcium channels (Cav1.2 and Cav1.3) are primarily located on post-synaptic channels that are involved in dorsal horn hyperexcitability and short- and long-term neuronal plasticity.68 As demonstrated in a preclinical study,66 L-type calcium channels contribute to the integration of afferent inputs and the maintenance of hyperexcitability in DRG neurons by controlling plateau potentials.

Retrograde Endocannabinoid Signaling Pathway

Retrograde endocannabinoid signaling is implicated in several forms of short- and long-term synaptic plasticity.6 The endocannabinoid system includes cannabinoid receptor subtypes 1 (CB1) and 2 (CB2), as well as their ligands, namely endocannabinoids.6 The endocannabinoids synthesized in response to activation of post-synaptic metabotropic glutamate receptors (mGluRs) travel retrogradely to bind CB1 receptors and impede neurotransmitter release through inhibition of presynaptic VGCCs.38 In addition, retrograde endocannabinoid signaling decreases presynaptic neurotransmitter release and balances glutamate/GABAergic transmission.38 Interestingly, while CB2 receptors are not found in the healthy brain, upregulation of CB2 receptors on microglia appears to be induced by neuroinflammatory processes.38 Therefore, increased endocannabinoid signaling may be associated with anti-inflammatory and neuroprotective phenotypes in microglia that suggests the therapeutic potential of targeting CB1 or CB2 receptors.23

Strengths and Limitations

While some limitations warrant consideration in that detailed information on the causes of cancer pain and analgesic use were not available for our patients, this study had a relatively large sample size and used LPA to identify distinct pain profiles. In addition, our sample represents the clinical reality in that oncology patients experience cancer and/or non-cancer pain. Additional strengths of this study include the performance of rigorous quality controls; utilization of two complementary methods to measure gene expression; the provision of results from independent tests across two samples; and the use of strict criteria for the selection of the perturbed neuroinflammatory pathways.

Conclusions

Our findings suggest that neuroimmune interactions are involved in the maintenance of chronic pain conditions in patients with cancer who are receiving chemotherapy. Our findings provide new evidence for potential therapeutic targets for the management of moderate to severe pain in oncology patients receiving chemotherapy. However, while no differences in cancer types and toxicity of the chemotherapy regimens was found between the None and Severe classes, additional research is warranted on the potential effects of the underlying tumor biology. Given that a growing body of evidence suggests a role for interactions between neuroimmune and endocrine systems in the maintenance of chronic pain, our subsequent study will evaluate for associations between pain in oncology patients receiving chemotherapy and perturbations in endocrine pathways.

Supplementary Material

Supp Data 4
Supp Data 2
Supp Data 3
Supp Data 1

Acknowledgments

We would like to acknowledge the support of Anatol Sucher and Ritu Roy on this project.

Disclosures:

This study was supported by a grant from the National Cancer Institute (CA134900). Dr. Miaskowski is an American Cancer Society Clinical Research Professor. Dr. Calvo-Schimmel and Ms. Harris and Oppegaard are supported by a T32 grant (NR016920) from the National Institute of Nursing Research. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Footnotes

Conflicts of interest: The authors have no conflicts of interest to declare.

Supplementary data

Supplementary data related to this article can be found at https://doi.org/10.1016/j.jpain.2022.08.007.

References

  • 1.Abdel Meguid MH, Hamad YH, Swilam RS, Barakat MS: Relation of interleukin-6 in rheumatoid arthritis patients to systemic bone loss and structural bone damage. Rheumatol Int 33:697–703, 2013 [DOI] [PubMed] [Google Scholar]
  • 2.Al-Massri KF, Ahmed LA, El-Abhar HS: Pregabalin and lacosamide ameliorate paclitaxel-induced peripheral neuropathy via inhibition of JAK/STAT signaling pathway and Notch-1 receptor. Neurochem Int 120:164–171, 2018 [DOI] [PubMed] [Google Scholar]
  • 3.Aoki-Kinoshita KF, Kanehisa M: Gene annotation and pathway mapping in KEGG. Methods Mol Biol (Clifton, N.J 396:71–91, 2007 [DOI] [PubMed] [Google Scholar]
  • 4.Bicer F, Altuntas CZ, Izgi K, Ozer A, Kavran M, Tuohy VK, Daneshgari F: Chronic pelvic allodynia is mediated by CCL2 through mast cells in an experimental autoimmune cystitis model. American J Physiol-Renal Physiol 308:F103–F113, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Bohn MJ, Babor TF, Kranzler HR: The Alcohol Use Disorders Identification Test (AUDIT): Validation of a screening instrument for use in medical settings. J Stud Alcohol 56:423–432, 1995 [DOI] [PubMed] [Google Scholar]
  • 6.Bouchet CA, Ingram SL: Cannabinoids in the descending pain modulatory circuit: Role in inflammation. Pharmacol Ther 209:107495, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Bourinet E, Alloui A, Monteil A, Barrère C, Couette B, Poirot O, Pages A, McRory J, Snutch TP, Eschalier A, Nargeot J: Silencing of the Cav3.2 T-type calcium channel gene in sensory neurons demonstrates its major role in nociception. Embo J 24:315–324, 2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Bourinet E, Altier C, Hildebrand ME, Trang T, Salter MW, Zamponi GW: Calcium-permeable ion channels in pain signaling. Physiol Rev 94:81–140, 2014 [DOI] [PubMed] [Google Scholar]
  • 9.Brown GC, Neher JJ: Microglial phagocytosis of live neurons. Nat Rev Neurosci 15:209–216, 2014 [DOI] [PubMed] [Google Scholar]
  • 10.Busch-Dienstfertig M, González-Rodríguez S: IL-4, JAK-STAT signaling, and pain. Jakstat 2:e27638, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Cao D-L, Qian B, Zhang Z-J, Gao Y-J, Wu X-B: Chemokine receptor CXCR2 in dorsal root ganglion contributes to the maintenance of inflammatory pain. Brain Res Bull 127:219–225, 2016 [DOI] [PubMed] [Google Scholar]
  • 12.Cao D-L, Zhang Z-J, Xie R-G, Jiang B-C, Ji R-R, Gao Y-J: Chemokine CXCL1 enhances inflammatory pain and increases NMDA receptor activity and COX-2 expression in spinal cord neurons via activation of CXCR2. Exp Neurol 261:328–336, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Cao S, Yuan J, Zhang D, Wen S, Wang J, Li Y, Deng W: Transcriptome changes in dorsal spinal cord of rats with neuropathic pain. J Pain Res 12:3013–3023, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Chapman CR, Tuckett RP, Song CW: Pain and stress in a systems perspective: reciprocal neural, endocrine, and immune interactions. J Pain 9:122–145, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Choi S, Na HS, Kim J, Lee J, Lee S, Kim D, Park J, Chen CC, Campbell KP, Shin HS: Attenuated pain responses in mice lacking Ca(V)3.2 T-type channels. Genes Brain Behav 6:425–431, 2007 [DOI] [PubMed] [Google Scholar]
  • 16.Daut RL, Cleeland CS, Flanery RC: Development of the Wisconsin brief pain questionnaire to assess pain in cancer and other diseases. Pain 17:197–210, 1983 [DOI] [PubMed] [Google Scholar]
  • 17.David W Hosmer SL Jr., Rodney X Sturdivant: Applied Logistic Regression. Third Edition edition, 2013. [Google Scholar]
  • 18.Draghici S, Khatri P, Tarca AL, Amin K, Done A, Voichita C, Georgescu C, Romero R: A systems biology approach for pathway level analysis. Genome Res 17:1537–1545, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Dunn OJ: Multiple comparisons among means. J Am Stat Assoc 56:52–64, 1961 [Google Scholar]
  • 20.Extermann M, Bonetti M, Sledge GW, O’Dwyer PJ, Bonomi P, Benson AB 3rd: MAX2−a convenient index to estimate the average per patient risk for chemotherapy toxicity; validation in ECOG trials. Eur J Cancer 40:1193–1198, 2004 [DOI] [PubMed] [Google Scholar]
  • 21.Fang D, Kong LY, Cai J, Li S, Liu XD, Han JS, Xing GG: Interleukin-6-mediated functional upregulation of TRPV1 receptors in dorsal root ganglion neurons through the activation of JAK/PI3K signaling pathway: Roles in the development of bone cancer pain in a rat model. Pain 156:1124–1144, 2015 [DOI] [PubMed] [Google Scholar]
  • 22.Fenton BW, Shih E, Zolton J: The neurobiology of pain perception in normal and persistent pain. Pain Manag 5:297–317, 2015 [DOI] [PubMed] [Google Scholar]
  • 23.Finn DP, Haroutounian S, Hohmann AG, Krane E, Soliman N, Rice ASC: Cannabinoids, the endocannabinoid system, and pain: A review of preclinical studies. Pain 162:S5–s25, 2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Fisher RA: Statistical Methods for Research Workers. Edinburgh, Oliver and Boyd, 1925 [Google Scholar]
  • 25.Fisher RA: Questions and answers #14. Am Stat 2:30–31, 1948 [Google Scholar]
  • 26.Gao Y-J, Chemokines Ji R-R: neuronal-glial interactions, and central processing of neuropathic pain. Pharmacol Ther 126:56–68, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Geraghty T, Winter DR, Miller RJ, Miller RE, Malfait AM: Neuroimmune interactions and osteoarthritis pain: Focus on macrophages. Pain Rep 6:e892, 2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Goncalves Dos Santos G, Delay L, Yaksh TL, Corr M: Neuraxial Cytokines in Pain States. Front Immunol 10:3061, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Gray KA, Daugherty LC, Gordon SM, Seal RL, Wright MW, Bruford EA: Genenames.org: the HGNC resources in 2013. Nucleic Acids Res 41:D545–D552, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Hagenston AM, Simonetti M: Neuronal calcium signaling in chronic pain. Cell Tissue Res 357:407–426, 2014 [DOI] [PubMed] [Google Scholar]
  • 31.Harrington R, Al Nokhatha SA, Conway RJAK: Inhibitors in rheumatoid arthritis: An evidence-based review on the emerging clinical data. J Inflamm Res 13:519–531, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Ji R-R, Chamessian A, Zhang Y-Q: Pain regulation by non-neuronal cells and inflammation. Science (New York, N.Y.) 354:572–577, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Ji R-R, Xu Z-Z, Gao Y-J: Emerging targets in neuroinflammation-driven chronic pain. Nat Rev Drug Discov 13:533–548, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Jiang B-C, Liu T, Gao Y-J: Chemokines in chronic pain: cellular and molecular mechanisms and therapeutic potential. Pharmacol Ther 212:107581, 2020 [DOI] [PubMed] [Google Scholar]
  • 35.Jose PJ, Moss IK, Maini RN, Williams TJ: Measurement of the chemotactic complement fragment C5a in rheumatoid synovial fluids by radioimmunoassay: role of C5a in the acute inflammatory phase. Ann Rheum Dis 49:747–752, 1990 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Jung T, Wickrama KAS: An Introduction to Latent Class Growth Analysis and growth mixture modeling. Soc Person Psychol Compass 2:302–317, 2008 [Google Scholar]
  • 37.Karnofsky D: Performance Scale. New York, Plenum Press, 1977 [Google Scholar]
  • 38.Kasatkina LA, Rittchen S, Sturm EM: Neuroprotective and immunomodulatory action of the endocannabinoid system under neuroinflammation. Int J Mol Sci 22, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Kawasaki Y, Zhang L, Cheng J-K, Ji R-R: Cytokine mechanisms of central sensitization: distinct and overlapping role of interleukin-1beta, interleukin-6, and tumor necrosis factor-alpha in regulating synaptic and neuronal activity in the superficial spinal cord. J Neurosci 28:5189–5194, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Kiener HP, Baghestanian M, Dominkus M, Walchshofer S, Ghannadan M, Willheim M, Sillaber C, Graninger WB, Smolen JS, Valent P: Expression of the C5a receptor (CD88) on synovial mast cells in patients with rheumatoid arthritis. Arthritis Rheum 41:233–245, 1998 [DOI] [PubMed] [Google Scholar]
  • 41.Landau WM, Liu P: Dispersion estimation and its effect on test performance in RNA-seq data analysis: a simulation-based comparison of methods. PLoS One 8:e81415, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Laumet G, Bavencoffe A, Edralin JD, Huo X-J, Walters ET, Dantzer R, Heijnen CJ, Kavelaars A: Interleukin-10 resolves pain hypersensitivity induced by cisplatin by reversing sensory neuron hyperexcitability. PAIN 161:2344–2352, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.LeBaron VT, Camacho F, Balkrishnan R, Yao NA, Gilson AM: Opioid epidemic or pain crisis? Using the virginia all payer claims database to describe opioid medication prescribing patterns and potential harms for patients with cancer. J Oncol Pract 15:e997–e1009, 2019 [DOI] [PubMed] [Google Scholar]
  • 44.Leek JT: svaseq: removing batch effects and other unwanted noise from sequencing data. Nucleic Acids Res 42, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Leek JT, Storey JD: Capturing heterogeneity in gene expression studies by surrogate variable analysis. PLoS Genet 3:1724–1735, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Leo M, Schmitt L-I, Erkel M, Melnikova M, Thomale J, Hagenacker T: Cisplatin-induced neuropathic pain is mediated by upregulation of N-type voltage-gated calcium channels in dorsal root ganglion neurons. Exp Neurol 288:62–74, 2017 [DOI] [PubMed] [Google Scholar]
  • 47.Liou J-T, Lee C-M, Day Y-J: The immune aspect in neuropathic pain: Role of chemokines. Acta Anaesthesiologica Taiwanica 51:127–132, 2013 [DOI] [PubMed] [Google Scholar]
  • 48.Long J-Y, Wang X-J, Li X-Y, Kong X-H, Yang G, Zhang D, Yang Y-T, Shi Z, Ma X-P: Spinal microglia and astrocytes: Two key players in chronic visceral pain pathogenesis. Neurochem Res, 2021 [DOI] [PubMed] [Google Scholar]
  • 49.Matsuda M, Huh Y, Ji RR: Roles of inflammation, neurogenic inflammation, and neuroinflammation in pain. J Anesth 33:131–139, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Matsuka Y, Afroz S, Dalanon JC, Iwasa T, Waskitho A, Oshima M: The role of chemical transmitters in neuron-glia interaction and pain in sensory ganglion. Neurosci Biobehav Rev 108:393–399, 2020 [DOI] [PubMed] [Google Scholar]
  • 51.McCarthy DJ, Chen Y, Smyth GK: Differential expression analysis of multifactor RNA-Seq experiments with respect to biological variation. Nucleic Acids Res 40:4288–4297, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Miaskowski C, Topp K, Conley YP, Paul SM, Melisko M, Schumacher M, Chesney M, Abrams G, Levine JD, Kober KM: Perturbations in neuroinflammatory pathways are associated with paclitaxel-induced peripheral neuropathy in breast cancer survivors. J Neuroimmunol 335:577019, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Mitrea C, Taghavi Z, Bokanizad B, Hanoudi S, Tagett R, Donato M, Voichita C, Draghici S: Methods and approaches in the topology-based analysis of biological pathways. Front Physiol 4:278, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Mori T, Miyamoto T, Yoshida H, Asakawa M, Kawasumi M, Kobayashi T, Morioka H, Chiba K, Toyama Y, Yoshimura A: IL-1β and TNFα-initiated IL-6−STAT3 pathway is critical in mediating inflammatory cytokines and RANKL expression in inflammatory arthritis. Int Immunol 23:701–712, 2011 [DOI] [PubMed] [Google Scholar]
  • 55.Morris R, Kershaw NJ, Babon JJ: The molecular details of cytokine signaling via the JAK/STAT pathway. Protein Sci 27:1984–2009, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Moryl N, Dave V, Glare P, Bokhari A, Malhotra VT, Gulati A, Hung J, Puttanniah V, Griffo Y, Tickoo R, Wiesenthal A, Horn SD, Inturrisi CE: Patient-Reported Outcomes and Opioid Use by Outpatient Cancer Patients. J Pain 19:278–290, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Muthen B, Shedden K: Finite mixture modeling with mixture outcomes using the EM algorithm. Biometrics 55:463–469, 1999 [DOI] [PubMed] [Google Scholar]
  • 58.Muthen LK, Muthen BO: Mplus User’s Guide, 8th ed. Los Angeles, CA, Muthen & Muthen, 1998, pp 2020. [Google Scholar]
  • 59.Nguyen THP, Hokstad I, Fagerland MW, Mollnes TE, Hollan I, Feinberg MW, Hjeltnes G, Eilertsen GO, Mikkelsen K, Agewall S: Antirheumatic therapy is associated with reduced complement activation in rheumatoid arthritis. PLoS One 17:e0264628, 2022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Oppegaard K, Harris CS, Shin J, Paul SM, Cooper BA, Chan A, Anguera JA, Levine J, Conley Y, Hammer M, Miaskowski CA, Chan RJ, Kober KM: Cancer-related cognitive impairment is associated with perturbations in inflammatory pathways. Cytokine 148:155653, 2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Pevida M, González-Rodríguez S, Lastra A, García-Suárez O, Hidalgo A, Menéndez L, Baamonde A: Involvement of spinal chemokine CCL2 in the hyperalgesia evoked by bone cancer in mice: A role for astroglia and microglia. Cell Mol Neurobiol 34:143–156, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Pevida M, Lastra A, Meana A, Hidalgo Á, Baamonde A, Menéndez L: The chemokine CCL5 induces CCR1-mediated hyperalgesia in mice inoculated with NCTC 2472 tumoral cells. Neuroscience 259:113–125, 2014 [DOI] [PubMed] [Google Scholar]
  • 63.Pinho-Ribeiro FA, Verri WA Jr, Chiu IM: Nociceptor Sensory Neuron-Immune Interactions in Pain and Inflammation. Trends Immunol 38:5–19, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Posternak V, Dunn LB, Dhruva A, Paul SM, Luce J, Mastick J, Levine JD, Aouizerat BE, Hammer M, Wright F, Miaskowski C: Differences in demographic, clinical, and symptom characteristics and quality of life outcomes among oncology patients with different types of pain. Pain 157:892–900, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Qiao Z, Tang J, Wu W, Tang J, Liu M: Acteoside inhibits inflammatory response via JAK/STAT signaling pathway in osteoarthritic rats. BMC Complement Alternat Med 19:264, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Radwani H, Lopez-Gonzalez MJ, Cattaert D, Roca-Lapirot O, Dobremez E, Bouali-Benazzouz R, Eiríksdóttir E, Langel Ü, Favereaux A, Errami M, Landry M, Fossat P: Cav1.2 and Cav1.3 L-type calcium channels independently control short- and long-term sensitization to pain. J Physiol 594:6607–6626, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Robinson MD, McCarthy DJ, Smyth GK: edgeR: a Bio-conductor package for differential expression analysis of digital gene expression data. Bioinformatics 26:139–140, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Roca-Lapirot O, Radwani H, Aby F, Nagy F, Landry M, Fossat P: Calcium signalling through L-type calcium channels: role in pathophysiology of spinal nociceptive transmission. Br J Pharmacol 175:2362–2374, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Saegusa H, Kurihara T, Zong S, Kazuno A, Matsuda Y, Nonaka T, Han W, Toriyama H, Tanabe T: Suppression of inflammatory and neuropathic pain symptoms in mice lacking the N-type Ca2+ channel. Embo j 20:2349–2356, 2001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Sangha O, Stucki G, Liang MH, Fossel AH, Katz JN: The self-administered comorbidity questionnaire: A new method to assess comorbidity for clinical and health services research. Arthritis & Rheumatism 49:156–163, 2003 [DOI] [PubMed] [Google Scholar]
  • 71.Santoni A, Mercadante S, Arcuri E: Chronic cancer and non-cancer pain and opioid-induced hyperalgesia share common mechanisms: neuroinflammation and central sensitization. Minerva Anestesiol 87:210–222, 2021 [DOI] [PubMed] [Google Scholar]
  • 72.Scheff NN, Ye Y, Bhattacharya A, MacRae J, Hickman DN, Sharma AK, Dolan JC, Schmidt BL: Tumor necrosis factor alpha secreted from oral squamous cell carcinoma contributes to cancer pain and associated inflammation. Pain 158:2396–2409, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Sekiguchi F, Tsubota M, Kawabata A: Involvement of voltage-gated calcium channels in inflammation and inflammatory pain. Biol Pharm Bull 41:1127–1134, 2018 [DOI] [PubMed] [Google Scholar]
  • 74.Shin J, Harris C, Oppegaard K, Kober KM, Paul SM, Cooper BA, Hammer M, Conley Y, Levine JD, Miaskowski C: Worst pain severity profiles of oncology patients are associated with significant stress and multiple co-occurring symptoms. J Pain 23:74–88, 2022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Singh KP, Dhruva A, Flowers E, Paul SM, Hammer MJ, Wright F, Cartwright F, Conley YP, Melisko M, Levine JD, Miaskowski C, Kober KM: Alterations in patterns of gene expression and perturbed pathways in the gut-brain axis are associated with chemotherapy-induced nausea. J Pain Symptom Manage 59:1248–1259, 2020. e1245 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Limma Smyth G:: Linear Models for Microarray Data. Gentleman RC, Carey VJ, Dudoit S, Irizarry R, Huber W, editors. Linear Models for Microarray Data. Bioinformatics and Computational Biology; 397–420, 2005 [Google Scholar]
  • 77.Smyth GK, Ritchie M, Thorne N, Wettenhall J: LIMMA: linear models for microarray data. In Bioinformatics and Computational Biology Solutions Using R and Bioconductor. Statistics for Biology and Health., 2005 [Google Scholar]
  • 78.Team RC: R: A Language and Environment for Statistical Computing. Vienna, Austria, R Foundation for Statistical Computing, 2019 [Google Scholar]
  • 79.Utne I, Loyland B, Grov EK, Rasmussen HL, Torstveit AH, Cooper BA, Mastick J, Mazor M, Wong M, Paul SM, Conley YP, Jahan T, Ritchie C, Levine JD, Miaskowski C: Distinct attentional function profiles in older adults receiving cancer chemotherapy. Eur J Oncol Nurs 36:32–39, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Vicario N, Turnaturi R, Spitale FM, Torrisi F, Zappalà A, Gulino R, Pasquinucci L, Chiechio S, Parenti C, Parenti R: Intercellular communication and ion channels in neuropathic pain chronicization. Inflamm Res 69:841–850, 2020 [DOI] [PubMed] [Google Scholar]
  • 81.Wang Q, Rozelle AL, Lepus CM, Scanzello CR, Song JJ, Larsen DM, Crish JF, Bebek G, Ritter SY, Lindstrom TM, Hwang I, Wong HH, Punzi L, Encarnacion A, Shamloo M, Goodman SB, Wyss-Coray T, Goldring SR, Banda NK, Thurman JM, Gobezie R, Crow MK, Holers VM, Lee DM, Robinson WH: Identification of a central role for complement in osteoarthritis. Nat Med 17:1674–1679, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Warwick CA, Keyes AL, Woodruff TM, Usachev YM: The complement cascade in the regulation of neuroinflammation, nociceptive sensitization, and pain. J Biol Chem 297:101085, 2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Wei XH, Na XD, Liao GJ, Chen QY, Cui Y, Chen FY, Li YY, Zang Y, Liu XG: The up-regulation of IL-6 in DRG and spinal dorsal horn contributes to neuropathic pain following L5 ventral root transection. Exp Neurol 241:159–168, 2013 [DOI] [PubMed] [Google Scholar]
  • 84.White FA, Bhangoo SK, Miller RJ: Chemokines: integrators of pain and inflammation. Nat Rev Drug Discov 4:834–844, 2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.White FA, Feldman P, Miller RJ: Chemokine signaling and the management of neuropathic pain. Mol Interv 9:188–195, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Woller SA, Eddinger KA, Corr M, Yaksh TL: An overview of pathways encoding nociception. Clin Exp Rheumatol 35 Suppl 107:40–46, 2017 [PMC free article] [PubMed] [Google Scholar]
  • 87.Xiao M, Sherman SL, Abrams GD: Inflammatory mechanisms in the development of osteoarthritis. Instr Course Lect 70:537–550, 2021 [PubMed] [Google Scholar]
  • 88.Xie R-G, Gao Y-J, Park C-K, Lu N, Luo C, Wang W-T, Wu S-X, Ji R-R: Spinal CCL2 promotes central sensitization, long-term potentiation, and inflammatory pain via CCR2: Further insights into molecular, synaptic, and cellular mechanisms. Neurosci Bull 34:13–21, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Xu J, Zhu M-D, Zhang X, Tian H, Zhang J-H, Wu X-B, Gao Y-J: NFκB-mediated CXCL1 production in spinal cord astrocytes contributes to the maintenance of bone cancer pain in mice. Journal of Neuroinflammation 11: 38, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Xu M, Cheng Z, Ding Z, Wang Y, Guo Q, Huang C: Resveratrol enhances IL-4 receptor-mediated anti-inflammatory effects in spinal cord and attenuates neuropathic pain following sciatic nerve injury. Mol Pain 14:174480691876754, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Yang J, Xie M-X, Hu L, Wang X-F, Mai J-Z, Li Y-Y, Wu N, Zhang C, Li J, Pang R-P, Liu X-G: Upregulation of N-type calcium channels in the soma of uninjured dorsal root ganglion neurons contributes to neuropathic pain by increasing neuronal excitability following peripheral nerve injury. Brain Behav Immun 71:52–65, 2018 [DOI] [PubMed] [Google Scholar]
  • 92.Yang QQ, Li HN, Zhang ST, Yu YL, Wei W, Zhang X, Wang JY, Zeng XY: Red nucleus IL-6 mediates the maintenance of neuropathic pain by inducing the productions of TNF-α and IL-1β through the JAK2/STAT3 and ERK signaling pathways. Neuropathology 40:347–357, 2020 [DOI] [PubMed] [Google Scholar]
  • 93.Yi M-H, Liu YU, Liu K, Chen T, Bosco DB, Zheng J, Xie M, Zhou L, Qu W, Wu L-J: Chemogenetic manipulation of microglia inhibits neuroinflammation and neuropathic pain in mice. Brain Behav Immun 92:78–89, 2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Zhou L-J, Peng J, Xu Y-N, Zeng W-J, Zhang J, Wei X, Mai C-L, Lin Z-J, Liu Y, Murugan M, Eyo UB, Umpierre AD, Xin W-J, Chen T, Li M, Wang H, Richardson JR, Tan Z, Liu X-G, Wu L-J: Microglia are indispensable for synaptic plasticity in the spinal dorsal horn and chronic pain. Cell Reports 27:3844–3859, 2019.e3846 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Zhu X, Cao S, Zhu MD, Liu JQ, Chen JJ, Gao YJ: Contribution of chemokine CCL2/CCR2 signaling in the dorsal root ganglion and spinal cord to the maintenance of neuropathic pain in a rat model of lumbar disc herniation. J Pain 15:516–526, 2014 [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supp Data 4
Supp Data 2
Supp Data 3
Supp Data 1

RESOURCES