Inflammatory pathway genes associated with inter-individual variability in the trajectories of morning and evening fatigue in patients receiving chemotherapy.
Journal: 2017/February - Cytokine
ISSN: 1096-0023
Abstract:
Fatigue, a highly prevalent and distressing symptom during chemotherapy (CTX), demonstrates diurnal and interindividual variability in severity. Little is known about the associations between variations in genes involved in inflammatory processes and morning and evening fatigue severity during CTX. The purposes of this study, in a sample of oncology patients (N=543) with breast, gastrointestinal (GI), gynecological (GYN), or lung cancer who received two cycles of CTX, were to determine whether variations in genes involved in inflammatory processes were associated with inter-individual variability in initial levels as well as in the trajectories of morning and evening fatigue. Patients completed the Lee Fatigue Scale to determine morning and evening fatigue severity a total of six times over two cycles of CTX. Using a whole exome array, 309 single nucleotide polymorphisms SNPs among the 64 candidate genes that passed all quality control filters were evaluated using hierarchical linear modeling (HLM). Based on the results of the HLM analyses, the final SNPs were evaluated for their potential impact on protein function using two bioinformational tools. The following inflammatory pathways were represented: chemokines (3 genes); cytokines (12 genes); inflammasome (11 genes); Janus kinase/signal transducers and activators of transcription (JAK/STAT, 10 genes); mitogen-activated protein kinase/jun amino-terminal kinases (MAPK/JNK, 3 genes); nuclear factor-kappa beta (NFkB, 18 genes); and NFkB and MAP/JNK (7 genes). After controlling for self-reported and genomic estimates of race and ethnicity, polymorphisms in six genes from the cytokine (2 genes); inflammasome (2 genes); and NFkB (2 genes) pathways were associated with both morning and evening fatigue. Polymorphisms in six genes from the inflammasome (1 gene); JAK/STAT (1 gene); and NFkB (4 genes) pathways were associated with only morning fatigue. Polymorphisms in three genes from the inflammasome (2 genes) and the NFkB (1 gene) pathways were associated with only evening fatigue. Taken together, these findings add to the growing body of evidence that suggests that morning and evening fatigue are distinct symptoms.
Relations:
Content
Citations
(5)
References
(135)
Affiliates
(7)
Similar articles
Articles by the same authors
Discussion board
Cytokine 91: 187-210

Inflammatory Pathway Genes Associated with Inter-Individual Variability in the Trajectories of Morning and Evening Fatigue in Patients Receiving Chemotherapy

+2 authors

1. Introduction

During chemotherapy (CTX), over 45% of patients experience clinically meaningful levels of fatigue that decrease their ability to tolerate treatments, engage in social relationships, and maintain regular work activities [1]. However, a growing body of evidence demonstrates that inter-individual variability exists in fatigue severity across cancer diagnosis [24] and treatments [5, 6]. In addition, recent work from our group [711] and others [4, 12] demonstrates that morning and evening fatigue are distinct yet related symptoms. Some of this inter-individual variability is explained by different phenotypic characteristics that distinguish between higher levels of morning (e.g., higher body mass index (BMI), lack of regular exercise, higher state anxiety) and evening (e.g., being white, higher years of education, child care responsibilities) fatigue [9, 10]. In addition to phenotypic differences, preliminary evidence suggests that variations in cytokine genes are associated with inter-individual differences in morning (e.g., tumor necrosis factor alpha (TNFA) [13] and evening (e.g., interleukin (IL)4 [14] and IL6 [15]) fatigue severity.

While considered to be multi-factorial, a growing body of evidence suggests that cytokine dysregulation, as well as many other neuroinflammatory processes may modulate fatigue severity in a number of chronic conditions [1619]. Increased knowledge of the mechanisms that underlie fatigue is essential for the development of effective treatments for this devastating symptom. However, no definitive conclusions can be drawn from studies that evaluated associations between fatigue severity and various biomarkers of cytokine dysregulation (for reviews see [18, 19]).

1.1 Associations between fatigue and serum markers of inflammation

To examine this cytokine dysregulation hypothesis, several studies evaluated the associations between fatigue severity and serum cytokine levels. To date, results are inconclusive, with some studies finding positive associations between fatigue severity and circulating levels of TNF-α [20, 21] and IL-6 [18, 20, 2229] and others finding no associations with TNF-α[18, 22, 3033], IL-6 [30, 3438], and IL-4 [25, 36, 38]. These inconsistent results may be related to the challenges associated with the measurement of serum cytokines, as well as circadian variations in cytokine levels [39].

An alternative approach is to measure circulating levels of biomarkers of immune activation (e.g., cellular receptors) [40]. Again, these results are inconclusive. Some studies found positive associations between fatigue and changes in serum levels of IL-1 receptor antagonist (IL-1ra)[24, 35, 41, 42], soluble TNF receptor II (sTNF-RII) [41, 43, 44], sTNF-RI [45], and sIL-6R [37, 46, 47]. However, other studies found no associations between fatigue severity and changes in serum levels of IL-1ra [27, 34, 36], sTNF-RII [36, 42], and sIL-6R [46]. Of note, none of these studies evaluated for associations between diurnal variations in fatigue severity and changes in these serum markers.

1.2 Changes in gene expression and fatigue

Another approach to examine the role of inflammation in fatigue is to evaluate for changes in the expression of inflammatory genes. To date, seven studies have evaluated for changes in gene expression associated with fatigue severity in oncology patients [34, 4853]. Five of these studies [34, 48, 50, 52, 53] examined changes in gene expression related to inflammation/immune function. In four of these studies higher levels of fatigue were associated with upregulation of genes that regulate cytokine production (i.e., interferon alpha-inducible protein 27 (IFI27) [48], α-synuclein (SNCA) [52], IL1 [34], IL6 [34], IL4 [50]). In another study [53], differentially perturbed cytokine pathways were associated with higher levels of evening fatigue. However, across these studies only mean or evening fatigue scores were evaluated. In addition, the sample sizes for these studies were relatively small (i.e., 15 [49] to 137 [50] patients).

1.3 Associations between fatigue and variations in cytokine genes

A third approach that can be used to examine the role of inflammation in fatigue is to evaluate for associations between fatigue severity and variations in cytokine genes. Single nucleotide polymorphisms (SNP) in TNFA [54, 55], IL6 [37, 56, 57], and IL1RA [58] were associated with increased levels of fatigue. To date, only three studies evaluated for associations between variations in cytokine genes and diurnal variations in fatigue severity [1315]. In a study of oncology patients (n=185) and their family caregivers (n=103), SNPs in TNFA (i.e., rs1800629, rs3093662) and IL6 (i.e., rs4719714) were associated with higher levels of morning and evening fatigue [1315]. Additionally, a polymorphism in IL4 rs2243248 was associated with lower levels of evening fatigue [14]. While the studies cited above provide preliminary evidence that variations in cytokine genes are associated with diurnal variations in fatigue severity, two of the studies evaluated only one polymorphism [1315] and none of them evaluated oncology patients undergoing CTX.

While evidence exists for the role of cytokine dysregulation as a modulator of neuroinflammation, recent studies found other pathways and processes that contribute to the development of inflammation (e.g., the mitogen-activated protein kinase (MAPK) pathway [59], and inflammasomes [19, 60]). However, the contribution of these pathways to fatigue severity in oncology patients undergoing CTX has not been evaluated. Increased knowledge of whether additional inflammatory pathways are associated with diurnal variations in fatigue severity would enhance our understanding of the various mechanisms that contribute to this devastating symptom.

Recently, we identified common and distinct phenotypic characteristics for morning [9] and evening [10] fatigue severity in oncology patients undergoing CTX. This study extends these findings to identify associations between variations in genes associated with a variety of inflammatory processes and the severity of morning and evening fatigue. Since genes interact with one another [61], the polymorphisms that were evaluated were grouped into common inflammatory pathways to provide insights into the role of related genes and the severity of morning and evening fatigue. The purposes of this study, in a sample of oncology patients with breast, gastrointestinal (GI), gynecological (GYN), or lung cancer who received two cycles of CTX, were to determine whether variations in genes involved in inflammatory processes were associated with inter-individual variability in initial levels as well as in the trajectories of morning and evening fatigue.

1.1 Associations between fatigue and serum markers of inflammation

To examine this cytokine dysregulation hypothesis, several studies evaluated the associations between fatigue severity and serum cytokine levels. To date, results are inconclusive, with some studies finding positive associations between fatigue severity and circulating levels of TNF-α [20, 21] and IL-6 [18, 20, 2229] and others finding no associations with TNF-α[18, 22, 3033], IL-6 [30, 3438], and IL-4 [25, 36, 38]. These inconsistent results may be related to the challenges associated with the measurement of serum cytokines, as well as circadian variations in cytokine levels [39].

An alternative approach is to measure circulating levels of biomarkers of immune activation (e.g., cellular receptors) [40]. Again, these results are inconclusive. Some studies found positive associations between fatigue and changes in serum levels of IL-1 receptor antagonist (IL-1ra)[24, 35, 41, 42], soluble TNF receptor II (sTNF-RII) [41, 43, 44], sTNF-RI [45], and sIL-6R [37, 46, 47]. However, other studies found no associations between fatigue severity and changes in serum levels of IL-1ra [27, 34, 36], sTNF-RII [36, 42], and sIL-6R [46]. Of note, none of these studies evaluated for associations between diurnal variations in fatigue severity and changes in these serum markers.

1.2 Changes in gene expression and fatigue

Another approach to examine the role of inflammation in fatigue is to evaluate for changes in the expression of inflammatory genes. To date, seven studies have evaluated for changes in gene expression associated with fatigue severity in oncology patients [34, 4853]. Five of these studies [34, 48, 50, 52, 53] examined changes in gene expression related to inflammation/immune function. In four of these studies higher levels of fatigue were associated with upregulation of genes that regulate cytokine production (i.e., interferon alpha-inducible protein 27 (IFI27) [48], α-synuclein (SNCA) [52], IL1 [34], IL6 [34], IL4 [50]). In another study [53], differentially perturbed cytokine pathways were associated with higher levels of evening fatigue. However, across these studies only mean or evening fatigue scores were evaluated. In addition, the sample sizes for these studies were relatively small (i.e., 15 [49] to 137 [50] patients).

1.3 Associations between fatigue and variations in cytokine genes

A third approach that can be used to examine the role of inflammation in fatigue is to evaluate for associations between fatigue severity and variations in cytokine genes. Single nucleotide polymorphisms (SNP) in TNFA [54, 55], IL6 [37, 56, 57], and IL1RA [58] were associated with increased levels of fatigue. To date, only three studies evaluated for associations between variations in cytokine genes and diurnal variations in fatigue severity [1315]. In a study of oncology patients (n=185) and their family caregivers (n=103), SNPs in TNFA (i.e., rs1800629, rs3093662) and IL6 (i.e., rs4719714) were associated with higher levels of morning and evening fatigue [1315]. Additionally, a polymorphism in IL4 rs2243248 was associated with lower levels of evening fatigue [14]. While the studies cited above provide preliminary evidence that variations in cytokine genes are associated with diurnal variations in fatigue severity, two of the studies evaluated only one polymorphism [1315] and none of them evaluated oncology patients undergoing CTX.

While evidence exists for the role of cytokine dysregulation as a modulator of neuroinflammation, recent studies found other pathways and processes that contribute to the development of inflammation (e.g., the mitogen-activated protein kinase (MAPK) pathway [59], and inflammasomes [19, 60]). However, the contribution of these pathways to fatigue severity in oncology patients undergoing CTX has not been evaluated. Increased knowledge of whether additional inflammatory pathways are associated with diurnal variations in fatigue severity would enhance our understanding of the various mechanisms that contribute to this devastating symptom.

Recently, we identified common and distinct phenotypic characteristics for morning [9] and evening [10] fatigue severity in oncology patients undergoing CTX. This study extends these findings to identify associations between variations in genes associated with a variety of inflammatory processes and the severity of morning and evening fatigue. Since genes interact with one another [61], the polymorphisms that were evaluated were grouped into common inflammatory pathways to provide insights into the role of related genes and the severity of morning and evening fatigue. The purposes of this study, in a sample of oncology patients with breast, gastrointestinal (GI), gynecological (GYN), or lung cancer who received two cycles of CTX, were to determine whether variations in genes involved in inflammatory processes were associated with inter-individual variability in initial levels as well as in the trajectories of morning and evening fatigue.

2. Methods

2.1 Patients and settings

Some of the details of the phenotypic [911, 62] and genotypic [63, 64] methods used in this study are published elsewhere. In brief, patients were recruited from two comprehensive cancer centers, one Veteran’s Affairs hospital, and four community-based oncology programs. Patients with a diagnosis of breast, GI, GYN, or lung cancer were eligible to participate if they were ≥18 years of age; had received CTX within the previous four weeks; were scheduled to receive at least two additional cycles of CTX; were able to read, write, and understand English; and gave written informed consent.

2.2 Instruments

Patients completed a demographic questionnaire, the Karnofsky Performance Status (KPS) scale [65], and the Self-Administered Comorbidity Questionnaire (SCQ) [66]. In addition, patients completed a number of questionnaires to evaluate anxiety [67], depression [68], and sleep disturbance [69].

Fatigue was evaluated using the 18 item Lee Fatigue Scale (LFS) that assesses physical fatigue and energy [70]. Each item was rated on a 0 to 10 numeric rating scale (NRS). Total fatigue and energy scores were calculated as the mean of the 13 fatigue and the 5 energy items, with higher scores indicating greater fatigue severity and higher levels of energy, respectively. Using separate LFS questionnaires, patients were asked to rate each item based on how they felt within 30 minutes of awakening (i.e., morning fatigue and morning energy) and prior to going to bed (i.e., evening fatigue and evening energy). The LFS has established cutoff scores for clinically meaningful levels of fatigue (i.e., ≥3.2 for morning fatigue, ≥5.6 for evening fatigue) [70]. The LFS is easy to administer, relatively short, and has well established validity and reliability [70]. As noted in previous reports (9,10), the Cronbach’s alphas were .95 for evening fatigue, .95 for morning fatigue, .93 for evening energy, and .95 for morning energy.

2.3 Study Procedures

Each of the sites’ Institutional Review Board approved the study. All patients provided written informed consent. Patients completed study questionnaires in their homes a total of six times over two cycles of CTX (prior to CTX administration (i.e., recovery from previous CTX cycle, assessments 1 and 4), approximately 1 week after CTX administration (i.e., acute symptoms, assessments 2 and 5), approximately 2 weeks after CTX administration (i.e., potential nadir, assessments 3 and 6)).

2.1 Patients and settings

Some of the details of the phenotypic [911, 62] and genotypic [63, 64] methods used in this study are published elsewhere. In brief, patients were recruited from two comprehensive cancer centers, one Veteran’s Affairs hospital, and four community-based oncology programs. Patients with a diagnosis of breast, GI, GYN, or lung cancer were eligible to participate if they were ≥18 years of age; had received CTX within the previous four weeks; were scheduled to receive at least two additional cycles of CTX; were able to read, write, and understand English; and gave written informed consent.

2.2 Instruments

Patients completed a demographic questionnaire, the Karnofsky Performance Status (KPS) scale [65], and the Self-Administered Comorbidity Questionnaire (SCQ) [66]. In addition, patients completed a number of questionnaires to evaluate anxiety [67], depression [68], and sleep disturbance [69].

Fatigue was evaluated using the 18 item Lee Fatigue Scale (LFS) that assesses physical fatigue and energy [70]. Each item was rated on a 0 to 10 numeric rating scale (NRS). Total fatigue and energy scores were calculated as the mean of the 13 fatigue and the 5 energy items, with higher scores indicating greater fatigue severity and higher levels of energy, respectively. Using separate LFS questionnaires, patients were asked to rate each item based on how they felt within 30 minutes of awakening (i.e., morning fatigue and morning energy) and prior to going to bed (i.e., evening fatigue and evening energy). The LFS has established cutoff scores for clinically meaningful levels of fatigue (i.e., ≥3.2 for morning fatigue, ≥5.6 for evening fatigue) [70]. The LFS is easy to administer, relatively short, and has well established validity and reliability [70]. As noted in previous reports (9,10), the Cronbach’s alphas were .95 for evening fatigue, .95 for morning fatigue, .93 for evening energy, and .95 for morning energy.

2.3 Study Procedures

Each of the sites’ Institutional Review Board approved the study. All patients provided written informed consent. Patients completed study questionnaires in their homes a total of six times over two cycles of CTX (prior to CTX administration (i.e., recovery from previous CTX cycle, assessments 1 and 4), approximately 1 week after CTX administration (i.e., acute symptoms, assessments 2 and 5), approximately 2 weeks after CTX administration (i.e., potential nadir, assessments 3 and 6)).

3. Genomic Analyses

3.1 Blood collection and genotyping

Genomic deoxyribonucleic acid (DNA) was isolated from peripheral blood mononuclear cells (PBMCs), using the PUREGene DNA Isolation System (Invitrogen, Carlsbad, CA). DNA was quantitated with a Nanodrop Spectrophotometer (ND-1000) and normalized to a concentration of 50 nanograms/microliter (diluted in 10 mM Tris/1 mM EDTA). Genotyping was performed using the HumanExome Array-12 v1.1 on the Infinium Beadchip genotyping platform which provides focused coverage in the coding regions (i.e., exons) of genes (Illumina, San Diego, CA). Data were processed according to the standard protocol using GenomeStudio (Illumina, San Diego, CA).

3.2 Candidate gene and SNP selection

Candidate genes were selected based on evidence in the literature of an association between each gene and fatigue. SNPs representing these genes were selected from the genome-wide SNP array.

Quality control filtering excluded SNPs with call rates of <95%. SNPs with less than (i.e., monomorphic) or more than (i.e., tri- or tetra-allelic) two alleles were excluded. Allele counts at different loci were assumed to be independent. The 309 SNPs among the 64 candidate genes that passed all quality control filters and whose occurrence rates were evaluated in this sample are listed in Supplemental Table 1. The genes are grouped within their common inflammatory pathways based on a review of the literature as well as the description of each gene’s function and pathway found in the National Center for Biotechnology Information (NCBI) gene database and pathcards (http://pathcards.genecards.org). The following inflammatory pathways are represented: chemokines (3 genes); cytokines (12 genes); inflammasome (11 genes); Janus kinase/signal transducers and activators of transcription (JAK/STAT, 10 genes); mitogen-activated protein kinase/jun amino-terminal kinases (MAPK/JNK, 3 genes); nuclear factor-kappa beta (NFkB, 18 genes); and NFkB and MAP/JNK (7 genes).

3.1 Blood collection and genotyping

Genomic deoxyribonucleic acid (DNA) was isolated from peripheral blood mononuclear cells (PBMCs), using the PUREGene DNA Isolation System (Invitrogen, Carlsbad, CA). DNA was quantitated with a Nanodrop Spectrophotometer (ND-1000) and normalized to a concentration of 50 nanograms/microliter (diluted in 10 mM Tris/1 mM EDTA). Genotyping was performed using the HumanExome Array-12 v1.1 on the Infinium Beadchip genotyping platform which provides focused coverage in the coding regions (i.e., exons) of genes (Illumina, San Diego, CA). Data were processed according to the standard protocol using GenomeStudio (Illumina, San Diego, CA).

3.2 Candidate gene and SNP selection

Candidate genes were selected based on evidence in the literature of an association between each gene and fatigue. SNPs representing these genes were selected from the genome-wide SNP array.

Quality control filtering excluded SNPs with call rates of <95%. SNPs with less than (i.e., monomorphic) or more than (i.e., tri- or tetra-allelic) two alleles were excluded. Allele counts at different loci were assumed to be independent. The 309 SNPs among the 64 candidate genes that passed all quality control filters and whose occurrence rates were evaluated in this sample are listed in Supplemental Table 1. The genes are grouped within their common inflammatory pathways based on a review of the literature as well as the description of each gene’s function and pathway found in the National Center for Biotechnology Information (NCBI) gene database and pathcards (http://pathcards.genecards.org). The following inflammatory pathways are represented: chemokines (3 genes); cytokines (12 genes); inflammasome (11 genes); Janus kinase/signal transducers and activators of transcription (JAK/STAT, 10 genes); mitogen-activated protein kinase/jun amino-terminal kinases (MAPK/JNK, 3 genes); nuclear factor-kappa beta (NFkB, 18 genes); and NFkB and MAP/JNK (7 genes).

4. Statistical analyses

4.1 Demographic and clinical data

The sample’s demographic and clinical characteristics and symptom severity scores at enrollment were determined with descriptive statistics and frequency distributions. These analyses were done using the Statistical Package for the Social Sciences (SPSS) version 22 [71].

4.2 Genetic data

Gene counting determined allele and genotype frequencies. To be included in subsequent evaluations, each SNP needed to have a total of six occurrences of the rare allele (i.e., heterozygous or homozygous) in order not to over- or under- estimate the effect of the rare allele. After applying this criterion, 93 SNPs among 49 genes were evaluated as potential predictors of inter-individual variability in morning and evening fatigue. Liability scores composed of the number of rare allele occurrences across all SNPs for each candidate gene were generated by summing the number of rare alleles carried by each patient.

To minimize confounding due to population stratification, ancestry informative markers (AIMs) identified with principal component (PC) analysis were used in subsequent analyses [7275]. Approximately 3,468 AIMS were included in this analysis. To adjust for potential confounding due to population substructure (i.e., race/ethnicity) the first three PCs were included as covariates in the hierarchical linear modeling (HLM) analyses.

4.3 HLM Analysis

Details of the HLM analysis are published elsewhere [9, 10]. In brief, HLM based on full maximum likelihood estimation was performed in two stages to evaluate the effects of individual SNPs and liability scores on initial levels as well as on changes over time in the severity of morning and evening fatigue [76]. Morning and evening fatigue were evaluated in separate HLM analyses. Since the six assessments encompassed two cycles of CTX, a piecewise model strategy was employed to evaluate the pattern of change in morning and evening fatigue over time. The first piece (PW1) modeled change over time during the first CTX cycle (i.e., Assessments 1, 2, and 3). The second piece (PW2) modeled change during the second CTX cycle (i.e. Assessments 4, 5, and 6).

Then, inter-individual differences in the piecewise trajectories of morning and evening fatigue were examined by modeling the individual change parameters (i.e., intercept and slope parameters) as a function of proposed predictors at level 2. First, each of the SNPs and liability scores that passed the quality control filters was evaluated in an exploratory analysis to determine whether it would result in a better fitting model if it alone was added as a predictor. To improve estimation efficiency and construct a parsimonious model, SNP predictors with a t value of <2.0 were excluded from subsequent model testing.

Each of the SNPs and liability scores, identified in the exploratory analyses (Tables 1 and and2),2), were entered into the model that controlled for self-reported and genomic estimates of race and ethnicity to predict each individual change parameter in morning or evening fatigue severity. Only SNPs that maintained a statistically significant contribution were retained in the final models. A p-value of <.05 indicated statistical significance.

Table 1

Candidate Genotype Predictors of Intercept, Piecewise 1 and Piecewise 2 Linear and Quadratic Components for Morning Fatigue

Gene
Symbol
SNP IDPositionChrAllelesModelGenotype
Counts1
IPW1-LPW1-QPW2-LPW2-Q
Cytokine genes
IL10rs30244932069439681G>TR411, 117, 15
IL12Brs32130941587507695T>CR292, 202, 49
TNFArs1041981315407846C>AD233, 241, 69
Inflammasome pathways genes
NLRC5rs284388575706035316T>CD380, 132, 31
NLRC5rs71853205710137316A>GR484, 54, 5
NLRP5rs4719795653897619G>CR417, 119, 7
NLRP6rs7404441128447711T>CD521, 20, 2
NOD2rs20767565075688116A>GR347, 167, 29
JAK/STAT pathway genes
IL4RLiability Score0: 356
1: 124
2: 42
3: 18
4:2
5:1
IFNGR2rs49869583478729421C>GD528, 14, 1
IL10RBrs28341673464078821A>GR277, 212, 54
MAP/JNK pathway genes
PSMD2rs115451691840205423T>GR408, 123, 12
NFkB pathway genes
LTBRLiability Score0: 536
1: 7
IL17RBLiability Score0: 391
1: 129
2: 23
TNFRSF1Ars4149584644264312T>CD525, 17, 1
TNFRSF1Ars4149637644300112A>GD537, 6, 0
TNFRSF10Ars17620230602568T>CA143, 238, 162
TNFRSF10DLiability Score0: 134
1: 243
2: 138
3: 23
4: 5
TNFRSF11ALiability Score0: 99
1: 196
2: 185
3: 46
4: 17
TNFRSF14rs2234163249130622A>GD533, 10, 0
TNFRSF21Liability Score0: 435
1: 94
2: 14
MAP/JNK and NFkB pathway genes
TRAF1rs37618471236902399A>GD176, 265, 102
TRAF4rs359327782707533417A>GD535, 8, 0

█ = From morning fatigue exploratory analysis had a t-value of ≥2.0

= The order of the genotypes for which counts are provided is homozygous common, heterozygote, and homozygous rare.

Liability Score= The total number of rare allele occurrences across all SNPs and models tested for a given gene in the sample.

For the liability scores - The total number of rare allele occurrences across all SNPs for a given gene are coded as follows for IL4R as an example 0: 356 means that 356 patients did not have any doses of the rare alleles across IL4R; 1: 124 means that 124 patients had 1 dose of the rare alleles across IL4R; 2: 42 means that 42 patients had 2 doses of the rare alleles across IL4R; 3: 18 means that 18 patients had 3 doses of the rare alleles across IL4R; and 4: 2 means that 2 patients had 4 doses of the rare alleles across IL4R.

Abbreviations: A = additive model; Chr = chromosome; D = dominant model; I = intercept; IFNGR2 = Interferon gamma receptor 2; IL4R = Interleukin 4 receptor; IL10 = Interleukin 10; IL10RB = Interleukin 10 receptor subunit beta; IL12B= Interleukin 12B; IL17RB = Interleukin 17 receptor B; JAK/STAT = Janus kinase/signal transducers and activators of transcription; JNK = jun amino-terminal kinases; LTBR = Lymphotoxin beta receptor; MAP = mitogen-activated protein kinase; NFkB = nuclear factor-kappa beta; NLRC5 = Nod-Like receptor family, caspase recruitment domain containing 5; NLRP5 = Nod-Like receptor family, pyrin domain containing 5; NLRP6 = Nod-Like receptor family, pyrin domain containing 6; NOD2 = Nucleotide-Binding oligomerization domain containing 2; PSMD2 = Proteasome 26S subunit non-ATPase 2; PW1-L = piecewise 1 linear component; PW1-Q = piecewise 1 quadratic component; PW2-L = piecewise 2 linear component; PW2-Q = piecewise 2 quadratic component; R = recessive model; rs = reference SNP cluster identification number; SNP= single nucleotide polymorphism; TNFA = Tumor necrosis factor alpha; TNFRSF1A = Tumor necrosis factor receptor superfamily member 1A; TNFRSF10A = Tumor necrosis factor receptor superfamily member 10A; TNFRSF10D = Tumor necrosis factor receptor superfamily member 10D; TNFRSF11A = Tumor necrosis factor receptor superfamily member 11A; TNFRSF14 = Tumor necrosis factor receptor superfamily member 14; TNFRSF21 = Tumor necrosis factor receptor superfamily member 21; TRAF1 = Tumor necrosis factor receptor associated factor 1; TRAF4 = Tumor necrosis factor receptor associated factor 4

Table 2

Candidate Genotype Predictors of Intercept, Piecewise 1 and Piecewise 2 Linear and Quadratic Components for Evening Fatigue

Gene
Symbol
SNP IDPositionChrAllelesModelGenotype
Counts1
IPW1-LPW1-QPW2-LPW2-Q
Cytokine genes
IL12Brs32130941587507695T>CR292, 202, 49
IL12BLiability Score0: 275
1: 211
2: 49
3: 8
IL17Frs11465553521017586T>CD513, 30, 0
IL17Liability Score0: 511
1: 32
TNFArs1041981315407846C>AD233, 241, 69
Inflammasome pathways genes
CARD6rs10512747408417415T>CA441, 92, 10
CARD6Liability Score0: 401
1: 127
2: 15
NLRC5rs169651505705948416T>CD514, 27, 2
NLRP4rs178573735636990819C>GD500, 42, 1
NLRP5rs4719795653897619G>CR417, 119, 7
NLRP6rs7404441128447711T>CD521, 20, 2
NOD2rs20767565075688116A>GR347, 167, 29
JAK/STAT pathway genes
IFNGR2rs49869583478729421C>GD528, 14, 1
IL2RArs791587608869910A>GR149, 268, 126
IL2RBrs22840333753403422A>GD180, 261, 102
MAP/JNK pathway genes
IL17RDrs61742267571320353A>GD528, 18, 0
NFkB pathway genes
IL17RBrs2232346538928303T>CD520, 23, 0
IL17RBrs1043261538992763T>CR437, 96, 10
LTBRLiability Score0: 536
1: 7
TNFRSF14rs2234163249130622A>GD533, 10, 0

█ = From evening fatigue exploratory analysis had a t-value of ≥2.0

= The order of the genotypes for which counts are provided is homozygous common, heterozygote, and homozygous rare.

Liability Score= The total number of rare allele occurrences across all SNPs and models tested for a given gene in the sample.

For the liability scores - The total number of rare allele occurrences across all SNPs for a given gene are coded as follows for IL12B as an example 0: 275 means that 275 patients did not have any doses of the rare alleles across IL12B; 1: 211 means that 211 patients had 1 dose of the rare alleles across IL12B; 2: 49 means that 49 patients had 2 doses of the rare alleles across IL12B; 3: 8 means that 8 patients had 3 doses of the rare alleles across IL12B.

Abbreviations: A = additive model; CARD6 = Caspase recruitment domain family member 6; Chr = chromosome; D = dominant model; I = intercept; IFNGR2 = Interferon gamma receptor 2; IL2RA = Interleukin 2 receptor alpha; IL2RB = Interleukin 2 receptor beta; IL12B= Interleukin 12B; IL17 = Interleukin 17; IL17F = Interleukin 17F; IL17RB = Interleukin 17 receptor B; IL17RD = Interleukin 17 receptor D; JAK/STAT = janus kinase/signal transducers and activators of transcription; JNK = jun amino-terminal kinases; LTBR = Lymphotoxin beta receptor; MAP = mitogen-activated protein kinase; NFkB = nuclear factor-kappa beta; NLRC5 = Nod-Like receptor family, caspase recruitment domain containing 5; NLRP4 = Nod-Like receptor family, pyrin domain containing 4; NLRP5 = Nod-Like receptor family, pyrin domain containing 5; NLRP6 = Nod-Like receptor family, pyrin domain containing 6; NOD2 = Nucleotide-Binding oligomerization domain containing 2; PW1-L = piecewise 1 linear component; PW1-Q = piecewise 1 quadratic component; PW2-L = piecewise 2 linear component; PW2-Q = piecewise 2 quadratic component; R = recessive model; rs = reference SNP cluster identification number; SNP= single nucleotide polymorphism; TNFA = Tumor necrosis factor alpha; TNFRSF14 = Tumor necrosis factor receptor superfamily member 14.

Consistent with our previous studies [14, 63, 7780], recommendations from the literature [81, 82], rigorous quality controls for genomic data and the exploratory nature of our analyses, adjustments were not made for multiple testing. Since significant SNPs and liability scores identified in the exploratory analysis were evaluated further in the HLM analyses that controlled for population stratification (i.e., genomic and self-reported estimates of race and ethnicity), and other variations in the same gene, the significant independent genetic associations reported are unlikely to be due solely to chance.

4.4 Estimation of polymorphism function

Based on the results of the HLM analyses, the SNPs associated with inter-individual differences in the initial levels or trajectories of morning or evening fatigue were evaluated for their potential impact on protein function using two bioinformational tools (i.e., Sorting Intolerant From Tolerant (SIFT) algorithm [83] and Polymorphism Phenotyping v2 (PolyPhen-2) [84]). SIFT predicts whether a SNP in a coding region results in an amino acid substitution that may affect protein function. This prediction is based on an analysis of the conservation of amino acid residues in sequence alignments of closely related sequences [84]. PolyPhen-2 compares the SNP to sequence-based and structure-based predictive features to predict the functional significance of the SNP [84]. Results from the bioinformational tools are described in the discussion.

4.1 Demographic and clinical data

The sample’s demographic and clinical characteristics and symptom severity scores at enrollment were determined with descriptive statistics and frequency distributions. These analyses were done using the Statistical Package for the Social Sciences (SPSS) version 22 [71].

4.2 Genetic data

Gene counting determined allele and genotype frequencies. To be included in subsequent evaluations, each SNP needed to have a total of six occurrences of the rare allele (i.e., heterozygous or homozygous) in order not to over- or under- estimate the effect of the rare allele. After applying this criterion, 93 SNPs among 49 genes were evaluated as potential predictors of inter-individual variability in morning and evening fatigue. Liability scores composed of the number of rare allele occurrences across all SNPs for each candidate gene were generated by summing the number of rare alleles carried by each patient.

To minimize confounding due to population stratification, ancestry informative markers (AIMs) identified with principal component (PC) analysis were used in subsequent analyses [7275]. Approximately 3,468 AIMS were included in this analysis. To adjust for potential confounding due to population substructure (i.e., race/ethnicity) the first three PCs were included as covariates in the hierarchical linear modeling (HLM) analyses.

4.3 HLM Analysis

Details of the HLM analysis are published elsewhere [9, 10]. In brief, HLM based on full maximum likelihood estimation was performed in two stages to evaluate the effects of individual SNPs and liability scores on initial levels as well as on changes over time in the severity of morning and evening fatigue [76]. Morning and evening fatigue were evaluated in separate HLM analyses. Since the six assessments encompassed two cycles of CTX, a piecewise model strategy was employed to evaluate the pattern of change in morning and evening fatigue over time. The first piece (PW1) modeled change over time during the first CTX cycle (i.e., Assessments 1, 2, and 3). The second piece (PW2) modeled change during the second CTX cycle (i.e. Assessments 4, 5, and 6).

Then, inter-individual differences in the piecewise trajectories of morning and evening fatigue were examined by modeling the individual change parameters (i.e., intercept and slope parameters) as a function of proposed predictors at level 2. First, each of the SNPs and liability scores that passed the quality control filters was evaluated in an exploratory analysis to determine whether it would result in a better fitting model if it alone was added as a predictor. To improve estimation efficiency and construct a parsimonious model, SNP predictors with a t value of <2.0 were excluded from subsequent model testing.

Each of the SNPs and liability scores, identified in the exploratory analyses (Tables 1 and and2),2), were entered into the model that controlled for self-reported and genomic estimates of race and ethnicity to predict each individual change parameter in morning or evening fatigue severity. Only SNPs that maintained a statistically significant contribution were retained in the final models. A p-value of <.05 indicated statistical significance.

Table 1

Candidate Genotype Predictors of Intercept, Piecewise 1 and Piecewise 2 Linear and Quadratic Components for Morning Fatigue

Gene
Symbol
SNP IDPositionChrAllelesModelGenotype
Counts1
IPW1-LPW1-QPW2-LPW2-Q
Cytokine genes
IL10rs30244932069439681G>TR411, 117, 15
IL12Brs32130941587507695T>CR292, 202, 49
TNFArs1041981315407846C>AD233, 241, 69
Inflammasome pathways genes
NLRC5rs284388575706035316T>CD380, 132, 31
NLRC5rs71853205710137316A>GR484, 54, 5
NLRP5rs4719795653897619G>CR417, 119, 7
NLRP6rs7404441128447711T>CD521, 20, 2
NOD2rs20767565075688116A>GR347, 167, 29
JAK/STAT pathway genes
IL4RLiability Score0: 356
1: 124
2: 42
3: 18
4:2
5:1
IFNGR2rs49869583478729421C>GD528, 14, 1
IL10RBrs28341673464078821A>GR277, 212, 54
MAP/JNK pathway genes
PSMD2rs115451691840205423T>GR408, 123, 12
NFkB pathway genes
LTBRLiability Score0: 536
1: 7
IL17RBLiability Score0: 391
1: 129
2: 23
TNFRSF1Ars4149584644264312T>CD525, 17, 1
TNFRSF1Ars4149637644300112A>GD537, 6, 0
TNFRSF10Ars17620230602568T>CA143, 238, 162
TNFRSF10DLiability Score0: 134
1: 243
2: 138
3: 23
4: 5
TNFRSF11ALiability Score0: 99
1: 196
2: 185
3: 46
4: 17
TNFRSF14rs2234163249130622A>GD533, 10, 0
TNFRSF21Liability Score0: 435
1: 94
2: 14
MAP/JNK and NFkB pathway genes
TRAF1rs37618471236902399A>GD176, 265, 102
TRAF4rs359327782707533417A>GD535, 8, 0

█ = From morning fatigue exploratory analysis had a t-value of ≥2.0

= The order of the genotypes for which counts are provided is homozygous common, heterozygote, and homozygous rare.

Liability Score= The total number of rare allele occurrences across all SNPs and models tested for a given gene in the sample.

For the liability scores - The total number of rare allele occurrences across all SNPs for a given gene are coded as follows for IL4R as an example 0: 356 means that 356 patients did not have any doses of the rare alleles across IL4R; 1: 124 means that 124 patients had 1 dose of the rare alleles across IL4R; 2: 42 means that 42 patients had 2 doses of the rare alleles across IL4R; 3: 18 means that 18 patients had 3 doses of the rare alleles across IL4R; and 4: 2 means that 2 patients had 4 doses of the rare alleles across IL4R.

Abbreviations: A = additive model; Chr = chromosome; D = dominant model; I = intercept; IFNGR2 = Interferon gamma receptor 2; IL4R = Interleukin 4 receptor; IL10 = Interleukin 10; IL10RB = Interleukin 10 receptor subunit beta; IL12B= Interleukin 12B; IL17RB = Interleukin 17 receptor B; JAK/STAT = Janus kinase/signal transducers and activators of transcription; JNK = jun amino-terminal kinases; LTBR = Lymphotoxin beta receptor; MAP = mitogen-activated protein kinase; NFkB = nuclear factor-kappa beta; NLRC5 = Nod-Like receptor family, caspase recruitment domain containing 5; NLRP5 = Nod-Like receptor family, pyrin domain containing 5; NLRP6 = Nod-Like receptor family, pyrin domain containing 6; NOD2 = Nucleotide-Binding oligomerization domain containing 2; PSMD2 = Proteasome 26S subunit non-ATPase 2; PW1-L = piecewise 1 linear component; PW1-Q = piecewise 1 quadratic component; PW2-L = piecewise 2 linear component; PW2-Q = piecewise 2 quadratic component; R = recessive model; rs = reference SNP cluster identification number; SNP= single nucleotide polymorphism; TNFA = Tumor necrosis factor alpha; TNFRSF1A = Tumor necrosis factor receptor superfamily member 1A; TNFRSF10A = Tumor necrosis factor receptor superfamily member 10A; TNFRSF10D = Tumor necrosis factor receptor superfamily member 10D; TNFRSF11A = Tumor necrosis factor receptor superfamily member 11A; TNFRSF14 = Tumor necrosis factor receptor superfamily member 14; TNFRSF21 = Tumor necrosis factor receptor superfamily member 21; TRAF1 = Tumor necrosis factor receptor associated factor 1; TRAF4 = Tumor necrosis factor receptor associated factor 4

Table 2

Candidate Genotype Predictors of Intercept, Piecewise 1 and Piecewise 2 Linear and Quadratic Components for Evening Fatigue

Gene
Symbol
SNP IDPositionChrAllelesModelGenotype
Counts1
IPW1-LPW1-QPW2-LPW2-Q
Cytokine genes
IL12Brs32130941587507695T>CR292, 202, 49
IL12BLiability Score0: 275
1: 211
2: 49
3: 8
IL17Frs11465553521017586T>CD513, 30, 0
IL17Liability Score0: 511
1: 32
TNFArs1041981315407846C>AD233, 241, 69
Inflammasome pathways genes
CARD6rs10512747408417415T>CA441, 92, 10
CARD6Liability Score0: 401
1: 127
2: 15
NLRC5rs169651505705948416T>CD514, 27, 2
NLRP4rs178573735636990819C>GD500, 42, 1
NLRP5rs4719795653897619G>CR417, 119, 7
NLRP6rs7404441128447711T>CD521, 20, 2
NOD2rs20767565075688116A>GR347, 167, 29
JAK/STAT pathway genes
IFNGR2rs49869583478729421C>GD528, 14, 1
IL2RArs791587608869910A>GR149, 268, 126
IL2RBrs22840333753403422A>GD180, 261, 102
MAP/JNK pathway genes
IL17RDrs61742267571320353A>GD528, 18, 0
NFkB pathway genes
IL17RBrs2232346538928303T>CD520, 23, 0
IL17RBrs1043261538992763T>CR437, 96, 10
LTBRLiability Score0: 536
1: 7
TNFRSF14rs2234163249130622A>GD533, 10, 0

█ = From evening fatigue exploratory analysis had a t-value of ≥2.0

= The order of the genotypes for which counts are provided is homozygous common, heterozygote, and homozygous rare.

Liability Score= The total number of rare allele occurrences across all SNPs and models tested for a given gene in the sample.

For the liability scores - The total number of rare allele occurrences across all SNPs for a given gene are coded as follows for IL12B as an example 0: 275 means that 275 patients did not have any doses of the rare alleles across IL12B; 1: 211 means that 211 patients had 1 dose of the rare alleles across IL12B; 2: 49 means that 49 patients had 2 doses of the rare alleles across IL12B; 3: 8 means that 8 patients had 3 doses of the rare alleles across IL12B.

Abbreviations: A = additive model; CARD6 = Caspase recruitment domain family member 6; Chr = chromosome; D = dominant model; I = intercept; IFNGR2 = Interferon gamma receptor 2; IL2RA = Interleukin 2 receptor alpha; IL2RB = Interleukin 2 receptor beta; IL12B= Interleukin 12B; IL17 = Interleukin 17; IL17F = Interleukin 17F; IL17RB = Interleukin 17 receptor B; IL17RD = Interleukin 17 receptor D; JAK/STAT = janus kinase/signal transducers and activators of transcription; JNK = jun amino-terminal kinases; LTBR = Lymphotoxin beta receptor; MAP = mitogen-activated protein kinase; NFkB = nuclear factor-kappa beta; NLRC5 = Nod-Like receptor family, caspase recruitment domain containing 5; NLRP4 = Nod-Like receptor family, pyrin domain containing 4; NLRP5 = Nod-Like receptor family, pyrin domain containing 5; NLRP6 = Nod-Like receptor family, pyrin domain containing 6; NOD2 = Nucleotide-Binding oligomerization domain containing 2; PW1-L = piecewise 1 linear component; PW1-Q = piecewise 1 quadratic component; PW2-L = piecewise 2 linear component; PW2-Q = piecewise 2 quadratic component; R = recessive model; rs = reference SNP cluster identification number; SNP= single nucleotide polymorphism; TNFA = Tumor necrosis factor alpha; TNFRSF14 = Tumor necrosis factor receptor superfamily member 14.

Consistent with our previous studies [14, 63, 7780], recommendations from the literature [81, 82], rigorous quality controls for genomic data and the exploratory nature of our analyses, adjustments were not made for multiple testing. Since significant SNPs and liability scores identified in the exploratory analysis were evaluated further in the HLM analyses that controlled for population stratification (i.e., genomic and self-reported estimates of race and ethnicity), and other variations in the same gene, the significant independent genetic associations reported are unlikely to be due solely to chance.

4.4 Estimation of polymorphism function

Based on the results of the HLM analyses, the SNPs associated with inter-individual differences in the initial levels or trajectories of morning or evening fatigue were evaluated for their potential impact on protein function using two bioinformational tools (i.e., Sorting Intolerant From Tolerant (SIFT) algorithm [83] and Polymorphism Phenotyping v2 (PolyPhen-2) [84]). SIFT predicts whether a SNP in a coding region results in an amino acid substitution that may affect protein function. This prediction is based on an analysis of the conservation of amino acid residues in sequence alignments of closely related sequences [84]. PolyPhen-2 compares the SNP to sequence-based and structure-based predictive features to predict the functional significance of the SNP [84]. Results from the bioinformational tools are described in the discussion.

5. Results

5.1 Sample characteristics

As summarized in Table 3, of the 543 patients in the study, the majority of the patients were female, white, diagnosed with breast cancer, and were treated with CTX on a 21-day cycle. Most patients were well educated, married or partnered, and currently not employed. At enrollment, patients reported clinically meaningful sleep disturbance and anxiety levels. Morning and evening fatigue scores at enrollment were just below the cutoff for clinically meaningful levels (i.e., ≥3.2 for morning fatigue, ≥5.6 for evening fatigue) [70].

Table 3

Sample Characteristics (n=543)

CharacteristicsMean (SD)
Age (years)57.1 (11.7)
Education (years)16.3 (3.0)
Body mass index (kg/m)26.3 (5.8)
Hemoglobin (gm/dL)11.7 (1.4)
Karnofsky Performance Status score80.7 (11.8)
Self-administered Comorbidity Questionnaire score5.5 (3.0)
Time since cancer diagnosis (years)2.5 (4.4)
Number prior cancer treatments1.9 (1.6)
Number of metastatic sites including lymph node involvement1.4 (1.3)
Number of metastatic sites excluding lymph node involvement0.9 (1.2)
Symptom scores at enrollment
Lee Fatigue Scale: morning fatigue3.0 (2.2)
Lee Fatigue Scale: evening fatigue5.3 (2.1)
Lee Fatigue Scale: morning energy4.5 (2.2)
Lee Fatigue Scale: evening energy3.5 (1.9)
Center for Epidemiological Studies-Depression Scale12.5 (9.3)
General Sleep Disturbance Scale51.9 (19.3)
Trait Anxiety35.0 (10.3)
State Anxiety33.3 (11.9)

% (N)

Gender (female)80.8 (439)
Ethnicity
White69.4 (377)
Black6.8 (77)
Asian/Pacific Islander12.7 (69)
Hispanic/Mixed/Other11.0 (60)
Married or partnered68.0 (369)
Lives alone19.5 (106)
Currently employed34.3 (186)
Child care responsibilities (% yes)23.6 (128)
Exercise on a regular basis (% yes)70.7 (413)
Cancer diagnosis
Breast43.6 (237)
Gastrointestinal26.0 (141)
Gynecological21.0 (114)
Lung9.4 (51)
Chemotherapy cycle length
14 days36.1 (196)
21 days54.9 (298)
28 days9.0 (49)
Previous cancer treatments89.9 (450)
Pain present (% yes)72.9 (396)

Abbreviations: gm/dL = grams per deciliter; kg/m = kilograms per meter squared; SD = standard deviation

5.2 Changes in morning fatigue severity

HLM was used to examine how morning fatigue scores changed within the two cycles of CTX, controlling for self-reported and genomic estimates of race and ethnicity. The estimates for the initial piecewise model are presented in Table 4. Since the model was unconditional (i.e., no covariates included in the model), the average morning fatigue severity score at enrollment (i.e., 3.011 on a 0 to 10 scale) represents the intercept. The estimated linear piecewise rates of change were 1.192 and 0.532 (both p<.0001) for piecewise linear 1 and piecewise linear 2, respectively. The estimated quadratic piecewise rates of change were −.599 and −.153 (both p<.0001) for piecewise quadratic 1 and piecewise quadratic 2, respectively. Figure 1A displays the unconditional model for mean morning fatigue scores over the two cycles of CTX.

An external file that holds a picture, illustration, etc.
Object name is nihms845338f1.jpg

A–F – Unconditional piecewise model of mean morning fatigue scores for six assessment points over two cycles of chemotherapy (A). Influence of the recessive model (TT+TC vs. CC) of the rare C allele in IL12B rs3213094 on the inter-individual differences in the intercept for morning fatigue (B). Influence of the dominant model (CC vs. CA+AA) of the rare A allele in TNFA rs1041981 on the slope parameters for morning fatigue (C). Influence of the recessive model (AA+AG vs. GG) of the rare G allele in NOD2 rs2076756 on the inter-individual differences in the intercept for morning fatigue (D). Influence of the recessive model (GG+GC vs. CC) of the rare C allele in NLRP5 rs471979 on the slope parameters for morning fatigue (E). Influence of the dominant model (TT vs. TC+CC) of the rare C allele in NLRP6 rs74044411 on the slope parameters for morning fatigue (F).

Table 4

Hierarchical Linear Models for Morning Fatigue

Cytokine Genes
Morning Fatigue and IL12B rs3213094Coefficient (SE)
Unconditional ModelFinal Model
Fixed effects
Intercept3.011 (.099)+3.008 (.099)+
Ethnicitya
Black versus White−.845 (.917)−.735 (.913)
Asian versus White.214 (.675).265 (.672)
Hispanic versus White.328 (.331).348 (.329)
Ancestry informative markers principal componentsb
PC1−.102 (.104)−.105 (.103)
PC2−.052 (.088)−.031 (.088)
PC3.008 (.039).008 (.039)
PW1 – linear1.192 (.148)+1.191 (.148)+
PW1 – quadratic−.599 (.071)+−.599 (.071)+
PW2 – linear.532(.096)+.531 (.096)+
PW2 – quadratic−.153 (.031)+−.153 (.031)+
Time invariant covariates
Intercept - IL12B rs3213094−.760 (.319)*
Variance components
In intercept3.552+3.512+
Goodness-of-fit deviance (parameters estimated)11026.531695 (13)+11020.881398 (14)
Model comparison χ (df)5.650 (1)*
Morning Fatigue and TNFA rs1041981Coefficient (SE)
Final Model
Fixed effects
Intercept3.011 (.099)+
Ethnicitya
Black versus White−.807 (.917)
Asian versus White.187 (.675)
Hispanic versus White.304 (.331)
Ancestry informative markers principal componentsb
PC1−.106 (.103)
PC2−.045 (.088)
PC3.015 (.039)
PW1 – linear1.186 (.148)+
PW1 – quadratic−.597 (.071)+
PW2 – linear.533 (.096)+
PW2 – quadratic−.153 (.031)+
Time invariant covariates
PW1 – linear - TNFA rs1041981−.641 (.278)*
PW1 – quadratic - TNFA rs1041981.264 (.131)*
Variance components
In intercept3.549+
Goodness-of-fit deviance (parameters estimated)11019.804052 (15)
Model comparison χ (df)6.728 (2)*
Inflammasome Pathway Genes
Morning Fatigue and NLRP5 rs471979Coefficient (SE)
Final Model
Fixed effects
Intercept3.011 (.099)+
Ethnicitya
Black versus White−.852 (.918)
Asian versus White.212 (.676)
Hispanic versus White.310 (.332)
Ancestry informative markers principal componentsb
PC1−.101 (.104)
PC2−.051 (.088)
PC3.004 (.039)
PW1 – linear1.190 (.148)+
PW1 – quadratic−.598 (.071)+
PW2 – linear.533 (.096)+
PW2 – quadratic−.153 (.031)+
Time invariant covariates
PW1 – linear - NLRP5 rs471979−3.292 (1.258)*
PW1 – quadratic - NLRP5 rs4719791.323 (.595)*
Variance components
In intercept3.566+
Goodness-of-fit deviance (parameters estimated)11016.309635 (15)
Model comparison χ (df)10.222 (2)*
Morning Fatigue and NLRP6 rs74044411Coefficient (SE)
Final Model
Fixed effects
Intercept3.010 (.099)+
Ethnicitya
Black versus White−.727 (.914)
Asian versus White.229 (.672)
Hispanic versus White.339 (.330)
Ancestry informative markers principal componentsb
PC1−.128 (.103)
PC2−.068 (.087)
PC3.011 (.039)
PW1 – linear1.186 (.148)+
PW1 – quadratic−.597 (.071)+
PW2 – linear.534 (.096)+
PW2 – quadratic−.153 (.031)+
Time invariant covariates
Intercept
PW1 – linear - NLRP6 rs74044411−1.675 (.785)*
PW1 – quadratic - NLRP6 rs74044411.539 (.373)
Variance components
In intercept3.522+
Goodness-of-fit deviance (parameters estimated)11010.712889 (15)
Model comparison χ (df)15.819 (2)**
Morning Fatigue and NOD2 rs2076756Coefficient (SE)
Final Model
Fixed effects
Intercept3.010 (.099)+
Ethnicitya
Black versus White−.916 (.912)
Asian versus White.280 (.671)
Hispanic versus White.359 (.329)
Ancestry informative markers principal componentsb
PC1−.096 (.103)
PC2−.068 (.087)
PC3.009 (.039)
PW1 – linear1.192 (.148)+
PW1 – quadratic−.599 (.071)+
PW2 – linear.532 (.096)+
PW2 – quadratic−.153 (.031)+
Time invariant covariates
Intercept - NOD2 rs2076756−.960 (.383)*
Variance components
In intercept3.507+
Goodness-of-fit deviance (parameters estimated)11020.293155 (14)
Model comparison χ (df)6.239 (1)*
JAK/STAT Pathway Genes
Morning Fatigue and IL4R Liability ScoreCoefficient (SE)
Final Model
Fixed Effects
Intercept3.010 (.099)+
Ethnicitya
Black versus White−.772 (.917)
Asian versus White.233 (.674)
Hispanic versus White.336 (.331)
Ancestry informative markers principal componentsb
PC1−.103 (.103)
PC2−.061 (.088)
PC3.012 (.039)
PW1 – linear1.191 (.148)+
PW1 – quadratic−.599 (.071)+
PW2 – linear.532 (.096)+
PW2 – quadratic−.153 (.031)+
Time invariant covariates
Intercept
PW1 – linear - IL4R Liability Score−.401(.168)*
PW1 – quadratic - IL4R Liability Score.167 (.079)*
Variance components
In intercept3.548+
Goodness-of-fit deviance (parameters estimated)11019.542001 (15)
Model comparison χ (df)6.990 (2)*
NFkB pathway Genes
Morning Fatigue and IL17RB Liability ScoreCoefficient (SE)
Final Model
Fixed effects
Intercept3.011 (.099)+
Ethnicitya
Black versus White−.843 (.913)
Asian versus White.256 (.672)
Hispanic versus White.324 (.330)
Ancestry informative markers principal componentsb
PC1−.111 (.103)
PC2−.037 (.088)
PC3.013 (.039)
PW1 – linear1.191 (.148)+
PW1 – quadratic−.599 (.071)+
PW2 – linear.531 (.096)+
PW2 – quadratic−.153 (.031)+
Time invariant covariates
Intercept - IL17RB Liability Score−.352 (.162)*
Variance components
In intercept3.519+
Goodness-of-fit deviance (parameters estimated)11021.825332 (14)
Model comparison χ (df)4.706 (1)*
Morning Fatigue and TNFRSF10A rs17620Coefficient (SE)
Final Model
Fixed effects
Intercept3.010 (.099)+
Ethnicitya
Black versus White−.873 (.916)
Asian versus White.235 (.674)
Hispanic versus White.330 (.331)
Ancestry informative markers principal componentsb
PC1−.099 (.103)
PC2−.073 (.088)
PC3.012 (.039)
PW1 – linear1.194 (.148)+
PW1 – quadratic−.601 (.071)+
PW2 – linear.535 (.096)+
PW2 – quadratic−.154 (.031)+
Time invariant covariates
PW1 – linear - TNFRSF10A rs17620.402 (.189)*
PW1 – quadratic - TNFRSF10A rs17620−.193 (.093)*
PW2 – linear - TNFRSF10A rs17620.197 (.127)
PW2 – quadratic - TNFRSF10A rs17620−.051 (.041)
Variance components
In intercept3.544+
Goodness-of-fit deviance (parameters estimated)11018.710558 (17)
Model comparison χ (df)7.821 (4)
Morning Fatigue and TNFRSF10D Liability ScoreCoefficient (SE)
Final Model
Fixed effects
Intercept3.011 (.099)+
Ethnicitya
Black versus White−.810 (.917)
Asian versus White.203 (.675)
Hispanic versus White.323 (.331)
Ancestry informative markers principal componentsb
PC1−.106 (.104)
PC2−.048 (.088)
PC3.008 (.039)
PW1 – linear1.195 (.148)+
PW1 – quadratic−.601 (.071)+
PW2 – linear.529 (.096)+
PW2 – quadratic−.152 (.031)+
Time invariant covariates
PW2 – linear - TNFRSF10D Liability Score−.211 (.094)*
PW2 – quadratic - TNFRSF10D Liability Score.057 (.033)
Variance components
In intercept3.552+
Goodness-of-fit deviance (parameters estimated)11018.923385 (15)
Model comparison χ (df)7.608 (2)*
Morning Fatigue and TNFRSF11A Liability ScoreCoefficient (SE)
Final Model
Fixed effects
Intercept3.011 (.099)+
Ethnicitya
Black versus White−.866 (.914)
Asian versus White.172 (.673)
Hispanic versus White.330 (.330)
Ancestry informative markers principal componentsb
PC1−.107 (.103)
PC2−.046 (.087)
PC3.005 (.039)
PW1 – linear1.193 (.148)+
PW1 – quadratic−.600 (.071)+
PW2 – linear.535 (.096)+
PW2 – quadratic−.154 (.031)+
Time invariant covariates
PW1 – linear - TNFRSF11A Liability Score−.381 (.141)*
PW1 – quadratic - TNFRSF11A Liability Score.174 (.070)*
PW2 – linear - TNFRSF11A Liability Score−.130 (.097)
PW2 – quadratic - TNFRSF11A Liability Score.022 (.031)
Variance components
In intercept3.532+
Goodness-of-fit deviance (parameters estimated)11009.606255 (17)
Model comparison χ (df)16.925 (4)*
Morning Fatigue and TNFRSF14 rs2234163Coefficient (SE)
Final Model
Fixed effects
Intercept3.011 (.099)+
Ethnicitya
Black versus White−.903 (.913)
Asian versus White.112 (.673)
Hispanic versus White.311 (.330)
Ancestry informative markers principal componentsb
PC1−.104 (.103)
PC2−.055 (.087)
PC3.010 (.039)
PW1 – linear1.192 (.148)+
PW1 – quadratic−.599 (.071)+
PW2 – linear.532 (.096)+
PW2 – quadratic−.153 (.031)+
Time invariant covariates
Intercept - TNFRSF14 rs22341631.426 (.638)*
Variance components
In intercept3.514+
Goodness-of-fit deviance (parameters estimated)11021.551571 (14)
Model comparison χ (df)4.980 (1)*
Morning Fatigue and TNFRSF21 Liability ScoreCoefficient (SE)
Final Model
Intercept3.012 (.099)+
Ethnicitya
Black versus White−.815 (.913)
Asian versus White.281 (.672)
Hispanic versus White.310 (.330)
Ancestry informative markers principal componentsb
PC1−.104 (.103)
PC2−.069 (.088)
PC3.001 (.039)
PW1 – linear1.194 (.148)+
PW1 – quadratic−.601 (.071)+
PW2 – linear.531 (.096)+
PW2 – quadratic−.153 (.031)+
Time invariant covariates
Intercept TNFRSF21 Liability Score.416 (.183)*
Variance components
In intercept3.517+
Goodness-of-fit deviance (parameters estimated)11021.369941 (14)
Model comparison χ (df)5.162 (1)*
Self-reported ethnicity – represented by three “dummy” coded variables
Ancestry informative markers - represented by the first three PCs (i.e., PC1, PC2, PC3)
p<.05,
p<.001,
p<.0001

Abbreviations: df = degrees of freedom; IL4R = Interleukin 4 receptor; IL12B= Interleukin 12B; IL17RB = Interleukin 17 receptor B; JAK/STAT = Janus kinase/signal transducers and activators of transcription; NFkB = nuclear factor-kappa beta; NLRC5 = Nod-Like receptor family, caspase recruitment domain containing 5; NLRP5 = Nod-Like receptor family, pyrin domain containing 5; NLRP6 = Nod-Like receptor family, pyrin domain containing 6; NOD2 = Nucleotide-Binding oligomerization domain containing 2; PC=principal component; PW1 = piecewise 1 PW2 = piecewise 2; rs = reference SNP cluster identification number; SE = standard error; TNFA = Tumor necrosis factor alpha; TNFRSF10A = Tumor necrosis factor receptor superfamily member 10A; TNFRSF10D = Tumor necrosis factor receptor superfamily member 10D; TNFRSF11A = Tumor necrosis factor receptor superfamily member 11A; TNFRSF14 = Tumor necrosis factor receptor superfamily member 14; TNFRSF21 = Tumor necrosis factor receptor superfamily member 21

5.3 Genomic predictors of inter-individual differences in morning fatigue

Table 4 shows the final HLM models for morning fatigue. For the cytokine genes, two SNPs were associated with inter-individual differences in morning fatigue. Figure 1B illustrates the adjusted initial level of morning fatigue (i.e., intercept) based on a recessive model for IL12B rs3213094 (i.e., TT+TC vs. CC). Figure 1C illustrates the predicted changes in the trajectory of morning fatigue (i.e., slope) based on a dominant model for TNFA rs1041981 (i.e., CC vs. CA+AA).

For the inflammasome pathway genes, three SNPs were associated with changes in morning fatigue. Figure 1D illustrates the adjusted initial levels of morning fatigue based on the recessive model for nucleotide-binding oligomerization domain containing 2 (NOD2) rs2076756 (i.e., AA+AG vs. GG). Figures 1E and 1F respectively, illustrate the predicted changes in the trajectories of morning fatigue based on the recessive model for non-like receptor family, pyrin containing domain 5 (NLRP5) rs471979 (i.e., GG+GC vs. CC) and a dominant model for NLRP6 rs74044411 (i.e., TT vs. TC+CC).

For the JAK/STAT pathway, the liability score for all IL4R, was associated with inter-individual differences in the slope of morning fatigue (Figure 2A).

An external file that holds a picture, illustration, etc.
Object name is nihms845338f2.jpg

A–D – Influence of the liability score for the sum of the occurrence of the rare alleles in IL4R on the slope parameters for morning fatigue (A). Influence of the dominant model (AA vs. AG+GG) of the rare G allele for TNFRSF14 rs2234163 on the inter-individual differences in the intercept for morning fatigue (B). Influence of the liability score for the sum of the occurrence of the rare alleles in IL17RB, on the inter-individual differences in the intercept for morning fatigue (C). Influence of the liability score for the sum of the occurrence of the rare alleles in TNFRSF21 on the inter-individual differences in the intercept for morning fatigue (D).

For the NFkB pathway genes, two SNPs and four liability scores were associated with inter-individual differences in morning fatigue. Figure 2B illustrates the adjusted initial level of morning fatigue based on the dominant model for tumor necrosis factor receptor super family, member 14 (TNFRSF14) rs2234163 (i.e., AA vs. AG+GG). Figures 2C and 2D respectively, illustrate the adjusted initial levels of morning fatigue based on the liability scores for IL17RB and TNFRSF21. Figures 3A, 3B, and 3C, respectively, illustrate the predicted changes in the trajectories of morning fatigue for the additive model for TNFRSF10A rs17620 (i.e., TT vs. TC vs. CC) and the liability scores for TNFRSF10D and TNFRSF11A.

An external file that holds a picture, illustration, etc.
Object name is nihms845338f3.jpg

A–C – Influence of the additive model (TT vs. TC vs. CC) of the rare C allele for TNFRSF10A rs17620 on the slope parameters for morning fatigue (A). Influence of the liability score for the sum of the occurrence of the rare alleles in TNFRSF10D on the slope parameters for morning fatigue (B). Influence of the liability score for the sum of the occurrence of the rare alleles in TNFRSF11A on the slope parameters for morning fatigue (C).

5.4 Changes in evening fatigue severity

HLM was used to examine how evening fatigue scores changed within the two cycles of CTX, controlling for self-reported and genomic estimates of race and ethnicity. The estimates for the initial piecewise model are presented in Table 5. Since the model was unconditional (i.e., no covariates), the intercept represents the average evening fatigue severity score at enrollment (i.e., 5.310 on a 0 to 10 scale). The estimated linear piecewise rates of change were 0.601 and 0.394 (both, p<.0001) for piecewise linear 1 and piecewise linear 2, respectively. The estimated quadratic piecewise rates of change were −.306 and −.113 (both, p<.0001) for piecewise quadratic 1 and piecewise quadratic 2, respectively. Figure 4A displays the unconditional model for mean evening fatigue scores over two cycles of CTX.

An external file that holds a picture, illustration, etc.
Object name is nihms845338f4.jpg

A–D – Unconditional piecewise model of mean evening fatigue scores for six assessment points over two cycles of chemotherapy (A). Influence of the recessive model (TT+TC vs. CC) of the rare C allele in IL12B rs3213094 on the inter-individual differences in the intercept for evening fatigue (B). Influence of the liability score for the sum of the occurrence of the rare alleles in IL12B on the inter-individual differences in the intercept for evening fatigue (C). Influence of the dominant model (CC vs. CA+AA) of the rare A allele in TNFA rs1041981 on the slope parameters for evening fatigue (D).

Table 5

Hierarchical Linear Models for Evening Fatigue

Cytokine Genes
Evening Fatigue and IL12B rs3213094Coefficient (SE)
Unconditional ModelFinal Model
Fixed effects
Intercept5.310 (.090)+5.307 (.090)+
Ethnicitya
Black versus White−1.584 (.824)−1.501 (.822)
Asian versus White−.269 (.608)−.231 (.606)
Hispanic versus White−.348 (.298)−.332 (.297)
Ancestry informative markers principal components
PC1−.070 (.093)−.072 (.093)
PC2−.044 (.079)−.029 (.079)
PC3−.009 (.035)−.008 (.035)
PW 1 – linear.601 (.137)+.601 (.137)+
PW 1 – quadratic−.306 (.066)+−.306 (.066)+
PW 2 – linear.394 (.088)+.394 (.088)+
PW 2 – quadratic−.113 (.028)+−.113 (.028)+
Time invariant covariates
Intercept IL12B rs3213094−.573 (.288)*
Variance components
In intercept2.867+2.843+
Goodness-of-fit deviance (parameters estimated)10399.632349 (13)+10395.680232 (14)
Model comparison χ (df)3.952 (1)*
Evening Fatigue and IL12B Liability ScoreCoefficient (SE)
Final Model
Fixed effects
Intercept5.308 (.090)+
Ethnicitya
Black versus White−1.579 (.820)
Asian versus White−.322 (.605)
Hispanic versus White−.347 (.297)
Ancestry informative markers principal componentsb
PC1−.086 (.093)
PC2−.021 (.079)
PC3−.003 (.035)
PW 1 – linear.600 (.137) +
PW 1 – quadratic−.306 (.066) +
PW 2 – linear.393 (.088) +
PW 2 – quadratic−.112 (.028) +
Time invariant covariates
Intercept - IL12B Liability Score−.258 (.115)*
Variance components
In intercept2.838+
Goodness-of-fit deviance (parameters estimated)10394.643652 (14)
Model comparison χ (df)4.989 (1)*
Evening Fatigue and TNFA rs1041981Coefficient (SE)
Final Model
Fixed effects
Intercept5.309 (.090)+
Ethnicitya
Black versus White−1.548 (.822)
Asian versus White−.293 (.606)
Hispanic versus White−.369 (.298)
Ancestry informative markers principal componentsb
PC1−.073 (.093)
PC2−.038 (.079)
PC3−.002 (.035)
PW 1 – linear.596 (.137)+
PW 1 – quadratic−.304 (.066)+
PW 2 – linear.395 (.088)+
PW 2 – quadratic−.113 (.028)+
Time invariant covariates
Intercept
PW 1 – linear - TNFA rs1041981−.602 (.256)*
PW 1 – quadratic - TNFA rs1041981.252 (.121)*
Variance components
In intercept2.852+
Goodness-of-fit deviance (parameters estimated)10392.936781 (15)
Model comparison χ (df)6.696 (2)*
Inflammasome Pathway Genes
Evening Fatigue and CARD6 rs10512747Coefficient (SE)
Final Model
Fixed effects
Intercept5.309 (.089)+
Ethnicitya
Black versus White−1.591 (.819)
Asian versus White−.350 (.605)
Hispanic versus White−.397 (.297)
Ancestry informative markers principal componentsb
PC1−.057 (.093)
PC2−.043 (.078)
PC3−.008 (.035)
PW 1 – linear.599 (.137)+
PW 1 – quadratic−.306 (.066)+
PW 2 – linear.394 (.088)+
PW 2 – quadratic−.113 (.028)+
Time invariant covariates
Intercept - CARD6 rs10512747−.441 (.176)*
Variance components
In intercept2.829+
Goodness-of-fit deviance (parameters estimated)10393.393966 (14)
Model comparison χ (df)6.238 (1)*
Evening Fatigue and NLRP4 rs17857373Coefficient (SE)
Final Model
Fixed effects
Intercept5.308 (.090)+
Ethnicitya
Black versus White−1.600 (.821)
Asian versus White−.298 (.605)
Hispanic versus White−.365 (.297)
Ancestry informative markers principal componentsb
PC1−.061 (.093)
PC2−.046 (.079)
PC3−.003 (.035)
PW 1 – linear.601 (.137)+
PW 1 – quadratic−.306 (.066)+
PW 2 – linear.394 (.088)+
PW 2 – quadratic−.113 (.028)+
Time invariant covariates
Intercept - NLRP4 rs17857373−.603 (.291)*
Variance components
In intercept2.841+
Goodness-of-fit deviance (parameters estimated)10395.340962 (14)
Model comparison χ (df)4.291 (1)*
Evening Fatigue and NLRP6 rs74044411Coefficient (SE)
Final Model
Fixed effects
Intercept5.309 (.090)+
Ethnicitya
Black versus White−1.491 (.822)
Asian versus White−.257 (.606)
Hispanic versus White−.339 (.297)
Ancestry informative markers principal componentsb
PC1−.093 (.093)
PC2−.060 (.079)
PC3−.007 (.035)
PW 1 – linear.598 (.136)+
PW 1 – quadratic−.306 (.066)+
PW 2 – linear.395 (.088)+
PW 2 – quadratic−.113 (.028)+
Time invariant covariates
Intercept
PW 1 – linear - NLRP6 rs74044411−1.417 (.682)*
PW 1 – quadratic - NLRP6 rs74044411.449 (.322)
Variance components
In intercept2.847+
Goodness-of-fit deviance (parameters estimated)10385.256709 (15)
Model comparison χ (df)14.376 (2)**
Evening Fatigue and NOD2 rs2076756Coefficient (SE)
Final Model
Fixed effects
Intercept5.308 (.089)+
Ethnicitya
Black versus White−1.684 (.813)*
Asian versus White−.177 (.599)
Hispanic versus White−.304 (.294)
Ancestry informative markers principal componentsb
PC1−.061 (.092)
PC2−.067 (.078)
PC3−.008 (.035)
PW 1 – linear.601 (.137)+
PW 1 – quadratic−.306 (.066)+
PW 2 – linear.394 (.088)+
PW 2 – quadratic−.113 (.028)+
Time invariant covariates
Intercept - NOD2 rs2076756−1.350 (.342)+
Variance components
In intercept2.777+
Goodness-of-fit deviance (parameters estimated)10384.268252 (14)
Model comparison χ (df)15.364 (1)+
MAP/JNK Pathway Genes
Evening Fatigue and IL17RD rs61742267Coefficient (SE)
Final Model
Fixed effects
Intercept5.310 (.090)+
Ethnicitya
Black versus White−1.539 (.821)
Asian versus White−.265 (.605)
Hispanic versus White−.319 (.297)
Ancestry informative markers principal componentsb
PC1−.059 (.093)
PC2−.044 (.079)
PC3−.013 (.035)
PW 1 – linear.601 (.137)+
PW 1 – quadratic−.306 (.066)+
PW 2 – linear.393 (.088)+
PW 2 – quadratic−.112 (.028)+
Time invariant covariates
Intercept - IL17RD rs61742267−.933 (.429)*
Variance components
In intercept2.840+
Goodness-of-fit deviance (parameters estimated)10394.915944 (14)
Model comparison χ (df)4.716 (1)*
NfKB Pathway Genes
Evening Fatigue and IL17RB rs2232346 and
IL17RB rs1043261
Coefficient (SE)
Final Model
Fixed effects
Intercept5.308 (.089)+
Ethnicitya
Black versus White−1.573 (.817)
Asian versus White−.179 (.603)
Hispanic versus White−.344 (.296)
Ancestry informative markers principal componentsb
PC1−.065 (.092)
PC2−.037 (.078)
PC3−.009 (.035)
PW 1 – linear.600 (.137) +
PW 1 – quadratic−.306 (.066) +
PW 2 – linear.394 (.088) +
PW 2 – quadratic−.113 (.028) +
Time invariant covariates
Intercept - IL17RB rs2232346−.791 (.388)*
Intercept - IL17RB rs1043261−1.456 (.584)*
Variance components
In intercept2.809+
Goodness-of-fit deviance (parameters estimated)10389.389046 (15)
Model comparison χ (df)10.243 (2)*
Evening Fatigue and LTBR Liability ScoreCoefficient (SE)
Final Model
Fixed effects
Intercept5.309 (.090)+
Ethnicitya
Black versus White−1.562 (.826)
Asian versus White−.277 (.609)
Hispanic versus White−.346 (.299)
Ancestry informative markers principal componentsb
PC1−.072 (.093)
PC2−.044 (.079)
PC3−.010 (.035)
PW 1 – linear.604 (.136)+
PW 1 – quadratic−.308 (.066)+
PW 2 – linear.396 (.088)+
PW 2 – quadratic−.113 (.028)+
Time invariant covariates
Intercept
PW 1 – linear - LTBR Liability Score−3.188 (1.080)*
PW 1 – quadratic - LTBR Liability Score1.540 (.513)*
Variance components
In intercept2.885+
Goodness-of-fit deviance (parameters estimated)10390.660476 (15)
Model comparison χ (df)8.972 (2)*
Evening Fatigue and TNFRSF14 rs2234163Coefficient (SE)
Final Model
Fixed effects
Intercept5.309 (.090)+
Ethnicitya
Black versus White−1.648 (.818)
Asian versus White−.383 (.605)
Hispanic versus White−.366 (.296)
Ancestry informative markers principal componentsb
PC1−.072 (.092)
PC2−.048 (.078)
PC3−.007 (.035)
PW 1 – linear.600 (.137)+
PW 1 – quadratic−.306 (.066)+
PW 2 – linear.394 (.088)+
PW 2 – quadratic−.113 (.028)+
Time invariant covariates
Intercept - TNFRSF14 rs22341631.590 (.572)*
Variance components
In intercept2.821+
Goodness-of-fit deviance (parameters estimated)10391.966279 (14)
Model comparison χ (df)7.666 (1)*
Self-reported ethnicity – represented by three “dummy” coded variables
Ancestry informative markers - represented by the first three PCs (i.e., PC1, PC2, PC3)
p<.05,
p<.001,
p<.0001

Abbreviations: CARD6 = Caspase recruitment domain family member 6; df = degrees of freedom; IL12B= Interleukin 12B; IL17RB = Interleukin 17 receptor B; IL17RD = Interleukin 17 receptor D; JAK = jaunts kinase transducers and activators of transcription; LTBR = Lymphotoxin beta receptor; MAP = mitogen-activated protein kinase; NFkB = nuclear factor-kappa beta; NLRP4 = Nod-Like receptor family, pyrin domain containing 4; NLRP6 = Nod-Like receptor family, pyrin domain containing 6; NOD2 = Nucleotide-Binding oligomerization domain containing 2; PC=principal component; PW1 = piecewise 1; PW2 = piecewise 2; rs = reference SNP cluster identification number; SE = standard error; TNFA = Tumor necrosis factor alpha; TNFRSF14 = Tumor necrosis factor receptor superfamily member 14.

5.5 Genomic predictors of inter-individual differences in evening fatigue

Table 5 shows the final HLM models for evening fatigue. For the cytokine genes, two SNPs and one liability score were associated with inter-individual differences in evening fatigue. Figures 4B and 4C respectively, illustrate the adjusted initial level of evening fatigue based on the recessive model for IL12B rs3213094 (i.e., TT+TC vs. CC) and the liability score for IL12B. Figure 4D illustrates the predicted changes in the trajectories of evening fatigue for a dominant model for TNFA rs1041981 (i.e., CC vs. CA+AA).

For the inflammasome pathway genes, four SNPs were associated with evening fatigue. Figures 5A, 5B, and 5C respectively, illustrate the adjusted initial levels of evening fatigue based on the additive model for capsase recruitment domain family member 6 (CARD6) rs10512747 (i.e., TT vs. TC vs. CC); the dominant model for NLRP4 rs17857373 (i.e., CC vs. CG+GG) and the recessive model for NOD2 rs2076756 (i.e., AA+AG vs. GG). Figure 5D illustrates the predicted changes in the trajectory of evening fatigue based on the dominant model for NLRP6 rs74044411 (i.e., TT vs. TC+CC).

An external file that holds a picture, illustration, etc.
Object name is nihms845338f5.jpg

A–E – Influence of the additive model (TT vs. TC vs. CC) of the rare C allele in CARD6 rs10512747 on the inter-individual differences in the intercept for evening fatigue (A). Influence of the dominant model (CC vs. CG+GG) of the rare G allele for NLRP4 rs17857373 on the inter-individual differences in the intercept for evening fatigue (B). Influence of the recessive model (AA+AG vs. GG) of the rare G allele for NOD2 rs2076756 on the inter-individual differences in the intercept for evening fatigue (C). Influence of the dominant model (TT vs. TC+CC) of the rare C allele for NLRP6 rs74044411 on the slope parameters for evening fatigue (D). Influence of the dominant model (AA vs. AG+GG) for the rare G allele for IL17RD rs61742267 on the inter-individual differences in the intercept for evening fatigue (E).

For the MAP/JNK pathway genes, Figure 5E illustrates the adjusted initial level of evening fatigue based on the dominant model for IL17RD rs61742267 (i.e., AA vs. AG+GG).

For the NFkB pathway genes, three SNPs and one liability score were associated with inter-individual differences in evening fatigue. Figure 6A illustrates the adjusted initial level of evening fatigue based on the dominant model for IL17RB rs2232346 (i.e., TT vs. TC+CC) and the recessive model for IL17RB rs1043261 (i.e., TT+TC vs. CC). Figures 6B and 6C respectively, illustrate the adjusted initial level of evening fatigue based on the dominate model for TNFRSF14 rs2234163 (i.e., AA vs. AG+GG) and the predicted change in the trajectory of evening fatigue based on the liability score for lymphotoxin beta receptor (LTBR).

An external file that holds a picture, illustration, etc.
Object name is nihms845338f6.jpg

A–C – Influence of the dominant model (TT vs. TC+CC) of the rare C allele for IL17RB rs2232346 and the recessive model (TT+TC vs. CC) of the rare C allele for IL17RB rs1043261 on the inter-individual differences in the intercept for evening fatigue (A). Influence of the dominant model (AA vs. AG+GG) of the rare G allele for TNFRSF14 rs2234163 on the inter-individual differences in the intercept for evening fatigue (B). Influence of the liability score for the sum of the occurrence of the rare alleles in LTBR on the slope parameters for evening fatigue (C).

5.1 Sample characteristics

As summarized in Table 3, of the 543 patients in the study, the majority of the patients were female, white, diagnosed with breast cancer, and were treated with CTX on a 21-day cycle. Most patients were well educated, married or partnered, and currently not employed. At enrollment, patients reported clinically meaningful sleep disturbance and anxiety levels. Morning and evening fatigue scores at enrollment were just below the cutoff for clinically meaningful levels (i.e., ≥3.2 for morning fatigue, ≥5.6 for evening fatigue) [70].

Table 3

Sample Characteristics (n=543)

CharacteristicsMean (SD)
Age (years)57.1 (11.7)
Education (years)16.3 (3.0)
Body mass index (kg/m)26.3 (5.8)
Hemoglobin (gm/dL)11.7 (1.4)
Karnofsky Performance Status score80.7 (11.8)
Self-administered Comorbidity Questionnaire score5.5 (3.0)
Time since cancer diagnosis (years)2.5 (4.4)
Number prior cancer treatments1.9 (1.6)
Number of metastatic sites including lymph node involvement1.4 (1.3)
Number of metastatic sites excluding lymph node involvement0.9 (1.2)
Symptom scores at enrollment
Lee Fatigue Scale: morning fatigue3.0 (2.2)
Lee Fatigue Scale: evening fatigue5.3 (2.1)
Lee Fatigue Scale: morning energy4.5 (2.2)
Lee Fatigue Scale: evening energy3.5 (1.9)
Center for Epidemiological Studies-Depression Scale12.5 (9.3)
General Sleep Disturbance Scale51.9 (19.3)
Trait Anxiety35.0 (10.3)
State Anxiety33.3 (11.9)

% (N)

Gender (female)80.8 (439)
Ethnicity
White69.4 (377)
Black6.8 (77)
Asian/Pacific Islander12.7 (69)
Hispanic/Mixed/Other11.0 (60)
Married or partnered68.0 (369)
Lives alone19.5 (106)
Currently employed34.3 (186)
Child care responsibilities (% yes)23.6 (128)
Exercise on a regular basis (% yes)70.7 (413)
Cancer diagnosis
Breast43.6 (237)
Gastrointestinal26.0 (141)
Gynecological21.0 (114)
Lung9.4 (51)
Chemotherapy cycle length
14 days36.1 (196)
21 days54.9 (298)
28 days9.0 (49)
Previous cancer treatments89.9 (450)
Pain present (% yes)72.9 (396)

Abbreviations: gm/dL = grams per deciliter; kg/m = kilograms per meter squared; SD = standard deviation

5.2 Changes in morning fatigue severity

HLM was used to examine how morning fatigue scores changed within the two cycles of CTX, controlling for self-reported and genomic estimates of race and ethnicity. The estimates for the initial piecewise model are presented in Table 4. Since the model was unconditional (i.e., no covariates included in the model), the average morning fatigue severity score at enrollment (i.e., 3.011 on a 0 to 10 scale) represents the intercept. The estimated linear piecewise rates of change were 1.192 and 0.532 (both p<.0001) for piecewise linear 1 and piecewise linear 2, respectively. The estimated quadratic piecewise rates of change were −.599 and −.153 (both p<.0001) for piecewise quadratic 1 and piecewise quadratic 2, respectively. Figure 1A displays the unconditional model for mean morning fatigue scores over the two cycles of CTX.

An external file that holds a picture, illustration, etc.
Object name is nihms845338f1.jpg

A–F – Unconditional piecewise model of mean morning fatigue scores for six assessment points over two cycles of chemotherapy (A). Influence of the recessive model (TT+TC vs. CC) of the rare C allele in IL12B rs3213094 on the inter-individual differences in the intercept for morning fatigue (B). Influence of the dominant model (CC vs. CA+AA) of the rare A allele in TNFA rs1041981 on the slope parameters for morning fatigue (C). Influence of the recessive model (AA+AG vs. GG) of the rare G allele in NOD2 rs2076756 on the inter-individual differences in the intercept for morning fatigue (D). Influence of the recessive model (GG+GC vs. CC) of the rare C allele in NLRP5 rs471979 on the slope parameters for morning fatigue (E). Influence of the dominant model (TT vs. TC+CC) of the rare C allele in NLRP6 rs74044411 on the slope parameters for morning fatigue (F).

Table 4

Hierarchical Linear Models for Morning Fatigue

Cytokine Genes
Morning Fatigue and IL12B rs3213094Coefficient (SE)
Unconditional ModelFinal Model
Fixed effects
Intercept3.011 (.099)+3.008 (.099)+
Ethnicitya
Black versus White−.845 (.917)−.735 (.913)
Asian versus White.214 (.675).265 (.672)
Hispanic versus White.328 (.331).348 (.329)
Ancestry informative markers principal componentsb
PC1−.102 (.104)−.105 (.103)
PC2−.052 (.088)−.031 (.088)
PC3.008 (.039).008 (.039)
PW1 – linear1.192 (.148)+1.191 (.148)+
PW1 – quadratic−.599 (.071)+−.599 (.071)+
PW2 – linear.532(.096)+.531 (.096)+
PW2 – quadratic−.153 (.031)+−.153 (.031)+
Time invariant covariates
Intercept - IL12B rs3213094−.760 (.319)*
Variance components
In intercept3.552+3.512+
Goodness-of-fit deviance (parameters estimated)11026.531695 (13)+11020.881398 (14)
Model comparison χ (df)5.650 (1)*
Morning Fatigue and TNFA rs1041981Coefficient (SE)
Final Model
Fixed effects
Intercept3.011 (.099)+
Ethnicitya
Black versus White−.807 (.917)
Asian versus White.187 (.675)
Hispanic versus White.304 (.331)
Ancestry informative markers principal componentsb
PC1−.106 (.103)
PC2−.045 (.088)
PC3.015 (.039)
PW1 – linear1.186 (.148)+
PW1 – quadratic−.597 (.071)+
PW2 – linear.533 (.096)+
PW2 – quadratic−.153 (.031)+
Time invariant covariates
PW1 – linear - TNFA rs1041981−.641 (.278)*
PW1 – quadratic - TNFA rs1041981.264 (.131)*
Variance components
In intercept3.549+
Goodness-of-fit deviance (parameters estimated)11019.804052 (15)
Model comparison χ (df)6.728 (2)*
Inflammasome Pathway Genes
Morning Fatigue and NLRP5 rs471979Coefficient (SE)
Final Model
Fixed effects
Intercept3.011 (.099)+
Ethnicitya
Black versus White−.852 (.918)
Asian versus White.212 (.676)
Hispanic versus White.310 (.332)
Ancestry informative markers principal componentsb
PC1−.101 (.104)
PC2−.051 (.088)
PC3.004 (.039)
PW1 – linear1.190 (.148)+
PW1 – quadratic−.598 (.071)+
PW2 – linear.533 (.096)+
PW2 – quadratic−.153 (.031)+
Time invariant covariates
PW1 – linear - NLRP5 rs471979−3.292 (1.258)*
PW1 – quadratic - NLRP5 rs4719791.323 (.595)*
Variance components
In intercept3.566+
Goodness-of-fit deviance (parameters estimated)11016.309635 (15)
Model comparison χ (df)10.222 (2)*
Morning Fatigue and NLRP6 rs74044411Coefficient (SE)
Final Model
Fixed effects
Intercept3.010 (.099)+
Ethnicitya
Black versus White−.727 (.914)
Asian versus White.229 (.672)
Hispanic versus White.339 (.330)
Ancestry informative markers principal componentsb
PC1−.128 (.103)
PC2−.068 (.087)
PC3.011 (.039)
PW1 – linear1.186 (.148)+
PW1 – quadratic−.597 (.071)+
PW2 – linear.534 (.096)+
PW2 – quadratic−.153 (.031)+
Time invariant covariates
Intercept
PW1 – linear - NLRP6 rs74044411−1.675 (.785)*
PW1 – quadratic - NLRP6 rs74044411.539 (.373)
Variance components
In intercept3.522+
Goodness-of-fit deviance (parameters estimated)11010.712889 (15)
Model comparison χ (df)15.819 (2)**
Morning Fatigue and NOD2 rs2076756Coefficient (SE)
Final Model
Fixed effects
Intercept3.010 (.099)+
Ethnicitya
Black versus White−.916 (.912)
Asian versus White.280 (.671)
Hispanic versus White.359 (.329)
Ancestry informative markers principal componentsb
PC1−.096 (.103)
PC2−.068 (.087)
PC3.009 (.039)
PW1 – linear1.192 (.148)+
PW1 – quadratic−.599 (.071)+
PW2 – linear.532 (.096)+
PW2 – quadratic−.153 (.031)+
Time invariant covariates
Intercept - NOD2 rs2076756−.960 (.383)*
Variance components
In intercept3.507+
Goodness-of-fit deviance (parameters estimated)11020.293155 (14)
Model comparison χ (df)6.239 (1)*
JAK/STAT Pathway Genes
Morning Fatigue and IL4R Liability ScoreCoefficient (SE)
Final Model
Fixed Effects
Intercept3.010 (.099)+
Ethnicitya
Black versus White−.772 (.917)
Asian versus White.233 (.674)
Hispanic versus White.336 (.331)
Ancestry informative markers principal componentsb
PC1−.103 (.103)
PC2−.061 (.088)
PC3.012 (.039)
PW1 – linear1.191 (.148)+
PW1 – quadratic−.599 (.071)+
PW2 – linear.532 (.096)+
PW2 – quadratic−.153 (.031)+
Time invariant covariates
Intercept
PW1 – linear - IL4R Liability Score−.401(.168)*
PW1 – quadratic - IL4R Liability Score.167 (.079)*
Variance components
In intercept3.548+
Goodness-of-fit deviance (parameters estimated)11019.542001 (15)
Model comparison χ (df)6.990 (2)*
NFkB pathway Genes
Morning Fatigue and IL17RB Liability ScoreCoefficient (SE)
Final Model
Fixed effects
Intercept3.011 (.099)+
Ethnicitya
Black versus White−.843 (.913)
Asian versus White.256 (.672)
Hispanic versus White.324 (.330)
Ancestry informative markers principal componentsb
PC1−.111 (.103)
PC2−.037 (.088)
PC3.013 (.039)
PW1 – linear1.191 (.148)+
PW1 – quadratic−.599 (.071)+
PW2 – linear.531 (.096)+
PW2 – quadratic−.153 (.031)+
Time invariant covariates
Intercept - IL17RB Liability Score−.352 (.162)*
Variance components
In intercept3.519+
Goodness-of-fit deviance (parameters estimated)11021.825332 (14)
Model comparison χ (df)4.706 (1)*
Morning Fatigue and TNFRSF10A rs17620Coefficient (SE)
Final Model
Fixed effects
Intercept3.010 (.099)+
Ethnicitya
Black versus White−.873 (.916)
Asian versus White.235 (.674)
Hispanic versus White.330 (.331)
Ancestry informative markers principal componentsb
PC1−.099 (.103)
PC2−.073 (.088)
PC3.012 (.039)
PW1 – linear1.194 (.148)+
PW1 – quadratic−.601 (.071)+
PW2 – linear.535 (.096)+
PW2 – quadratic−.154 (.031)+
Time invariant covariates
PW1 – linear - TNFRSF10A rs17620.402 (.189)*
PW1 – quadratic - TNFRSF10A rs17620−.193 (.093)*
PW2 – linear - TNFRSF10A rs17620.197 (.127)
PW2 – quadratic - TNFRSF10A rs17620−.051 (.041)
Variance components
In intercept3.544+
Goodness-of-fit deviance (parameters estimated)11018.710558 (17)
Model comparison χ (df)7.821 (4)
Morning Fatigue and TNFRSF10D Liability ScoreCoefficient (SE)
Final Model
Fixed effects
Intercept3.011 (.099)+
Ethnicitya
Black versus White−.810 (.917)
Asian versus White.203 (.675)
Hispanic versus White.323 (.331)
Ancestry informative markers principal componentsb
PC1−.106 (.104)
PC2−.048 (.088)
PC3.008 (.039)
PW1 – linear1.195 (.148)+
PW1 – quadratic−.601 (.071)+
PW2 – linear.529 (.096)+
PW2 – quadratic−.152 (.031)+
Time invariant covariates
PW2 – linear - TNFRSF10D Liability Score−.211 (.094)*
PW2 – quadratic - TNFRSF10D Liability Score.057 (.033)
Variance components
In intercept3.552+
Goodness-of-fit deviance (parameters estimated)11018.923385 (15)
Model comparison χ (df)7.608 (2)*
Morning Fatigue and TNFRSF11A Liability ScoreCoefficient (SE)
Final Model
Fixed effects
Intercept3.011 (.099)+
Ethnicitya
Black versus White−.866 (.914)
Asian versus White.172 (.673)
Hispanic versus White.330 (.330)
Ancestry informative markers principal componentsb
PC1−.107 (.103)
PC2−.046 (.087)
PC3.005 (.039)
PW1 – linear1.193 (.148)+
PW1 – quadratic−.600 (.071)+
PW2 – linear.535 (.096)+
PW2 – quadratic−.154 (.031)+
Time invariant covariates
PW1 – linear - TNFRSF11A Liability Score−.381 (.141)*
PW1 – quadratic - TNFRSF11A Liability Score.174 (.070)*
PW2 – linear - TNFRSF11A Liability Score−.130 (.097)
PW2 – quadratic - TNFRSF11A Liability Score.022 (.031)
Variance components
In intercept3.532+
Goodness-of-fit deviance (parameters estimated)11009.606255 (17)
Model comparison χ (df)16.925 (4)*
Morning Fatigue and TNFRSF14 rs2234163Coefficient (SE)
Final Model
Fixed effects
Intercept3.011 (.099)+
Ethnicitya
Black versus White−.903 (.913)
Asian versus White.112 (.673)
Hispanic versus White.311 (.330)
Ancestry informative markers principal componentsb
PC1−.104 (.103)
PC2−.055 (.087)
PC3.010 (.039)
PW1 – linear1.192 (.148)+
PW1 – quadratic−.599 (.071)+
PW2 – linear.532 (.096)+
PW2 – quadratic−.153 (.031)+
Time invariant covariates
Intercept - TNFRSF14 rs22341631.426 (.638)*
Variance components
In intercept3.514+
Goodness-of-fit deviance (parameters estimated)11021.551571 (14)
Model comparison χ (df)4.980 (1)*
Morning Fatigue and TNFRSF21 Liability ScoreCoefficient (SE)
Final Model
Intercept3.012 (.099)+
Ethnicitya
Black versus White−.815 (.913)
Asian versus White.281 (.672)
Hispanic versus White.310 (.330)
Ancestry informative markers principal componentsb
PC1−.104 (.103)
PC2−.069 (.088)
PC3.001 (.039)
PW1 – linear1.194 (.148)+
PW1 – quadratic−.601 (.071)+
PW2 – linear.531 (.096)+
PW2 – quadratic−.153 (.031)+
Time invariant covariates
Intercept TNFRSF21 Liability Score.416 (.183)*
Variance components
In intercept3.517+
Goodness-of-fit deviance (parameters estimated)11021.369941 (14)
Model comparison χ (df)5.162 (1)*
Self-reported ethnicity – represented by three “dummy” coded variables
Ancestry informative markers - represented by the first three PCs (i.e., PC1, PC2, PC3)
p<.05,
p<.001,
p<.0001

Abbreviations: df = degrees of freedom; IL4R = Interleukin 4 receptor; IL12B= Interleukin 12B; IL17RB = Interleukin 17 receptor B; JAK/STAT = Janus kinase/signal transducers and activators of transcription; NFkB = nuclear factor-kappa beta; NLRC5 = Nod-Like receptor family, caspase recruitment domain containing 5; NLRP5 = Nod-Like receptor family, pyrin domain containing 5; NLRP6 = Nod-Like receptor family, pyrin domain containing 6; NOD2 = Nucleotide-Binding oligomerization domain containing 2; PC=principal component; PW1 = piecewise 1 PW2 = piecewise 2; rs = reference SNP cluster identification number; SE = standard error; TNFA = Tumor necrosis factor alpha; TNFRSF10A = Tumor necrosis factor receptor superfamily member 10A; TNFRSF10D = Tumor necrosis factor receptor superfamily member 10D; TNFRSF11A = Tumor necrosis factor receptor superfamily member 11A; TNFRSF14 = Tumor necrosis factor receptor superfamily member 14; TNFRSF21 = Tumor necrosis factor receptor superfamily member 21

5.3 Genomic predictors of inter-individual differences in morning fatigue

Table 4 shows the final HLM models for morning fatigue. For the cytokine genes, two SNPs were associated with inter-individual differences in morning fatigue. Figure 1B illustrates the adjusted initial level of morning fatigue (i.e., intercept) based on a recessive model for IL12B rs3213094 (i.e., TT+TC vs. CC). Figure 1C illustrates the predicted changes in the trajectory of morning fatigue (i.e., slope) based on a dominant model for TNFA rs1041981 (i.e., CC vs. CA+AA).

For the inflammasome pathway genes, three SNPs were associated with changes in morning fatigue. Figure 1D illustrates the adjusted initial levels of morning fatigue based on the recessive model for nucleotide-binding oligomerization domain containing 2 (NOD2) rs2076756 (i.e., AA+AG vs. GG). Figures 1E and 1F respectively, illustrate the predicted changes in the trajectories of morning fatigue based on the recessive model for non-like receptor family, pyrin containing domain 5 (NLRP5) rs471979 (i.e., GG+GC vs. CC) and a dominant model for NLRP6 rs74044411 (i.e., TT vs. TC+CC).

For the JAK/STAT pathway, the liability score for all IL4R, was associated with inter-individual differences in the slope of morning fatigue (Figure 2A).

An external file that holds a picture, illustration, etc.
Object name is nihms845338f2.jpg

A–D – Influence of the liability score for the sum of the occurrence of the rare alleles in IL4R on the slope parameters for morning fatigue (A). Influence of the dominant model (AA vs. AG+GG) of the rare G allele for TNFRSF14 rs2234163 on the inter-individual differences in the intercept for morning fatigue (B). Influence of the liability score for the sum of the occurrence of the rare alleles in IL17RB, on the inter-individual differences in the intercept for morning fatigue (C). Influence of the liability score for the sum of the occurrence of the rare alleles in TNFRSF21 on the inter-individual differences in the intercept for morning fatigue (D).

For the NFkB pathway genes, two SNPs and four liability scores were associated with inter-individual differences in morning fatigue. Figure 2B illustrates the adjusted initial level of morning fatigue based on the dominant model for tumor necrosis factor receptor super family, member 14 (TNFRSF14) rs2234163 (i.e., AA vs. AG+GG). Figures 2C and 2D respectively, illustrate the adjusted initial levels of morning fatigue based on the liability scores for IL17RB and TNFRSF21. Figures 3A, 3B, and 3C, respectively, illustrate the predicted changes in the trajectories of morning fatigue for the additive model for TNFRSF10A rs17620 (i.e., TT vs. TC vs. CC) and the liability scores for TNFRSF10D and TNFRSF11A.

An external file that holds a picture, illustration, etc.
Object name is nihms845338f3.jpg

A–C – Influence of the additive model (TT vs. TC vs. CC) of the rare C allele for TNFRSF10A rs17620 on the slope parameters for morning fatigue (A). Influence of the liability score for the sum of the occurrence of the rare alleles in TNFRSF10D on the slope parameters for morning fatigue (B). Influence of the liability score for the sum of the occurrence of the rare alleles in TNFRSF11A on the slope parameters for morning fatigue (C).

5.4 Changes in evening fatigue severity

HLM was used to examine how evening fatigue scores changed within the two cycles of CTX, controlling for self-reported and genomic estimates of race and ethnicity. The estimates for the initial piecewise model are presented in Table 5. Since the model was unconditional (i.e., no covariates), the intercept represents the average evening fatigue severity score at enrollment (i.e., 5.310 on a 0 to 10 scale). The estimated linear piecewise rates of change were 0.601 and 0.394 (both, p<.0001) for piecewise linear 1 and piecewise linear 2, respectively. The estimated quadratic piecewise rates of change were −.306 and −.113 (both, p<.0001) for piecewise quadratic 1 and piecewise quadratic 2, respectively. Figure 4A displays the unconditional model for mean evening fatigue scores over two cycles of CTX.

An external file that holds a picture, illustration, etc.
Object name is nihms845338f4.jpg

A–D – Unconditional piecewise model of mean evening fatigue scores for six assessment points over two cycles of chemotherapy (A). Influence of the recessive model (TT+TC vs. CC) of the rare C allele in IL12B rs3213094 on the inter-individual differences in the intercept for evening fatigue (B). Influence of the liability score for the sum of the occurrence of the rare alleles in IL12B on the inter-individual differences in the intercept for evening fatigue (C). Influence of the dominant model (CC vs. CA+AA) of the rare A allele in TNFA rs1041981 on the slope parameters for evening fatigue (D).

Table 5

Hierarchical Linear Models for Evening Fatigue

Cytokine Genes
Evening Fatigue and IL12B rs3213094Coefficient (SE)
Unconditional ModelFinal Model
Fixed effects
Intercept5.310 (.090)+5.307 (.090)+
Ethnicitya
Black versus White−1.584 (.824)−1.501 (.822)
Asian versus White−.269 (.608)−.231 (.606)
Hispanic versus White−.348 (.298)−.332 (.297)
Ancestry informative markers principal components
PC1−.070 (.093)−.072 (.093)
PC2−.044 (.079)−.029 (.079)
PC3−.009 (.035)−.008 (.035)
PW 1 – linear.601 (.137)+.601 (.137)+
PW 1 – quadratic−.306 (.066)+−.306 (.066)+
PW 2 – linear.394 (.088)+.394 (.088)+
PW 2 – quadratic−.113 (.028)+−.113 (.028)+
Time invariant covariates
Intercept IL12B rs3213094−.573 (.288)*
Variance components
In intercept2.867+2.843+
Goodness-of-fit deviance (parameters estimated)10399.632349 (13)+10395.680232 (14)
Model comparison χ (df)3.952 (1)*
Evening Fatigue and IL12B Liability ScoreCoefficient (SE)
Final Model
Fixed effects
Intercept5.308 (.090)+
Ethnicitya
Black versus White−1.579 (.820)
Asian versus White−.322 (.605)
Hispanic versus White−.347 (.297)
Ancestry informative markers principal componentsb
PC1−.086 (.093)
PC2−.021 (.079)
PC3−.003 (.035)
PW 1 – linear.600 (.137) +
PW 1 – quadratic−.306 (.066) +
PW 2 – linear.393 (.088) +
PW 2 – quadratic−.112 (.028) +
Time invariant covariates
Intercept - IL12B Liability Score−.258 (.115)*
Variance components
In intercept2.838+
Goodness-of-fit deviance (parameters estimated)10394.643652 (14)
Model comparison χ (df)4.989 (1)*
Evening Fatigue and TNFA rs1041981Coefficient (SE)
Final Model
Fixed effects
Intercept5.309 (.090)+
Ethnicitya
Black versus White−1.548 (.822)
Asian versus White−.293 (.606)
Hispanic versus White−.369 (.298)
Ancestry informative markers principal componentsb
PC1−.073 (.093)
PC2−.038 (.079)
PC3−.002 (.035)
PW 1 – linear.596 (.137)+
PW 1 – quadratic−.304 (.066)+
PW 2 – linear.395 (.088)+
PW 2 – quadratic−.113 (.028)+
Time invariant covariates
Intercept
PW 1 – linear - TNFA rs1041981−.602 (.256)*
PW 1 – quadratic - TNFA rs1041981.252 (.121)*
Variance components
In intercept2.852+
Goodness-of-fit deviance (parameters estimated)10392.936781 (15)
Model comparison χ (df)6.696 (2)*
Inflammasome Pathway Genes
Evening Fatigue and CARD6 rs10512747Coefficient (SE)
Final Model
Fixed effects
Intercept5.309 (.089)+
Ethnicitya
Black versus White−1.591 (.819)
Asian versus White−.350 (.605)
Hispanic versus White−.397 (.297)
Ancestry informative markers principal componentsb
PC1−.057 (.093)
PC2−.043 (.078)
PC3−.008 (.035)
PW 1 – linear.599 (.137)+
PW 1 – quadratic−.306 (.066)+
PW 2 – linear.394 (.088)+
PW 2 – quadratic−.113 (.028)+
Time invariant covariates
Intercept - CARD6 rs10512747−.441 (.176)*
Variance components
In intercept2.829+
Goodness-of-fit deviance (parameters estimated)10393.393966 (14)
Model comparison χ (df)6.238 (1)*
Evening Fatigue and NLRP4 rs17857373Coefficient (SE)
Final Model
Fixed effects
Intercept5.308 (.090)+
Ethnicitya
Black versus White−1.600 (.821)
Asian versus White−.298 (.605)
Hispanic versus White−.365 (.297)
Ancestry informative markers principal componentsb
PC1−.061 (.093)
PC2−.046 (.079)
PC3−.003 (.035)
PW 1 – linear.601 (.137)+
PW 1 – quadratic−.306 (.066)+
PW 2 – linear.394 (.088)+
PW 2 – quadratic−.113 (.028)+
Time invariant covariates
Intercept - NLRP4 rs17857373−.603 (.291)*
Variance components
In intercept2.841+
Goodness-of-fit deviance (parameters estimated)10395.340962 (14)
Model comparison χ (df)4.291 (1)*
Evening Fatigue and NLRP6 rs74044411Coefficient (SE)
Final Model
Fixed effects
Intercept5.309 (.090)+
Ethnicitya
Black versus White−1.491 (.822)
Asian versus White−.257 (.606)
Hispanic versus White−.339 (.297)
Ancestry informative markers principal componentsb
PC1−.093 (.093)
PC2−.060 (.079)
PC3−.007 (.035)
PW 1 – linear.598 (.136)+
PW 1 – quadratic−.306 (.066)+
PW 2 – linear.395 (.088)+
PW 2 – quadratic−.113 (.028)+
Time invariant covariates
Intercept
PW 1 – linear - NLRP6 rs74044411−1.417 (.682)*
PW 1 – quadratic - NLRP6 rs74044411.449 (.322)
Variance components
In intercept2.847+
Goodness-of-fit deviance (parameters estimated)10385.256709 (15)
Model comparison χ (df)14.376 (2)**
Evening Fatigue and NOD2 rs2076756Coefficient (SE)
Final Model
Fixed effects
Intercept5.308 (.089)+
Ethnicitya
Black versus White−1.684 (.813)*
Asian versus White−.177 (.599)
Hispanic versus White−.304 (.294)
Ancestry informative markers principal componentsb
PC1−.061 (.092)
PC2−.067 (.078)
PC3−.008 (.035)
PW 1 – linear.601 (.137)+
PW 1 – quadratic−.306 (.066)+
PW 2 – linear.394 (.088)+
PW 2 – quadratic−.113 (.028)+
Time invariant covariates
Intercept - NOD2 rs2076756−1.350 (.342)+
Variance components
In intercept2.777+
Goodness-of-fit deviance (parameters estimated)10384.268252 (14)
Model comparison χ (df)15.364 (1)+
MAP/JNK Pathway Genes
Evening Fatigue and IL17RD rs61742267Coefficient (SE)
Final Model
Fixed effects
Intercept5.310 (.090)+
Ethnicitya
Black versus White−1.539 (.821)
Asian versus White−.265 (.605)
Hispanic versus White−.319 (.297)
Ancestry informative markers principal componentsb
PC1−.059 (.093)
PC2−.044 (.079)
PC3−.013 (.035)
PW 1 – linear.601 (.137)+
PW 1 – quadratic−.306 (.066)+
PW 2 – linear.393 (.088)+
PW 2 – quadratic−.112 (.028)+
Time invariant covariates
Intercept - IL17RD rs61742267−.933 (.429)*
Variance components
In intercept2.840+
Goodness-of-fit deviance (parameters estimated)10394.915944 (14)
Model comparison χ (df)4.716 (1)*
NfKB Pathway Genes
Evening Fatigue and IL17RB rs2232346 and
IL17RB rs1043261
Coefficient (SE)
Final Model
Fixed effects
Intercept5.308 (.089)+
Ethnicitya
Black versus White−1.573 (.817)
Asian versus White−.179 (.603)
Hispanic versus White−.344 (.296)
Ancestry informative markers principal componentsb
PC1−.065 (.092)
PC2−.037 (.078)
PC3−.009 (.035)
PW 1 – linear.600 (.137) +
PW 1 – quadratic−.306 (.066) +
PW 2 – linear.394 (.088) +
PW 2 – quadratic−.113 (.028) +
Time invariant covariates
Intercept - IL17RB rs2232346−.791 (.388)*
Intercept - IL17RB rs1043261−1.456 (.584)*
Variance components
In intercept2.809+
Goodness-of-fit deviance (parameters estimated)10389.389046 (15)
Model comparison χ (df)10.243 (2)*
Evening Fatigue and LTBR Liability ScoreCoefficient (SE)
Final Model
Fixed effects
Intercept5.309 (.090)+
Ethnicitya
Black versus White−1.562 (.826)
Asian versus White−.277 (.609)
Hispanic versus White−.346 (.299)
Ancestry informative markers principal componentsb
PC1−.072 (.093)
PC2−.044 (.079)
PC3−.010 (.035)
PW 1 – linear.604 (.136)+
PW 1 – quadratic−.308 (.066)+
PW 2 – linear.396 (.088)+
PW 2 – quadratic−.113 (.028)+
Time invariant covariates
Intercept
PW 1 – linear - LTBR Liability Score−3.188 (1.080)*
PW 1 – quadratic - LTBR Liability Score1.540 (.513)*
Variance components
In intercept2.885+
Goodness-of-fit deviance (parameters estimated)10390.660476 (15)
Model comparison χ (df)8.972 (2)*
Evening Fatigue and TNFRSF14 rs2234163Coefficient (SE)
Final Model
Fixed effects
Intercept5.309 (.090)+
Ethnicitya
Black versus White−1.648 (.818)
Asian versus White−.383 (.605)
Hispanic versus White−.366 (.296)
Ancestry informative markers principal componentsb
PC1−.072 (.092)
PC2−.048 (.078)
PC3−.007 (.035)
PW 1 – linear.600 (.137)+
PW 1 – quadratic−.306 (.066)+
PW 2 – linear.394 (.088)+
PW 2 – quadratic−.113 (.028)+
Time invariant covariates
Intercept - TNFRSF14 rs22341631.590 (.572)*
Variance components
In intercept2.821+
Goodness-of-fit deviance (parameters estimated)10391.966279 (14)
Model comparison χ (df)7.666 (1)*
Self-reported ethnicity – represented by three “dummy” coded variables
Ancestry informative markers - represented by the first three PCs (i.e., PC1, PC2, PC3)
p<.05,
p<.001,
p<.0001

Abbreviations: CARD6 = Caspase recruitment domain family member 6; df = degrees of freedom; IL12B= Interleukin 12B; IL17RB = Interleukin 17 receptor B; IL17RD = Interleukin 17 receptor D; JAK = jaunts kinase transducers and activators of transcription; LTBR = Lymphotoxin beta receptor; MAP = mitogen-activated protein kinase; NFkB = nuclear factor-kappa beta; NLRP4 = Nod-Like receptor family, pyrin domain containing 4; NLRP6 = Nod-Like receptor family, pyrin domain containing 6; NOD2 = Nucleotide-Binding oligomerization domain containing 2; PC=principal component; PW1 = piecewise 1; PW2 = piecewise 2; rs = reference SNP cluster identification number; SE = standard error; TNFA = Tumor necrosis factor alpha; TNFRSF14 = Tumor necrosis factor receptor superfamily member 14.

5.5 Genomic predictors of inter-individual differences in evening fatigue

Table 5 shows the final HLM models for evening fatigue. For the cytokine genes, two SNPs and one liability score were associated with inter-individual differences in evening fatigue. Figures 4B and 4C respectively, illustrate the adjusted initial level of evening fatigue based on the recessive model for IL12B rs3213094 (i.e., TT+TC vs. CC) and the liability score for IL12B. Figure 4D illustrates the predicted changes in the trajectories of evening fatigue for a dominant model for TNFA rs1041981 (i.e., CC vs. CA+AA).

For the inflammasome pathway genes, four SNPs were associated with evening fatigue. Figures 5A, 5B, and 5C respectively, illustrate the adjusted initial levels of evening fatigue based on the additive model for capsase recruitment domain family member 6 (CARD6) rs10512747 (i.e., TT vs. TC vs. CC); the dominant model for NLRP4 rs17857373 (i.e., CC vs. CG+GG) and the recessive model for NOD2 rs2076756 (i.e., AA+AG vs. GG). Figure 5D illustrates the predicted changes in the trajectory of evening fatigue based on the dominant model for NLRP6 rs74044411 (i.e., TT vs. TC+CC).

An external file that holds a picture, illustration, etc.
Object name is nihms845338f5.jpg

A–E – Influence of the additive model (TT vs. TC vs. CC) of the rare C allele in CARD6 rs10512747 on the inter-individual differences in the intercept for evening fatigue (A). Influence of the dominant model (CC vs. CG+GG) of the rare G allele for NLRP4 rs17857373 on the inter-individual differences in the intercept for evening fatigue (B). Influence of the recessive model (AA+AG vs. GG) of the rare G allele for NOD2 rs2076756 on the inter-individual differences in the intercept for evening fatigue (C). Influence of the dominant model (TT vs. TC+CC) of the rare C allele for NLRP6 rs74044411 on the slope parameters for evening fatigue (D). Influence of the dominant model (AA vs. AG+GG) for the rare G allele for IL17RD rs61742267 on the inter-individual differences in the intercept for evening fatigue (E).

For the MAP/JNK pathway genes, Figure 5E illustrates the adjusted initial level of evening fatigue based on the dominant model for IL17RD rs61742267 (i.e., AA vs. AG+GG).

For the NFkB pathway genes, three SNPs and one liability score were associated with inter-individual differences in evening fatigue. Figure 6A illustrates the adjusted initial level of evening fatigue based on the dominant model for IL17RB rs2232346 (i.e., TT vs. TC+CC) and the recessive model for IL17RB rs1043261 (i.e., TT+TC vs. CC). Figures 6B and 6C respectively, illustrate the adjusted initial level of evening fatigue based on the dominate model for TNFRSF14 rs2234163 (i.e., AA vs. AG+GG) and the predicted change in the trajectory of evening fatigue based on the liability score for lymphotoxin beta receptor (LTBR).

An external file that holds a picture, illustration, etc.
Object name is nihms845338f6.jpg

A–C – Influence of the dominant model (TT vs. TC+CC) of the rare C allele for IL17RB rs2232346 and the recessive model (TT+TC vs. CC) of the rare C allele for IL17RB rs1043261 on the inter-individual differences in the intercept for evening fatigue (A). Influence of the dominant model (AA vs. AG+GG) of the rare G allele for TNFRSF14 rs2234163 on the inter-individual differences in the intercept for evening fatigue (B). Influence of the liability score for the sum of the occurrence of the rare alleles in LTBR on the slope parameters for evening fatigue (C).

6. Discussion

In our prior studies [9, 10], common and unique phenotypic predictors of morning and evening fatigue were identified that provided evidence that they are distinct but related symptoms. As summarized in Table 6, this study extends these findings by identifying common as well as unique genetic associations for morning and evening fatigue. Controlling for self-reported and genomic estimates of race and ethnicity, five SNPs were associated with inter-individual variability in both morning and evening fatigue. Three SNPs (i.e., NOD2 rs2076756, TNFRSF14 rs2234163, IL12B rs3213094) were associated with changes in the initial levels and two SNPs (i.e., NLRP6 rs74044411, TNFA rs1041981) were associated with the trajectories of morning and evening fatigue severity. Two unique polymorphisms (i.e., NLRP5 rs471979, TNFRSF10A rs17620) and five liability scores (i.e., IL4R, IL17RB, TNFRSF10D, TNFRSF11A, TNFRSF21) were associated with only morning fatigue. Five unique polymorphisms on four genes (i.e., CARD6 rs10512747, IL17RB rs2232346, IL17RB rs1043261, IL17RD rs61742267, NLRP4 rs17857373) and two liability scores (i.e., IL12B, LTBR) were associated with only evening fatigue.

Table 6

Comparison of Genes Associated with Interindividual Differences in Morning and Evening Fatigue in Patients Receiving Chemotherapy

PathwayMorning FatigueEvening Fatigue
Cytokine genesTNFA rs1041981TNFA rs1041981
IL12B rs3213094IL12B rs3213094
IL12B liability score
Inflammasome pathway genesNLRP6 rs74044411NLRP6 rs74044411
NOD2 rs2076756NOD2 rs2076756
NLRP5 rs471979CARD6 rs10512747
NLRP4 rs1785737
JAK/STAT pathway genesIL4R liability scoreNone
MAP/JNK pathway genesNoneIL17RD rs61742267
NfKB pathway genesTNFRSF14 rs2234163TNFRSF14 rs2234163
IL17RB liability scoreIL17RB rs2232346
TNFRSF10A rs17620IL17RB rs1043261
TNFRSF10D liability scoreLTBR liability score
TNFRSF11A liability score
TNFRSF21 liability score

Abbreviations: CARD6 = caspase recruitment domain family member 6; IL4R = interleukin 4 receptor; IL12B = interleukin 12B; IL17RB = interleukin 17 receptor B; IL17RD = interleukin 17 receptor D: JAK/STAT = Janus kinase/signal transducers and activators of transcription; LTBR = lymphotoxin beta receptor; MAP = mitogen-activated protein kinase; NFkB = nuclear factor-kappa beta; NLRC5 = nod-like receptor family, caspase recruitment domain containing 5; NLRP4 = nod-like receptor family, pyrin domain containing 4; NLRP5 = nod-like receptor family, pyrin domain containing 5; NLRP6 = nod-like receptor family, pyrin domain containing 6; NOD2 = nucleotide-binding oligomerization domain containing 2; rs = reference SNP cluster identification number; TNFA = tumor necrosis factor alpha; TNFRSF10A = tumor necrosis factor receptor superfamily member 10A; TNFRSF10D = tumor necrosis factor receptor superfamily member 10D; TNFRSF11A = Tumor necrosis factor receptor superfamily member 11A; TNFRSF14 = tumor necrosis factor receptor superfamily member 14; TNFRSF21 = tumor necrosis factor receptor superfamily member 21

In terms of the genes themselves, as summarized in Table 6, polymorphisms in six genes (i.e., TNFA, IL12B, NLRP6, NOD2, TNFRSF14, IL17RB) were associated with both morning and evening fatigue. Polymorphisms in six genes (i.e., NLRP5, IL4R, TNFRSF10A, TNFRSF10D, TNFRSF11A, TNFRSF21) were associated with only morning fatigue. Polymorphisms in three genes (i.e., NLRP6, NLRP4, LTBR) were associated with only evening fatigue. Taken together, these findings add to the growing body of evidence that suggests that morning and evening fatigue are distinct but related symptoms.

One of the goals of this study was to evaluate the effects of polymorphisms in genes involved in inflammation on initial levels as well as on the trajectories of morning and evening fatigue. As noted in the introduction, while most studies evaluated for associations between cytokine dysregulation and fatigue, additional pathways are involved in the regulation of inflammatory processes. Therefore, the findings from this study are discussed in the context of each of the inflammatory pathways investigated in our study.

6.1 Cytokine genes

As key regulators of inflammation and immune responses, polymorphisms in cytokine genes were the most common variations evaluated as potential mechanisms for fatigue [14, 38, 55, 8590]. While previous studies found associations between polymorphisms in a number of cytokine genes and fatigue (for reviews see [29, 91]), in our study, the same polymorphisms in TNFA and IL12B were associated with decreases in both morning and evening fatigue severity.

TNFA encodes for a pleiotropic cytokine that regulates immune responses, as well as cell proliferation and differentiation. For TNFA rs1041981, individuals who carried one or two doses of the rare A allele reported slightly lower morning and evening fatigue severity scores (Figures 1C and and4D).4D). TNFA rs1041981 is located on chromosome 6p21.3. At this locus, the lymphotoxin-alpha (LTA) and TNFA genes are closely juxtaposed with the LTA coding region part of the TNFA early promoter region. While previous findings suggested that LTA shared a common signaling pathway with TNFA to mediate inflammatory responses [92], recent evidence suggests that the LTA and TNFA signaling pathways are unique [93, 94]. In the literature [95], rs1041981 is described as a SNP in both the LTA (i.e., coding region) and TNFA (i.e., promoter region) genes. When cited as a SNP in the LTA gene, rs1041981 was associated with inflammatory processes involved in atherogenesis [96], periodontal disease [97], and glucose intolerance [98]. When cited as a SNP in the promoter region of TNFA, rs1041981 was associated with sleep disturbance in patients with HIV [99]. These results suggest that as a promotor variant, this SNP may influence inflammatory responses through both TNFA and LTA signaling pathways.

In contrast to our findings, in the only study that evaluated the association between TNFA rs1041981 and morning and evening fatigue severity [100], patients with HIV disease who carried one or two doses of the rare A allele reported higher fatigue scores. These inconsistent findings may be related to differences in the way that the fatigue phenotypes were created. In the HIV study, mean scores were calculated. In our study, the effect of the polymorphism on initial levels as well as on the trajectories of morning and evening fatigue were evaluated. In addition, immune function in patients with HIV may have different signaling mechanisms than in oncology patients.

While, SIFT [83] and PolyPhen-2 [84] predicted a subtle impact of TNFA rs1041981 on function (i.e., tolerated by SIFT and benign by Polyphen-2), its impact on TNFA transcription is potentially more pronounced. The occurrence of the rare A allele changes the profile of transcription factors that recognize this sequence. With the homozygous common allele genotype (CC), six transcription factors are predicted to recognize the C-containing sequence (i.e., binding sites) (i.e., c_Ets-1 68, C/EBPalpha, C/EBPbeta, FOXP3, HNF-4alpha2, HNF-4alpha1) compared to only three transcription factors when the rare A allele is present (i.e., RXR-alpha, ETF, c-Ets-1 68). Reduced transcription factor activity was linked to reduced inflammatory signaling and decreased levels of fatigue in breast cancer survivors [101] and may be a potential explanation for the decreases in morning and evening fatigue severity reported by patients with the CA or AA genotype.

IL12B encodes for a proinflamatory cytokine that initiates the release of interferon gamma. The IL12B rs3213094 polymorphism was associated with lower initial levels of both morning and evening fatigue. No predictions for the functional effects of IL12B rs3213094 were identified using SIFT or PolyPhen-2. While no studies were found on an association between this SNP and fatigue severity, previous research found an association with the development of psoriasis [102].

A higher liability score for IL12B was associated with lower initial levels of evening fatigue. When the six SNPs found on IL12B were evaluated, four were predicted to be tolerated or benign damaging by SIFT and PolyPhen-2 respectively, and two were not identified in these bioinformational tools. As shown in Figure 4C, for patients with zero occurrences of the rare alleles, evening fatigue scores were above the clinically meaningful cutoff score for four of the six assessments (i.e., assessments 2, 4, 5, and 6). In contrast, patients with only three doses of the rare allele had evening fatigue scores that were below the clinically meaningful cutoff score for all six assessments. While no studies were found on associations between polymorphisms in IL12B and fatigue, one potential explanation for the predicted lower levels of evening fatigue could be decreased release of interferon gamma. Lower levels of interferon gamma were associated with lower levels of fatigue in patients with Sjögren’s syndrome [103].

It is important to note that cytokine dysregulation is associated with the co-occurrence of sleep disturbance, depressive symptoms, anxiety, and fatigue [56, 104]. Possible links among these co-occurring symptoms and polymorphisms in TNFA and IL12B require further study.

6.2 Inflammasome Pathway

Inflammasomes are multimeric protein complexes that serve as platforms for activation of the innate immune response through mediation of the NFkB and interferon signaling pathways [105, 106]. The formation of inflammasome complexes is activated by pattern recognition receptors (PRR) that detect danger-and pathogen-associated molecular patterns (i.e., DAMPs and PAMPs, respectively) [106]. These inflammasome complexes initiate a signaling pathway that activates capase-1 and transcription factor NFkB, which drives the expression of IL1β and IL18 [107]. To date, the most studied inflammasome complexes are activated by the Nod-Like Receptors (NLRs) family of PRRs.

In our study, five SNPs on five different NLR genes involved in inflammasome activation were associated with multiple effects on morning and evening fatigue. The same two SNPs, NOD2 rs2076756 and NLRP6 rs74044411 were associated with lower initial levels as well as with the trajectories of both morning and evening fatigue, respectively. One of the other SNPs (i.e., NLRP5 rs471979) was associated with the trajectory of morning fatigue. The other two SNPs (i.e., CARD6 rs10512747, NLRP4 rs17857373) were associated with initial levels of evening fatigue.

NOD2 expression results in activation of the MAPK and NFkB inflammatory pathways and a reduction in toll-like receptor mediated inflammatory responses [105]. These effects appear to be dependent on the degree of NOD2 expression [108]. While predictions of the functional effect of NOD2 rs2076756 were not identified by SIFT or Poly-Phen-2, rs2076756 is strongly associated with increased cancer risk [109], Crohn’s disease [110], and NOD2-associated autoinflammatory disease [111]. In addition, patients who are heterozygous (AG) or homozygous (GG) for the rare G allele are diagnosed with inflammatory bowel disease at a younger age [112]. In our study, patients who were homozygous (GG) for the rare G allele in NOD2 rs2076756 were predicted to have morning and evening fatigue below clinically meaningful levels across all six assessments (Figures 1D and and5C,5C, respectively). Explanations for this apparent protective effect of NOD2 on fatigue severity in oncology patients are not readily apparent. Given that the effects of NOD2 appear to be dependent on the degree of gene expression [108], future studies need to examine the effects of this specific polymorphism on differential gene expression in oncology patients with clinically meaningful differences in the severity of both morning and evening fatigue.

Recent evidence suggests that NLRP6 inhibits inflammasome and non-inflammasome dependent inflammatory responses [113, 114]. NLRP6 is associated with maintaining mucosal integrity of the gut and bacterial symbiosis [115]. NLRP6 rs74044411 is a missense mutation that substitutes alanine for valine. The SNP is predicted to be tolerated by SIFT and benign by PolyPhen-2. Patients who were heterozygous or homozygous for the rare C allele reported levels of morning (Figure 1F) and evening (Figure 5D) fatigue that were below the clinically meaningful cutoff levels (i.e., 3.2 and 5.6 respectively) at five of the six assessments. Given recent evidence that dysbiosis of the gut membrane was associated with increased fatigue in oncology patients receiving pelvic radiation [116] and our findings, future studies need to evaluate the functional effects of this SNP as well as the inter-relationships between alterations in the gut microbiome and fatigue severity.

Only one polymorphism in a gene from the inflammasome pathway (i.e., NLRP5 rs471979) predicted changes in the trajectory of morning fatigue. NLRP5 encodes for a protein that plays a role for zygotes to progress beyond the first embryonic cell divisions and is essential for RNA stability. Functional effects of this SNP were not identified by SIFT or PolyPhen-2. In addition, no studies were founding linking it to fatigue. Variations in NLRP5 were found to be associated with congenital disorders of growth and development [117], while higher NLRP5 expression was associated with periodontal disease in older adults [118]. In our study, the morning fatigue scores of patients who were homozygous for the C allele of NLRP5 rs471979 were predicted to be below the clinically meaningful cutoff score for five of the six assessments (Figure 1E).

Two SNPs from the inflammasome pathway (i.e., NLRP4 rs17857373, CARD6 rs10512747) were associated only with evening fatigue. NLRP4 rs17857373, predicted lower initial levels of evening fatigue. NLRP4 expression regulates type-1 interferon signaling and NF-kB activity induced by intracellular adapter proteins and kinases that functionally connect TNF and IL-1R receptors to NF-kB responses [105]. NLRP4 rs17857373 is a missense mutation that substitutes aspartic acid for glutamic acid. This change is predicted to be tolerated by SIFT and benign by PolyPhen-2. While no studies were found that described the effect of NLRP4 rs17857373, NLRP4 overexpression was associated with a diagnosis of bladder cancer [119], with an increased potential for metastasis and CTX resistance in women with epithelial ovarian cancer [120], and with periodontal disease [118]. In our study, patients who were homozygous or heterozygous for the rare G allele were predicted to have evening fatigue scores below the clinically meaningful cutoff at all six assessments (Figure 5B).

CARD6 encodes for a protein that regulates the adaptive and innate immune responses through modulation of interferon and NFkB signaling [121]. CARD6 appears to play a role in the development of GI cancers [122] and may protect cardiac muscle from hypertrophy [123]. CARD6 rs10512747 is a missense mutation that substitutes leucine for serine. This change is predicted to be tolerated by SIFT and potentially damaging by PolyPhen-2. While no studies were found that described the effect of this polymorphism, in our study, each additional dose of the rare C allele was associated with lower predicted evening fatigue scores at enrollment (i.e., TT = 5.40, TC = 4.96, CC = 4.52) (Figure 5A).

The function of the inflammasomes is an evolving area of investigation. In our study, all of the SNPs across the five genes in this pathway were associated with decreases in fatigue severity. Additional research is warranted to determine if the functional effects of these polymorphisms prevent the initiation of the inflammasome complex which may decrease inflammatory responses. While no studies were found that discussed the role of inflammasomes in the development of fatigue, it is reasonable to hypothesize that dysregulation of inflammasomes could result in decreases in IL-1B activity and decreases in innate inflammatory responses, and subsequent decreases in fatigue severity [124]. Additional research is warranted on this potential new mechanism for fatigue in oncology patients.

6.3 JAK/STAT pathway

The pleiotropic JAK/STAT pathway signals a cascade of reactions to maintain homeostasis [125]. The liability score for SNPs in IL4R predicted changes in the trajectory of morning fatigue. IL4R encodes for a cellular receptor for IL-4 and IL-13 that coordinates inflammatory responses through cytokine receptor signal transduction [126]. Of the seven individual SNPs included in the liability score, four were predicted to be tolerated or possibly damaging by SIFT and PolyPhen-2, respectively and three were not predicted by either bioinformational tool. In our study, for those patients who carried three or more rare alleles, their morning fatigue levels were predicted to be below the clinically meaningful cutoff score for five of the six assessments (Figure 2A). While no studies reported on an association between IL4R polymorphisms and fatigue severity, increased expression of IL4R was correlated with higher levels of fatigue in a sample of breast cancer survivors [50]. One potential explanation for the decreases in fatigue severity found in our study is that an increased number of polymorphisms in this gene changes the affinity of the cellular receptor which decreases the inflammatory responses associated with IL4 and IL13 signal transduction.

6.4 MAPK/JNK pathway

The MAPK/JNK pathway is both an upstream and downstream regulator of the expression of pro-inflammatory cytokines [127]. Only one SNP (i.e., IL17RD rs61742267) from the MAPK/JNK pathway was associated with evening fatigue. IL17RD (also called Sef [similar expression to fibroblast growth factor]) was found to be a tumor suppressor gene [128] and an inhibitor of toll-like receptor signaling [129]. IL17RD rs61742267 is a missense mutation that substitutes serine for proline. While no effect was predicted by PolyPhen-2, it was predicted by SIFT to have a potentially damaging effect on gene function. No studies were found that discussed the role of IL17RD in the development of fatigue. In our study, patients who are homozygous or heterozygous for the rare G allele were predicted to have evening fatigue at levels below the clinically meaningful cutoff at all six assessments (Figure 5E). One possible explanation for this finding could be that this polymorphism modulates toll-like receptor signaling which may decrease inflammatory responses that results in decreases in fatigue severity.

6.5 NFkB pathway

The NFkB pathway mediates cellular functions including apoptosis, cellular proliferation, and inflammatory responses through a complex signaling network [130]. This NFkB signaling network is regulated by canonical and non-canonical pathways. The canonical pathway is triggered by a variety of signals (e.g., members of the TNFRS family, IL1-R, toll-like receptors). The non-canonical NFkB pathway is triggered by specific members of the inhibitor of kappa B and TNFRSF families and LTBR [131]. Together, the canonical and non-canonical pathways regulate pro-and anti-inflammatory responses to prevent disorders that are associated with dysregulation of the NFkB pathway (e.g., rheumatoid arthritis, multiple sclerosis, inflammatory bowel disease) [132].

For the NFkB pathway, four SNPs and five liability scores across seven genes were associated with inter-individual differences in morning and evening fatigue. Only one SNP (i.e., TNFRSF14 rs2234163) was associated with initial levels of both morning and evening fatigue. TNFRSF14, encodes for a protein that both activates and inhibits T-cells based on the cellular environment [133]. TNFRSF14 rs2234163 is a missense mutation that substitutes threonine for alanine. It was predicted to be tolerated or possibly damaging by SIFT and PolyPhen-2, respectively. This SNP was associated with p53 mutations in Chinese women with GYN cancers [134]. In our study, compared to patients who were homozygous for the common allele (i.e., morning LFS score = 2.98, evening LFS score = 5.28), patients who were heterozygous or homozygous for the rare G allele of TNFRSF14 rs2234163, were predicted to have higher morning (i.e., LFS = 4.41) and evening (i.e., LFS = 6.87) scores (Figures 2B and and6B).6B). When these differences in fatigue scores were compared, caring one or two copies of the rare G allele was associated with clinically meaningful increases in both morning (d=.97) and evening (d=.75) fatigue severity. One possible explanation for higher fatigue scores associated with this SNP could be increased signaling of the canonical NFkB pathway that results in increased inflammatory responses.

One SNP (i.e., TNFRSF10A rs17620) and four liability scores (i.e., IL17RB, TNFRSF10D, TNFRSF11A and TNFRSF21) were associated with inter-individual differences in morning fatigue severity. TNFRSF10A rs17620 predicted inter-individual differences in the trajectory of morning fatigue. TNFRSF10A, after activation by tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), encodes for a protein that induces cellular apoptosis (http://www.ncbi.nlm.nih.gov/gene/8797). No predictions of the effects of this SNP on protein function were identified by SIFT or PolyPhen-2. While studies of the associations between TNFRSF10A rs17620 and fatigue were not found, TRAIL polymorphisms are associated with increased cancer risk [135, 136]. In our study, patients who were heterozygous or homozygous for the rare C allele were predicted to have a steeper trajectory of morning fatigue scores that suggests clinically meaningful increases in fatigue at four of the six assessments (i.e., assessments 2, 4, 5 and 6) (Figure 3A).

For IL17RB, the liability score for this gene and two SNPs in this gene (i.e., rs2232346, rs1043261) were associated with lower levels of morning and evening fatigue, respectively. IL17RB encodes for a protein that mediates the activation of the NFkB pathway and the production of C-X-C motif chemokine ligand 8 (CXCL8), a mediator of inflammatory responses. While no studies were found on an association between IL17RB polymorphisms and fatigue severity, overexpression of IL17RB is associated with increased tumorigenesis and metastasis [137, 138]. In our study, each additional copy of the rare allele for IL17RB was associated with lower predicted morning fatigue scores at enrollment (i.e., 0 alleles = 3.12, 1 allele = 2.77, 2 alleles = 2.42) (Figure 2C).

In terms of evening fatigue, two SNPs in IL17RB (i.e., rs2232346 and rs1043261) predicted the initial levels of evening fatigue at enrollment. IL17RB rs2232346 is a missense mutation that substitutes leucine for phenylalanine and is predicted to be tolerated by SIFT and benign by PolyPhen-2. No citations were identified for the effects of rs2232346. For IL17RB rs1043261, while no predictions of the functional effects were identified by the bioinformational tools, it was found to be associated with new-onset diabetes after renal transplantation [139].

Patients who were homozygous or heterozygous for the rare C allele of IL17RB rs2232346 or homozygous for rare C allele of IL17RB rs1043261 were predicted to have evening fatigue scores below clinically meaningful levels at all six assessments (Figure 6A). At enrollment, the effect size calculations for differences in predicted evening fatigue scores were in the moderate to large range for IL17RB rs2232346 (d = .4) and IL17RB rs1043261 (d = .7). Studies are needed to determine if the functional effects of these polymorphisms decrease inflammatory responses by inhibiting NFkB activation and decreasing the production of CXCL8.

The liability score for TNFRSF10D predicted the trajectory of morning fatigue. TNFRSF10D encodes for a protein that protects against TRAIL-mediated apoptosis and is part of the canonical NFkB pathway (http://www.ncbi.nlm.nih.gov/gene/8793). While no change was predicted in the trajectory of assessments 1, 2, and 3 (i.e., PW1), carrying more copies of the rare alleles lowered morning fatigue scores for assessments 4, 5, and 6 (i.e., PW2) (Figure 3B).

The liability score for TNFRSF11A predicted the trajectory of morning fatigue. TNFRSF11A encodes for a protein that is an activator of the non-canonical NFkB pathway to initiate inflammatory responses (http://www.ncbi.nlm.nih.gov/gene/8792). In our study, each additional copy of the rare allele of TNFRSF11A predicted lower morning fatigue scores across both piecewise models (Figure 3C).

The liability score for TNFRSF21 predicted initial levels of morning fatigue (Figure 2D). TNFRSF21 encodes for a protein that activates the canonical NFkB pathway and plays a role in neural cell apoptosis that is potentially related to the development of Alzheimer’s disease [140]. In our study, each additional copy of the rare allele for TNFRSF21 was associated with higher predicted morning fatigue scores at enrollment (i.e., 0 alleles = 2.92, 1 allele = 3.33, 2 alleles = 3.75).

The LTBR liability score was a unique predictor of evening fatigue. LTBR activates the non-canonical NFkB pathway and mediates cancer-associated inflammation [141]. While the two SNPs in the LTBR liability score were predicted to be tolerated by SIFT, no clinical studies of either SNP were identified. In our study, carrying the rare allele predicted a sharply decreased slope at assessment 2 (Figure 6C). Explanations for this trajectory at assessment 2 are not readily apparent and warrant further study to determine the effect of LTBR on the severity of evening fatigue.

An evaluation of the associations between polymorphisms in genes in the NFkB pathway identified in this study and changes in morning and evening fatigue severity reveals the complex nature of the mechanisms that underlie fatigue. While polymorphisms in four genes (i.e., IL17RB, TNFRSF10D, TNFRSF11A, LTBR) were associated with lower levels of morning and evening fatigue, polymorphisms in two genes (i.e. TNFRSF10A, TNFRSF14, TNFRSF21) were associated with higher levels of morning and evening fatigue. Future studies need to evaluate the functional effects of these polymorphisms and the interactions among the polymorphisms within and outside the NFkB pathway.

6.1 Cytokine genes

As key regulators of inflammation and immune responses, polymorphisms in cytokine genes were the most common variations evaluated as potential mechanisms for fatigue [14, 38, 55, 8590]. While previous studies found associations between polymorphisms in a number of cytokine genes and fatigue (for reviews see [29, 91]), in our study, the same polymorphisms in TNFA and IL12B were associated with decreases in both morning and evening fatigue severity.

TNFA encodes for a pleiotropic cytokine that regulates immune responses, as well as cell proliferation and differentiation. For TNFA rs1041981, individuals who carried one or two doses of the rare A allele reported slightly lower morning and evening fatigue severity scores (Figures 1C and and4D).4D). TNFA rs1041981 is located on chromosome 6p21.3. At this locus, the lymphotoxin-alpha (LTA) and TNFA genes are closely juxtaposed with the LTA coding region part of the TNFA early promoter region. While previous findings suggested that LTA shared a common signaling pathway with TNFA to mediate inflammatory responses [92], recent evidence suggests that the LTA and TNFA signaling pathways are unique [93, 94]. In the literature [95], rs1041981 is described as a SNP in both the LTA (i.e., coding region) and TNFA (i.e., promoter region) genes. When cited as a SNP in the LTA gene, rs1041981 was associated with inflammatory processes involved in atherogenesis [96], periodontal disease [97], and glucose intolerance [98]. When cited as a SNP in the promoter region of TNFA, rs1041981 was associated with sleep disturbance in patients with HIV [99]. These results suggest that as a promotor variant, this SNP may influence inflammatory responses through both TNFA and LTA signaling pathways.

In contrast to our findings, in the only study that evaluated the association between TNFA rs1041981 and morning and evening fatigue severity [100], patients with HIV disease who carried one or two doses of the rare A allele reported higher fatigue scores. These inconsistent findings may be related to differences in the way that the fatigue phenotypes were created. In the HIV study, mean scores were calculated. In our study, the effect of the polymorphism on initial levels as well as on the trajectories of morning and evening fatigue were evaluated. In addition, immune function in patients with HIV may have different signaling mechanisms than in oncology patients.

While, SIFT [83] and PolyPhen-2 [84] predicted a subtle impact of TNFA rs1041981 on function (i.e., tolerated by SIFT and benign by Polyphen-2), its impact on TNFA transcription is potentially more pronounced. The occurrence of the rare A allele changes the profile of transcription factors that recognize this sequence. With the homozygous common allele genotype (CC), six transcription factors are predicted to recognize the C-containing sequence (i.e., binding sites) (i.e., c_Ets-1 68, C/EBPalpha, C/EBPbeta, FOXP3, HNF-4alpha2, HNF-4alpha1) compared to only three transcription factors when the rare A allele is present (i.e., RXR-alpha, ETF, c-Ets-1 68). Reduced transcription factor activity was linked to reduced inflammatory signaling and decreased levels of fatigue in breast cancer survivors [101] and may be a potential explanation for the decreases in morning and evening fatigue severity reported by patients with the CA or AA genotype.

IL12B encodes for a proinflamatory cytokine that initiates the release of interferon gamma. The IL12B rs3213094 polymorphism was associated with lower initial levels of both morning and evening fatigue. No predictions for the functional effects of IL12B rs3213094 were identified using SIFT or PolyPhen-2. While no studies were found on an association between this SNP and fatigue severity, previous research found an association with the development of psoriasis [102].

A higher liability score for IL12B was associated with lower initial levels of evening fatigue. When the six SNPs found on IL12B were evaluated, four were predicted to be tolerated or benign damaging by SIFT and PolyPhen-2 respectively, and two were not identified in these bioinformational tools. As shown in Figure 4C, for patients with zero occurrences of the rare alleles, evening fatigue scores were above the clinically meaningful cutoff score for four of the six assessments (i.e., assessments 2, 4, 5, and 6). In contrast, patients with only three doses of the rare allele had evening fatigue scores that were below the clinically meaningful cutoff score for all six assessments. While no studies were found on associations between polymorphisms in IL12B and fatigue, one potential explanation for the predicted lower levels of evening fatigue could be decreased release of interferon gamma. Lower levels of interferon gamma were associated with lower levels of fatigue in patients with Sjögren’s syndrome [103].

It is important to note that cytokine dysregulation is associated with the co-occurrence of sleep disturbance, depressive symptoms, anxiety, and fatigue [56, 104]. Possible links among these co-occurring symptoms and polymorphisms in TNFA and IL12B require further study.

6.2 Inflammasome Pathway

Inflammasomes are multimeric protein complexes that serve as platforms for activation of the innate immune response through mediation of the NFkB and interferon signaling pathways [105, 106]. The formation of inflammasome complexes is activated by pattern recognition receptors (PRR) that detect danger-and pathogen-associated molecular patterns (i.e., DAMPs and PAMPs, respectively) [106]. These inflammasome complexes initiate a signaling pathway that activates capase-1 and transcription factor NFkB, which drives the expression of IL1β and IL18 [107]. To date, the most studied inflammasome complexes are activated by the Nod-Like Receptors (NLRs) family of PRRs.

In our study, five SNPs on five different NLR genes involved in inflammasome activation were associated with multiple effects on morning and evening fatigue. The same two SNPs, NOD2 rs2076756 and NLRP6 rs74044411 were associated with lower initial levels as well as with the trajectories of both morning and evening fatigue, respectively. One of the other SNPs (i.e., NLRP5 rs471979) was associated with the trajectory of morning fatigue. The other two SNPs (i.e., CARD6 rs10512747, NLRP4 rs17857373) were associated with initial levels of evening fatigue.

NOD2 expression results in activation of the MAPK and NFkB inflammatory pathways and a reduction in toll-like receptor mediated inflammatory responses [105]. These effects appear to be dependent on the degree of NOD2 expression [108]. While predictions of the functional effect of NOD2 rs2076756 were not identified by SIFT or Poly-Phen-2, rs2076756 is strongly associated with increased cancer risk [109], Crohn’s disease [110], and NOD2-associated autoinflammatory disease [111]. In addition, patients who are heterozygous (AG) or homozygous (GG) for the rare G allele are diagnosed with inflammatory bowel disease at a younger age [112]. In our study, patients who were homozygous (GG) for the rare G allele in NOD2 rs2076756 were predicted to have morning and evening fatigue below clinically meaningful levels across all six assessments (Figures 1D and and5C,5C, respectively). Explanations for this apparent protective effect of NOD2 on fatigue severity in oncology patients are not readily apparent. Given that the effects of NOD2 appear to be dependent on the degree of gene expression [108], future studies need to examine the effects of this specific polymorphism on differential gene expression in oncology patients with clinically meaningful differences in the severity of both morning and evening fatigue.

Recent evidence suggests that NLRP6 inhibits inflammasome and non-inflammasome dependent inflammatory responses [113, 114]. NLRP6 is associated with maintaining mucosal integrity of the gut and bacterial symbiosis [115]. NLRP6 rs74044411 is a missense mutation that substitutes alanine for valine. The SNP is predicted to be tolerated by SIFT and benign by PolyPhen-2. Patients who were heterozygous or homozygous for the rare C allele reported levels of morning (Figure 1F) and evening (Figure 5D) fatigue that were below the clinically meaningful cutoff levels (i.e., 3.2 and 5.6 respectively) at five of the six assessments. Given recent evidence that dysbiosis of the gut membrane was associated with increased fatigue in oncology patients receiving pelvic radiation [116] and our findings, future studies need to evaluate the functional effects of this SNP as well as the inter-relationships between alterations in the gut microbiome and fatigue severity.

Only one polymorphism in a gene from the inflammasome pathway (i.e., NLRP5 rs471979) predicted changes in the trajectory of morning fatigue. NLRP5 encodes for a protein that plays a role for zygotes to progress beyond the first embryonic cell divisions and is essential for RNA stability. Functional effects of this SNP were not identified by SIFT or PolyPhen-2. In addition, no studies were founding linking it to fatigue. Variations in NLRP5 were found to be associated with congenital disorders of growth and development [117], while higher NLRP5 expression was associated with periodontal disease in older adults [118]. In our study, the morning fatigue scores of patients who were homozygous for the C allele of NLRP5 rs471979 were predicted to be below the clinically meaningful cutoff score for five of the six assessments (Figure 1E).

Two SNPs from the inflammasome pathway (i.e., NLRP4 rs17857373, CARD6 rs10512747) were associated only with evening fatigue. NLRP4 rs17857373, predicted lower initial levels of evening fatigue. NLRP4 expression regulates type-1 interferon signaling and NF-kB activity induced by intracellular adapter proteins and kinases that functionally connect TNF and IL-1R receptors to NF-kB responses [105]. NLRP4 rs17857373 is a missense mutation that substitutes aspartic acid for glutamic acid. This change is predicted to be tolerated by SIFT and benign by PolyPhen-2. While no studies were found that described the effect of NLRP4 rs17857373, NLRP4 overexpression was associated with a diagnosis of bladder cancer [119], with an increased potential for metastasis and CTX resistance in women with epithelial ovarian cancer [120], and with periodontal disease [118]. In our study, patients who were homozygous or heterozygous for the rare G allele were predicted to have evening fatigue scores below the clinically meaningful cutoff at all six assessments (Figure 5B).

CARD6 encodes for a protein that regulates the adaptive and innate immune responses through modulation of interferon and NFkB signaling [121]. CARD6 appears to play a role in the development of GI cancers [122] and may protect cardiac muscle from hypertrophy [123]. CARD6 rs10512747 is a missense mutation that substitutes leucine for serine. This change is predicted to be tolerated by SIFT and potentially damaging by PolyPhen-2. While no studies were found that described the effect of this polymorphism, in our study, each additional dose of the rare C allele was associated with lower predicted evening fatigue scores at enrollment (i.e., TT = 5.40, TC = 4.96, CC = 4.52) (Figure 5A).

The function of the inflammasomes is an evolving area of investigation. In our study, all of the SNPs across the five genes in this pathway were associated with decreases in fatigue severity. Additional research is warranted to determine if the functional effects of these polymorphisms prevent the initiation of the inflammasome complex which may decrease inflammatory responses. While no studies were found that discussed the role of inflammasomes in the development of fatigue, it is reasonable to hypothesize that dysregulation of inflammasomes could result in decreases in IL-1B activity and decreases in innate inflammatory responses, and subsequent decreases in fatigue severity [124]. Additional research is warranted on this potential new mechanism for fatigue in oncology patients.

6.3 JAK/STAT pathway

The pleiotropic JAK/STAT pathway signals a cascade of reactions to maintain homeostasis [125]. The liability score for SNPs in IL4R predicted changes in the trajectory of morning fatigue. IL4R encodes for a cellular receptor for IL-4 and IL-13 that coordinates inflammatory responses through cytokine receptor signal transduction [126]. Of the seven individual SNPs included in the liability score, four were predicted to be tolerated or possibly damaging by SIFT and PolyPhen-2, respectively and three were not predicted by either bioinformational tool. In our study, for those patients who carried three or more rare alleles, their morning fatigue levels were predicted to be below the clinically meaningful cutoff score for five of the six assessments (Figure 2A). While no studies reported on an association between IL4R polymorphisms and fatigue severity, increased expression of IL4R was correlated with higher levels of fatigue in a sample of breast cancer survivors [50]. One potential explanation for the decreases in fatigue severity found in our study is that an increased number of polymorphisms in this gene changes the affinity of the cellular receptor which decreases the inflammatory responses associated with IL4 and IL13 signal transduction.

6.4 MAPK/JNK pathway

The MAPK/JNK pathway is both an upstream and downstream regulator of the expression of pro-inflammatory cytokines [127]. Only one SNP (i.e., IL17RD rs61742267) from the MAPK/JNK pathway was associated with evening fatigue. IL17RD (also called Sef [similar expression to fibroblast growth factor]) was found to be a tumor suppressor gene [128] and an inhibitor of toll-like receptor signaling [129]. IL17RD rs61742267 is a missense mutation that substitutes serine for proline. While no effect was predicted by PolyPhen-2, it was predicted by SIFT to have a potentially damaging effect on gene function. No studies were found that discussed the role of IL17RD in the development of fatigue. In our study, patients who are homozygous or heterozygous for the rare G allele were predicted to have evening fatigue at levels below the clinically meaningful cutoff at all six assessments (Figure 5E). One possible explanation for this finding could be that this polymorphism modulates toll-like receptor signaling which may decrease inflammatory responses that results in decreases in fatigue severity.

6.5 NFkB pathway

The NFkB pathway mediates cellular functions including apoptosis, cellular proliferation, and inflammatory responses through a complex signaling network [130]. This NFkB signaling network is regulated by canonical and non-canonical pathways. The canonical pathway is triggered by a variety of signals (e.g., members of the TNFRS family, IL1-R, toll-like receptors). The non-canonical NFkB pathway is triggered by specific members of the inhibitor of kappa B and TNFRSF families and LTBR [131]. Together, the canonical and non-canonical pathways regulate pro-and anti-inflammatory responses to prevent disorders that are associated with dysregulation of the NFkB pathway (e.g., rheumatoid arthritis, multiple sclerosis, inflammatory bowel disease) [132].

For the NFkB pathway, four SNPs and five liability scores across seven genes were associated with inter-individual differences in morning and evening fatigue. Only one SNP (i.e., TNFRSF14 rs2234163) was associated with initial levels of both morning and evening fatigue. TNFRSF14, encodes for a protein that both activates and inhibits T-cells based on the cellular environment [133]. TNFRSF14 rs2234163 is a missense mutation that substitutes threonine for alanine. It was predicted to be tolerated or possibly damaging by SIFT and PolyPhen-2, respectively. This SNP was associated with p53 mutations in Chinese women with GYN cancers [134]. In our study, compared to patients who were homozygous for the common allele (i.e., morning LFS score = 2.98, evening LFS score = 5.28), patients who were heterozygous or homozygous for the rare G allele of TNFRSF14 rs2234163, were predicted to have higher morning (i.e., LFS = 4.41) and evening (i.e., LFS = 6.87) scores (Figures 2B and and6B).6B). When these differences in fatigue scores were compared, caring one or two copies of the rare G allele was associated with clinically meaningful increases in both morning (d=.97) and evening (d=.75) fatigue severity. One possible explanation for higher fatigue scores associated with this SNP could be increased signaling of the canonical NFkB pathway that results in increased inflammatory responses.

One SNP (i.e., TNFRSF10A rs17620) and four liability scores (i.e., IL17RB, TNFRSF10D, TNFRSF11A and TNFRSF21) were associated with inter-individual differences in morning fatigue severity. TNFRSF10A rs17620 predicted inter-individual differences in the trajectory of morning fatigue. TNFRSF10A, after activation by tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), encodes for a protein that induces cellular apoptosis (http://www.ncbi.nlm.nih.gov/gene/8797). No predictions of the effects of this SNP on protein function were identified by SIFT or PolyPhen-2. While studies of the associations between TNFRSF10A rs17620 and fatigue were not found, TRAIL polymorphisms are associated with increased cancer risk [135, 136]. In our study, patients who were heterozygous or homozygous for the rare C allele were predicted to have a steeper trajectory of morning fatigue scores that suggests clinically meaningful increases in fatigue at four of the six assessments (i.e., assessments 2, 4, 5 and 6) (Figure 3A).

For IL17RB, the liability score for this gene and two SNPs in this gene (i.e., rs2232346, rs1043261) were associated with lower levels of morning and evening fatigue, respectively. IL17RB encodes for a protein that mediates the activation of the NFkB pathway and the production of C-X-C motif chemokine ligand 8 (CXCL8), a mediator of inflammatory responses. While no studies were found on an association between IL17RB polymorphisms and fatigue severity, overexpression of IL17RB is associated with increased tumorigenesis and metastasis [137, 138]. In our study, each additional copy of the rare allele for IL17RB was associated with lower predicted morning fatigue scores at enrollment (i.e., 0 alleles = 3.12, 1 allele = 2.77, 2 alleles = 2.42) (Figure 2C).

In terms of evening fatigue, two SNPs in IL17RB (i.e., rs2232346 and rs1043261) predicted the initial levels of evening fatigue at enrollment. IL17RB rs2232346 is a missense mutation that substitutes leucine for phenylalanine and is predicted to be tolerated by SIFT and benign by PolyPhen-2. No citations were identified for the effects of rs2232346. For IL17RB rs1043261, while no predictions of the functional effects were identified by the bioinformational tools, it was found to be associated with new-onset diabetes after renal transplantation [139].

Patients who were homozygous or heterozygous for the rare C allele of IL17RB rs2232346 or homozygous for rare C allele of IL17RB rs1043261 were predicted to have evening fatigue scores below clinically meaningful levels at all six assessments (Figure 6A). At enrollment, the effect size calculations for differences in predicted evening fatigue scores were in the moderate to large range for IL17RB rs2232346 (d = .4) and IL17RB rs1043261 (d = .7). Studies are needed to determine if the functional effects of these polymorphisms decrease inflammatory responses by inhibiting NFkB activation and decreasing the production of CXCL8.

The liability score for TNFRSF10D predicted the trajectory of morning fatigue. TNFRSF10D encodes for a protein that protects against TRAIL-mediated apoptosis and is part of the canonical NFkB pathway (http://www.ncbi.nlm.nih.gov/gene/8793). While no change was predicted in the trajectory of assessments 1, 2, and 3 (i.e., PW1), carrying more copies of the rare alleles lowered morning fatigue scores for assessments 4, 5, and 6 (i.e., PW2) (Figure 3B).

The liability score for TNFRSF11A predicted the trajectory of morning fatigue. TNFRSF11A encodes for a protein that is an activator of the non-canonical NFkB pathway to initiate inflammatory responses (http://www.ncbi.nlm.nih.gov/gene/8792). In our study, each additional copy of the rare allele of TNFRSF11A predicted lower morning fatigue scores across both piecewise models (Figure 3C).

The liability score for TNFRSF21 predicted initial levels of morning fatigue (Figure 2D). TNFRSF21 encodes for a protein that activates the canonical NFkB pathway and plays a role in neural cell apoptosis that is potentially related to the development of Alzheimer’s disease [140]. In our study, each additional copy of the rare allele for TNFRSF21 was associated with higher predicted morning fatigue scores at enrollment (i.e., 0 alleles = 2.92, 1 allele = 3.33, 2 alleles = 3.75).

The LTBR liability score was a unique predictor of evening fatigue. LTBR activates the non-canonical NFkB pathway and mediates cancer-associated inflammation [141]. While the two SNPs in the LTBR liability score were predicted to be tolerated by SIFT, no clinical studies of either SNP were identified. In our study, carrying the rare allele predicted a sharply decreased slope at assessment 2 (Figure 6C). Explanations for this trajectory at assessment 2 are not readily apparent and warrant further study to determine the effect of LTBR on the severity of evening fatigue.

An evaluation of the associations between polymorphisms in genes in the NFkB pathway identified in this study and changes in morning and evening fatigue severity reveals the complex nature of the mechanisms that underlie fatigue. While polymorphisms in four genes (i.e., IL17RB, TNFRSF10D, TNFRSF11A, LTBR) were associated with lower levels of morning and evening fatigue, polymorphisms in two genes (i.e. TNFRSF10A, TNFRSF14, TNFRSF21) were associated with higher levels of morning and evening fatigue. Future studies need to evaluate the functional effects of these polymorphisms and the interactions among the polymorphisms within and outside the NFkB pathway.

7. Limitations and strengths

Several limitations and strengths need to be acknowledged. While our sample size was adequate, these findings warrant replication. Because patients completed the questionnaires in their homes rather than in the clinic, it may have influenced their reports of fatigue severity. In our study, a liability score assumes that the rare alleles across all of the SNPs in a specific gene region carry a similar risk (e.g., all protective). It is possible that discordant allele effects could result in false negatives or bias the results obtained from using liability scores. Future analyses of the functional effects of each SNP could be aggregated into a liability score that might account for discordant allele effects. However, this large, representative sample of oncology outpatients undergoing CTX, the evaluation of morning and evening fatigue across two cycles of CTX, and the use of HLM to identify genetic predictors of inter-individual variability in morning and evening fatigue are major strengths of this study. Our conceptual analysis of the genomic data within functional pathways contextualizes the results that can be used to inform future hypothesis of how functionally related genes collectively affect morning and evening fatigue severity.

8. Conclusion

This study extends the evidence that morning and evening fatigue are distinct yet related symptoms. In addition, new inflammatory pathways were identified that play potential roles in the complex mechanisms that are involved in the development of morning and evening fatigue. Future research with pathway analysis will help us to clarify the biological processes that contribute to inter-individual variability in the severity of morning and evening fatigue so that we can tailor interventions to prevent or alleviate these distinct but related symptoms.

Supplementary Material

Click here to view.(224K, docx)

Acknowledgments

This study was funded by the National Cancer Institute (NCI, {"type":"entrez-nucleotide","attrs":{"text":"CA134900","term_id":"35022380","term_text":"CA134900"}}CA134900). Dr. Miaskowski is supported by a grant from the American Cancer Society and NCI ({"type":"entrez-nucleotide","attrs":{"text":"CA168960","term_id":"35090906","term_text":"CA168960"}}CA168960). Dr. Wright is funded by the National Institute of Nursing Research post-doctoral training program T32NR008346 at Yale University School of Nursing.

Yale School of Nursing, New Haven, CT, USA
Department of Nursing, Mount Sinai Hospital, New York, NY, USA
Department of Physiologic Nursing, School of Nursing, University of California at San Francisco, San Francisco, CA, USA
Bluestone Center for Clinical Research, Department of Oral and Maxillofacial Surgery, School of Dentistry, New York University, New York, NY, USA
School of Nursing, University of Pittsburgh, Pittsburgh, PA, USA
Department of Psychiatry, School of Medicine, Stanford University, Palo Alto, CA, USA
Department of Oral and Maxillofacial Surgery, School of Dentistry, University of California at San Francisco, San Francisco, CA, USA
Florence S. Downs PhD Program in Nursing Research and Theory Development, College of Nursing, New York University, New York, NY, USA
Address correspondence to: Christine Miaskowski, RN, PhD, Professor, Department of Physiological Nursing, University of California, 2 Koret Way – N631Y, San Francisco, CA 94143-0610, 415-476-9407 (phone), 415-476-8899 (fax), ude.fscu@ikswoksaim.sirhc
Publisher's Disclaimer

Abstract

Fatigue, a highly prevalent and distressing symptom during chemotherapy (CTX), demonstrates diurnal and interindividual variability in severity. Little is known about the associations between variations in genes involved in inflammatory processes and morning and evening fatigue severity during CTX. The purposes of this study, in a sample of oncology patients (N=543) with breast, gastrointestinal (GI), gynecological (GYN), or lung cancer who received two cycles of CTX, were to determine whether variations in genes involved in inflammatory processes were associated with inter-individual variability in initial levels as well as in the trajectories of morning and evening fatigue. Patients completed the Lee Fatigue Scale to determine morning and evening fatigue severity a total of six times over two cycles of CTX. Using a whole exome array, 309 single nucleotide polymorphisms among the 64 candidate genes that passed all quality control filters were evaluated using hierarchical linear modeling (HLM). Based on the results of the HLM analyses, the final SNPs were evaluated for their potential impact on protein function using two bioinformational tools. The following inflammatory pathways were represented: chemokines (3 genes); cytokines (12 genes); inflammasome (11 genes); Janus kinase/signal transducers and activators of transcription (JAK/STAT, 10 genes); mitogen-activated protein kinase/jun amino-terminal kinases (MAPK/JNK, 3 genes); nuclear factor-kappa beta (NFkB, 18 genes); and NFkB and MAP/JNK (7 genes). After controlling for self-reported and genomic estimates of race and ethnicity, polymorphisms in six genes from the cytokine (2 genes); inflammasome (2 genes); and NFkB (2 genes) pathways were associated with both morning and evening fatigue. Polymorphisms in six genes from the inflammasome (1 gene); JAK/STAT (1 gene); and NFkB (4 genes) pathways were associated with only morning fatigue. Polymorphisms in three genes from the inflammasome (2 genes) and the NFkB (1 gene) pathways were associated with only evening fatigue. Taken together, these findings add to the growing body of evidence that suggests that morning and evening fatigue are distinct symptoms.

Keywords: inflammation, genes, fatigue, hierarchical linear modeling, diurnal variability, cancer, chemotherapy
Abstract
Highlights

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

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

Footnotes

References

  • 1. Berger AM, Mitchell SA, Jacobsen PB, Pirl WFScreening, evaluation, and management of cancer-related fatigue: Ready for implementation to practice? CA Cancer J Clin. 2015;65(3):190–211.[PubMed][Google Scholar]
  • 2. Fisch MJ, Zhao F, O'Mara AM, Wang XS, Cella D, Cleeland CSPredictors of significant worsening of patient-reported fatigue over a 1-month timeframe in ambulatory patients with common solid tumors. Cancer. 2014;120(3):442–450.[Google Scholar]
  • 3. Huang HP, Chen ML, Liang J, Miaskowski CChanges in and predictors of severity of fatigue in women with breast cancer: A longitudinal study. Int J Nurs Stud. 2014;51(4):582–592.[PubMed][Google Scholar]
  • 4. Ancoli-Israel S, Liu L, Rissling M, Natarajan L, Neikrug AB, Palmer BW, Mills PJ, Parker BA, Sadler GR, Maglione JSleep, fatigue, depression, and circadian activity rhythms in women with breast cancer before and after treatment: a 1-year longitudinal study. Support Care Cancer. 2014;22(9):2535–2545.[Google Scholar]
  • 5. Trudel-Fitzgerald C, Savard J, Ivers HEvolution of cancer-related symptoms over an 18-month period. J Pain Symptom Manage. 2013;45(6):1007–1018.[PubMed][Google Scholar]
  • 6. Torres MA, Pace TW, Liu T, Felger JC, Mister D, Doho GH, Kohn JN, Barsevick AM, Long Q, Miller AHPredictors of depression in breast cancer patients treated with radiation: role of prior chemotherapy and nuclear factor kappa B. Cancer. 2013;119(11):1951–1959.[Google Scholar]
  • 7. Miaskowski C, Paul SM, Cooper BA, Lee K, Dodd M, West C, Aouizerat BE, Swift PS, Wara WTrajectories of fatigue in men with prostate cancer before, during, and after radiation therapy. J Pain Symptom Manage. 2008;35(6):632–643.[Google Scholar]
  • 8. Dhruva A, Aouizerat BE, Cooper B, Paul SM, Dodd M, West C, Wara W, Lee K, Dunn LB, Langford DJ, Merriman JD, Baggott C, Cataldo J, Ritchie C, Kober K, Leutwyler H, Miaskowski CDifferences in morning and evening fatigue in oncology patients and their family caregivers. Eur J Oncol Nurs. 2013;17(6):841–848.[Google Scholar]
  • 9. Wright F, D'Eramo Melkus G, Hammer M, Schmidt BL, Knobf MT, Paul SM, Cartwright F, Mastick J, Cooper BA, Chen LM, Melisko M, Levine JD, Kober K, Aouizerat BE, Miaskowski CPredictors and trajectories of morning fatigue are distinct from evening fatigue. J Pain Symptom Manage. 2015;50(2):176–189.[Google Scholar]
  • 10. Wright F, D'Eramo Melkus G, Hammer M, Schmidt BL, Knobf MT, Paul SM, Cartwright F, Mastick J, Cooper BA, Chen LM, Melisko M, Levine JD, Kober K, Aouizerat BE, Miaskowski CTrajectories of evening fatigue in oncology outpatients receiving chemotherapy. J Pain Symptom Manage. 2015;50(2):163–175.[Google Scholar]
  • 11. Kober KM, Cooper BA, Paul SM, Dunn LB, Levine JD, Wright F, Hammer MJ, Mastick J, Venook A, Aouizerat BE, Miaskowski CSubgroups of chemotherapy patients with distinct morning and evening fatigue trajectories. Support Care Cancer. 2016;24(4):1473–1485.[Google Scholar]
  • 12. Ratcliff CG, Lam CY, Arun B, Valero V, Cohen LEcological momentary assessment of sleep, symptoms, and mood during chemotherapy for breast cancer. Psychooncology. 2014;23(11):1220–1228.[Google Scholar]
  • 13. Aouizerat BE, Dodd M, Lee K, West C, Paul SM, Cooper BA, Wara W, Swift P, Dunn LB, Miaskowski CPreliminary evidence of a genetic association between tumor necrosis factor alpha and the severity of sleep disturbance and morning fatigue. Biol Res Nurs. 2009;11(1):27–41.[PubMed][Google Scholar]
  • 14. Dhruva A, Aouizerat BE, Cooper B, Paul SM, Dodd M, West C, Wara W, Lee K, Dunn LB, Langford DJ, Merriman JD, Baggott C, Cataldo J, Ritchie C, Kober KM, Leutwyler H, Miaskowski CCytokine gene associations with self-report ratings of morning and evening fatigue in oncology patients and their family caregivers. Biol Res Nurs. 2015;17(2):175–184.[Google Scholar]
  • 15. Miaskowski C, Dodd M, Lee K, West C, Paul SM, Cooper BA, Wara W, Swift PS, Dunn LB, Aouizerat BEPreliminary evidence of an association between a functional interleukin-6 polymorphism and fatigue and sleep disturbance in oncology patients and their family caregivers. J Pain Symptom Manage. 2010;40(4):531–544.[Google Scholar]
  • 16. LaVoy EC, Fagundes CP, Dantzer RExercise, inflammation, and fatigue in cancer survivors. Exerc Immunol Rev. 2016;22:82–93.[Google Scholar]
  • 17. Dantzer R, Heijnen CJ, Kavelaars A, Laye S, Capuron LThe neuroimmune basis of fatigue. Trends Neurosci. 2014;37(1):39–46.[Google Scholar]
  • 18. Saligan LN, Olson K, Filler K, Larkin D, Cramp F, Yennurajalingam S, Escalante CP, del Giglio A, Kober KM, Kamath J, Palesh O, Mustian K, Multinational GAssociation of Supportive Care in Cancer Fatigue Study Group-Biomarker Working, The biology of cancer-related fatigue: a review of the literature. Support Care Cancer. 2015;23(8):2461–2478.[Google Scholar]
  • 19. Vichaya EG, Chiu GS, Krukowski K, Lacourt TE, Kavelaars A, Dantzer R, Heijnen CJ, Walker AKMechanisms of chemotherapy-induced behavioral toxicities. Front Neurosci. 2015;9:131.[Google Scholar]
  • 20. Pertl MM, Hevey D, Boyle NT, Hughes MM, Collier S, O'Dwyer AM, Harkin A, Kennedy MJ, Connor TJC-reactive protein predicts fatigue independently of depression in breast cancer patients prior to chemotherapy. Brain, Behav Immun. 2013;34:108–119.[PubMed][Google Scholar]
  • 21. Sha F, Zhuang S, Zhou L, Zhang L, Yang Y, Zhang S, Jiang Y, Qiu G, Chen C, Zheng J, Zhang SBiomarkers for cancer-related fatigue and adverse reactions to chemotherapy in lung cancer patients. Mol Clin Oncol. 2015;3(1):163–166.[Google Scholar]
  • 22. Ahlberg K, Ekman T, Gaston-Johansson FLevels of fatigue compared to levels of cytokines and hemoglobin during pelvic radiotherapy: a pilot study. Biol Res Nurs. 2004;5(3):203–210.[PubMed][Google Scholar]
  • 23. Clevenger L, Schrepf A, Christensen D, DeGeest K, Bender D, Ahmed A, Goodheart MJ, Penedo F, Lubaroff DM, Sood AK, Lutgendorf SKSleep disturbance, cytokines, and fatigue in women with ovarian cancer. Brain Behav Immun. 2012;26(7):1037–1044.[Google Scholar]
  • 24. de Raaf PJ, Sleijfer S, Lamers CH, Jager A, Gratama JW, van der Rijt CCInflammation and fatigue dimensions in advanced cancer patients and cancer survivors: an explorative study. Cancer. 2012;118(23):6005–6011.[PubMed][Google Scholar]
  • 25. De Sanctis V, Agolli L, Visco V, Monaco F, Muni R, Spagnoli A, Campanella B, Valeriani M, Minniti G, Osti MF, Amanti C, Pellegrini P, Brunetti S, Costantini A, Alfo M, Torrisi MR, Marchetti P, Enrici RMCytokines, fatigue, and cutaneous erythema in early stage breast cancer patients receiving adjuvant radiation therapy. BioMed Res Int. 2014;2014:523568.[Google Scholar]
  • 26. Inagaki M, Isono M, Okuyama T, Sugawara Y, Akechi T, Akizuki N, Fujimori M, Mizuno M, Shima Y, Kinoshita H, Uchitomi YPlasma interleukin-6 and fatigue in terminally ill cancer patients. J Pain Symptom Manage. 2008;35(2):153–161.[PubMed][Google Scholar]
  • 27. Liu L, Mills PJ, Rissling M, Fiorentino L, Natarajan L, Dimsdale JE, Sadler GR, Parker BA, Ancoli-Israel SFatigue and sleep quality are associated with changes in inflammatory markers in breast cancer patients undergoing chemotherapy. Brain Behav Immun. 2012;26(5):706–713.[Google Scholar]
  • 28. Schrepf A, Clevenger L, Christensen D, DeGeest K, Bender D, Ahmed A, Goodheart MJ, Dahmoush L, Penedo F, Lucci JA, 3rd, Ganjei-Azar P, Mendez L, Markon K, Lubaroff DM, Thaker PH, Slavich GM, Sood AK, Lutgendorf SKCortisol and inflammatory processes in ovarian cancer patients following primary treatment: relationships with depression, fatigue, and disability. Brain Behav Immun. 2013;30(Suppl):S126–S134.[Google Scholar]
  • 29. Schubert C, Hong S, Natarajan L, Mills PJ, Dimsdale JEThe association between fatigue and inflammatory marker levels in cancer patients: a quantitative review. Brain Behav Immun. 2007;21(4):413–427.[PubMed][Google Scholar]
  • 30. Kwak SM, Choi YS, Yoon HM, Kim DG, Song SH, Lee YJ, Yeom CH, Koh SJ, Park J, Lee MA, Suh SYThe relationship between interleukin-6, tumor necrosis factor-alpha, and fatigue in terminally ill cancer patients. Palliat Med. 2012;26(3):275–282.[PubMed][Google Scholar]
  • 31. Rich T, Innominato PF, Boerner J, Mormont MC, Iacobelli S, Baron B, Jasmin C, Levi FElevated serum cytokines correlated with altered behavior, serum cortisol rhythm, and dampened 24-hour rest-activity patterns in patients with metastatic colorectal cancer. Clin Cancer Res. 2005;11(5):1757–1764.[PubMed][Google Scholar]
  • 32. Shafqat A, Einhorn LH, Hanna N, Sledge GW, Hanna A, Juliar BE, Monahan P, Bhatia SScreening studies for fatigue and laboratory correlates in cancer patients undergoing treatment. Ann Oncol. 2005;16(9):1545–1550.[PubMed][Google Scholar]
  • 33. Von Ah DM, Kang DH, Carpenter JSPredictors of cancer-related fatigue in women with breast cancer before, during, and after adjuvant therapy. Cancer Nurs. 2008;31(2):134–144.[PubMed][Google Scholar]
  • 34. Bower JE, Ganz PA, Irwin MR, Arevalo JM, Cole SWFatigue and gene expression in human leukocytes: increased NF-kappaB and decreased glucocorticoid signaling in breast cancer survivors with persistent fatigue. Brain Behav Immun. 2011;25(1):147–150.[Google Scholar]
  • 35. Bower JE, Ganz PA, Tao ML, Hu W, Belin TR, Sepah S, Cole S, Aziz NInflammatory biomarkers and fatigue during radiation therapy for breast and prostate cancer. Clin Cancer Res. 2009;15(17):5534–5540.[Google Scholar]
  • 36. Cameron BA, Bennett B, Li H, Boyle F, DeSouza P, Wilcken N, Friedlander M, Goldstein D, Lloyd ARPost-cancer fatigue is not associated with immune activation or altered cytokine production. Ann Oncol. 2012;23(11):2890–2895.[PubMed][Google Scholar]
  • 37. Collado-Hidalgo A, Bower JE, Ganz PA, Irwin MR, Cole SWCytokine gene polymorphisms and fatigue in breast cancer survivors: early findings. Brain Behav Immun. 2008;22(8):1197–1200.[Google Scholar]
  • 38. Dirksen SR, Kirschner KF, Belyea MJAssociation of symptoms and cytokines in prostate cancer patients receiving radiation treatment. Biol Res Nurs. 2014;16(3):250–257.[PubMed][Google Scholar]
  • 39. Gilbertson-White S, Aouizerat BE, Miaskowski CMethodologic issues in the measurement of cytokines to elucidate the biological basis for cancer symptoms. Biol Res Nurs. 2011;13(1):15–24.[PubMed][Google Scholar]
  • 40. Bower JECancer-related fatigue--mechanisms, risk factors, and treatments. Nat Rev Clin Oncol. 2014;11(10):597–609.[Google Scholar]
  • 41. Bower JE, Ganz PA, Aziz N, Fahey JLFatigue and proinflammatory cytokine activity in breast cancer survivors. Psychosom Med. 2002;64(4):604–611.[PubMed][Google Scholar]
  • 42. Orre IJ, Murison R, Dahl AA, Ueland T, Aukrust P, Fossa SDLevels of circulating interleukin-1 receptor antagonist and C-reactive protein in long-term survivors of testicular cancer with chronic cancer-related fatigue. Brain Behav Immun. 2009;23(6):868–874.[PubMed][Google Scholar]
  • 43. Bower JE, Ganz PA, Irwin MR, Kwan L, Breen EC, Cole SWInflammation and behavioral symptoms after breast cancer treatment: do fatigue, depression, and sleep disturbance share a common underlying mechanism? J Clin Oncol. 2011;29(26):3517–3522.[Google Scholar]
  • 44. Smith AK, Conneely KN, Pace TW, Mister D, Felger JC, Kilaru V, Akel MJ, Vertino PM, Miller AH, Torres MAEpigenetic changes associated with inflammation in breast cancer patients treated with chemotherapy. Brain Behav Immun. 2014;38:227–236.[Google Scholar]
  • 45. Wang XS, Williams LA, Krishnan S, Liao Z, Liu P, Mao L, Shi Q, Mobley GM, Woodruff JF, Cleeland CSSerum sTNF-R1, IL-6, and the development of fatigue in patients with gastrointestinal cancer undergoing chemoradiation therapy. Brain Behav Immun. 2012;26(5):699–705.[Google Scholar]
  • 46. Courtier N, Gambling T, Enright S, Barrett-Lee P, Abraham J, Mason MDPsychological and immunological characteristics of fatigued women undergoing radiotherapy for early-stage breast cancer. Support Care Cancer. 2013;21(1):173–181.[PubMed][Google Scholar]
  • 47. Reinertsen KV, Grenaker Alnaes GI, Landmark-Hoyvik H, Loge JH, Wist E, Kristensen VN, Fossa SD, Edvardsen HFatigued breast cancer survivors and gene polymorphisms in the inflammatory pathway. Brain Behav Immun. 2011;25(7):1376–1383.[PubMed][Google Scholar]
  • 48. Hsiao CP, Araneta M, Wang XM, Saligan LNThe association of IFI27 expression and fatigue intensification during localized radiation therapy: implication of a para-inflammatory bystander response. Int J Mol Sci. 2013;14(8):16943–16957.[Google Scholar]
  • 49. Hsiao CP, Wang D, Kaushal A, Saligan LMitochondria-related gene expression changes are associated with fatigue in patients with nonmetastatic prostate cancer receiving external beam radiation therapy. Cancer Nurs. 2013;36(3):189–197.[Google Scholar]
  • 50. Landmark-Hoyvik H, Reinertsen KV, Loge JH, Fossa SD, Borresen-Dale AL, Dumeaux VAlterations of gene expression in blood cells associated with chronic fatigue in breast cancer survivors. Pharmacogenomics J. 2009;9(5):333–340.[PubMed][Google Scholar]
  • 51. Light KC, Agarwal N, Iacob E, White AT, Kinney AY, VanHaitsma TA, Aizad H, Hughen RW, Bateman L, Light ARDiffering leukocyte gene expression profiles associated with fatigue in patients with prostate cancer versus chronic fatigue syndrome. Psychoneuroendocrinology. 2013;38(12):2983–2995.[Google Scholar]
  • 52. Saligan LN, Hsiao CP, Wang D, Wang XM, St John L, Kaushal A, Citrin D, Barb JJ, Munson PJ, Dionne RAUpregulation of alpha-synuclein during localized radiation therapy signals the association of cancer-related fatigue with the activation of inflammatory and neuroprotective pathways. Brain Behav Immun. 2013;27(1):63–70.[Google Scholar]
  • 53. Kober KM, Dunn L, Mastick J, Cooper B, Langford D, Melisko M, Venook A, Chen LM, Wright F, Hammer M, Schmidt BL, Levine J, Miaskowski C, Aouizerat BEGene expression profiling of evening fatigue in women undergoing chemotherapy for breast cancer. Biol Res Nurs. 2016;18(4):370–385.[Google Scholar]
  • 54. Bower JE, Ganz PA, Irwin MR, Castellon S, Arevalo J, Cole SWCytokine genetic variations and fatigue among patients with breast cancer. J Clin Oncol. 2013;31(13):1656–1661.[Google Scholar]
  • 55. Jim HS, Park JY, Permuth-Wey J, Rincon MA, Phillips KM, Small BJ, Jacobsen PBGenetic predictors of fatigue in prostate cancer patients treated with androgen deprivation therapy: preliminary findings. Brain Behav Immun. 2012;26(7):1030–1036.[Google Scholar]
  • 56. Bower JE, Lamkin DMInflammation and cancer-related fatigue: mechanisms, contributing factors, and treatment implications. Brain Behav Immun. 2013;30(Suppl):S48–S57.[Google Scholar]
  • 57. Shi Q, Wang XS, Li G, Shah ND, Orlowski RZ, Williams LA, Mendoza TR, Cleeland CSRacial/ethnic disparities in inflammatory gene single-nucleotide polymorphisms as predictors of a high risk for symptom burden in patients with multiple myeloma 1 year after diagnosis. Cancer. 2015;121(7):1138–1146.[Google Scholar]
  • 58. Rausch SM, Clark MM, Patten C, Liu H, Felten S, Li Y, Sloan J, Yang PRelationship between cytokine gene single nucleotide polymorphisms and symptom burden and quality of life in lung cancer survivors. Cancer. 2010;116(17):4103–4113.[Google Scholar]
  • 59. Wood LJ, Weymann KInflammation and neural signaling: etiologic mechanisms of the cancer treatment-related symptom cluster. Current Opin Support Palliat Care. 2013;7(1):54–59.[Google Scholar]
  • 60. Zhang ZT, Du XM, Ma XJ, Zong Y, Chen JK, Yu CL, Liu YG, Chen YC, Zhao LJ, Lu GCActivation of the NLRP3 inflammasome in lipopolysaccharide-induced mouse fatigue and its relevance to chronic fatigue syndrome. J Neuroinflammation. 2016;13(1):71.[Google Scholar]
  • 61. Ramanan VK, Shen L, Moore JH, Saykin AJPathway analysis of genomic data: concepts, methods, and prospects for future development. Trends Genet. 2012;28(7):323–332.[Google Scholar]
  • 62. Miaskowski C, Cooper BA, Melisko M, Chen LM, Mastick J, West C, Paul SM, Dunn LB, Schmidt BL, Hammer M, Cartwright F, Wright F, Langford DJ, Lee K, Aouizerat BEDisease and treatment characteristics do not predict symptom occurrence profiles in oncology outpatients receiving chemotherapy. Cancer. 2014;120(15):2371–2378.[Google Scholar]
  • 63. Alfaro E, Dhruva A, Langford DJ, Koetters T, Merriman JD, West C, Dunn LB, Paul SM, Cooper B, Cataldo J, Hamolsky D, Elboim C, Kober K, Aouizerat BE, Miaskowski CAssociations between cytokine gene variations and self-reported sleep disturbance in women following breast cancer surgery. Eur J Oncol Nurs. 2014;18(1):85–93.[Google Scholar]
  • 64. Aouizerat BE, Dhruva A, Paul SM, Cooper BA, Kober KM, Miaskowski CPhenotypic and molecular evidence suggests that decrements in morning and evening energy are distinct but related symptoms. J Pain Symptom Manage. 2015;50(5):599 e3–614 e3.[Google Scholar]
  • 65. Karnofsky D Performance Scale. New York, NY: Plenum Press; 1977. [PubMed][Google Scholar]
  • 66. Sangha O, Stucki G, Liang MH, Fossel AH, Katz JNThe Self-Administered Comorbidity Questionnaire: a new method to assess comorbidity for clinical and health services research. Arthritis Rheum. 2003;49(2):156–163.[PubMed][Google Scholar]
  • 67. Spielberger CG, Gorsuch RL, Suchene R, Vagg PR, Jacobs GA Manual for the State-Anxiety (Form Y): Self Evaluation Questionnaire. Palo Alto, CA: Consulting Psychologists Press; 1983. [PubMed][Google Scholar]
  • 68. Radloff LSThe CES-D Scale: A self-report depression scale for research in the general population. Applied Psychological Measurement. 1977;1(3):385–401.[PubMed][Google Scholar]
  • 69. Lee KASelf-reported sleep disturbances in employed women. Sleep. 1992;15(6):493–498.[PubMed][Google Scholar]
  • 70. Lee KA, Hicks G, Nino-Murcia GValidity and reliability of a scale to assess fatigue. Psychiatry Res. 1991;36(3):291–298.[PubMed][Google Scholar]
  • 71. IBM SPSS Statistics for Windows, Version 22. Armonk, NY: IBM Corp; 2013. Released. [PubMed]
  • 72. Halder I, Shriver M, Thomas M, Fernandez JR, Frudakis TA panel of ancestry informative markers for estimating individual biogeographical ancestry and admixture from four continents: utility and applications. Human mutation. 2008;29(5):648–658.[PubMed][Google Scholar]
  • 73. Hoggart CJ, Parra EJ, Shriver MD, Bonilla C, Kittles RA, Clayton DG, McKeigue PMControl of confounding of genetic associations in stratified populations. Am J Hum Genet. 2003;72(6):1492–1504.[Google Scholar]
  • 74. Tian C, Gregersen PK, Seldin MFAccounting for ancestry: population substructure and genome-wide association studies. Hum Mol Genet. 2008;17(R2):R143–R150.[Google Scholar]
  • 75. Price AL, Patterson NJ, Plenge RM, Weinblatt ME, Shadick NA, Reich DPrincipal components analysis corrects for stratification in genome-wide association studies. Nat Genet. 2006;38(8):904–909.[PubMed][Google Scholar]
  • 76. Raudenbush SW, Bryk A Hierarchical linear models: Applications and data analysis methods. 2nd. Thousand Oaks, CA: Sage Publications; 2002. [PubMed][Google Scholar]
  • 77. McCann B, Miaskowski C, Koetters T, Baggott C, West C, Levine JD, Elboim C, Abrams G, Hamolsky D, Dunn L, Rugo H, Dodd M, Paul SM, Neuhaus J, Cooper B, Schmidt B, Langford D, Cataldo J, Aouizerat BEAssociations between pro- and anti-inflammatory cytokine genes and breast pain in women prior to breast cancer surgery. J Pain. 2012;13(5):425–437.[Google Scholar]
  • 78. Miaskowski C, Cooper BA, Dhruva A, Dunn LB, Langford DJ, Cataldo JK, Baggott CR, Merriman JD, Dodd M, Lee K, West C, Paul SM, Aouizerat BEEvidence of associations between cytokine genes and subjective reports of sleep disturbance in oncology patients and their family caregivers. PLoS One. 2012;7(7):e40560.[Google Scholar]
  • 79. Miaskowski C, Cataldo JK, Baggott CR, West C, Dunn LB, Dhruva A, Merriman JD, Langford DJ, Kober KM, Paul SM, Cooper BA, Aouizerat BECytokine gene variations associated with trait and state anxiety in oncology patients and their family caregivers. Support Care Cancer. 2015;23(4):953–965.[Google Scholar]
  • 80. Merriman JD, Aouizerat BE, Cataldo JK, Dunn L, Cooper BA, West C, Paul SM, Baggott CR, Dhruva A, Kober K, Langford DJ, Leutwyler H, Ritchie CS, Abrams G, Dodd M, Elboim C, Hamolsky D, Melisko M, Miaskowski CAssociation between an interleukin 1 receptor, type I promoter polymorphism and self-reported attentional function in women with breast cancer. Cytokine. 2014;65(2):192–201.[Google Scholar]
  • 81. Rothman KJNo adjustments are needed for multiple comparisons. Epidemiology. 1990;1(1):43–46.[PubMed][Google Scholar]
  • 82. Hattersley AT, McCarthy MIWhat makes a good genetic association study? Lancet. 2005;366(9493):1315–1323.[PubMed][Google Scholar]
  • 83. Kumar P, Henikoff S, Ng PCPredicting the effects of coding non-synonymous variants on protein function using the SIFT algorithm. Nat Protoc. 2009;4(7):1073–1081.[PubMed][Google Scholar]
  • 84. Adzhubei IA, Schmidt S, Peshkin L, Ramensky VE, Gerasimova A, Bork P, Kondrashov AS, Sunyaev SRA method and server for predicting damaging missense mutations. Nat Methods. 2010;7(4):248–249.[Google Scholar]
  • 85. Barsevick A, Frost M, Zwinderman A, Hall P, Halyard M, Consortium GI'm so tired: biological and genetic mechanisms of cancer-related fatigue. Qual Life Res. 2010;19(10):1419–1427.[Google Scholar]
  • 86. Berger AM, Abernethy AP, Atkinson A, Barsevick AM, Breitbart WS, Cella D, Cimprich B, Cleeland C, Eisenberger MA, Escalante CP, Jacobsen PB, Kaldor P, Ligibel JA, Murphy BA, O'Connor T, Pirl WF, Rodler E, Rugo HS, Thomas J, Wagner LINCCN Clinical Practice Guidelines Cancer-related fatigue. J Natl Compr Canc Netw. 2010;8(8):904–931.[PubMed][Google Scholar]
  • 87. Bower JEFatigue, brain, behavior, and immunity: summary of the 2012 Named Series on fatigue. Brain Behav Immun. 2012;26(8):1220–1223.[PubMed][Google Scholar]
  • 88. Cleeland CS, Bennett GJ, Dantzer R, Dougherty PM, Dunn AJ, Meyers CA, Miller AH, Payne R, Reuben JM, Wang XS, Lee BNAre the symptoms of cancer and cancer treatment due to a shared biologic mechanism? A cytokine-immunologic model of cancer symptoms. Cancer. 2003;97(11):2919–2925.[PubMed][Google Scholar]
  • 89. Lee BN, Dantzer R, Langley KE, Bennett GJ, Dougherty PM, Dunn AJ, Meyers CA, Miller AH, Payne R, Reuben JM, Wang XS, Cleeland CSA cytokine-based neuroimmunologic mechanism of cancer-related symptoms. Neuroimmunomodulation. 2004;11(5):279–292.[PubMed][Google Scholar]
  • 90. Seruga B, Zhang H, Bernstein LJ, Tannock IFCytokines and their relationship to the symptoms and outcome of cancer. Nat Rev Cancer. 2008;8(11):887–899.[PubMed][Google Scholar]
  • 91. Saligan LN, Kim HSA systematic review of the association between immunogenomic markers and cancer-related fatigue. Brain Behav Immun. 2012;26(6):830–848.[Google Scholar]
  • 92. Ruddle NHLymphotoxin and TNF: how it all began-a tribute to the travelers. Cytokine Growth Factor Rev. 2014;25(2):83–89.[Google Scholar]
  • 93. Upadhyay V, Fu YXLymphotoxin signalling in immune homeostasis and the control of microorganisms. Nat Rev Immunol. 2013;13(4):270–279.[Google Scholar]
  • 94. Upadhyay V, Fu YXLymphotoxin organizes contributions to host defense and metabolic illness from innate lymphoid cells. Cytokine Growth Factor Rev. 2014;25(2):227–233.[Google Scholar]
  • 95. Boraska V, Rayner NW, Groves CJ, Frayling TM, Diakite M, Rockett KA, Kwiatkowski DP, Day-Williams AG, McCarthy MI, Zeggini ELarge-scale association analysis of TNF/LTA gene region polymorphisms in type 2 diabetes. BMC Med Genet. 2010;11:69.[Google Scholar]
  • 96. Ikeda S, Tanaka N, Arai T, Chida K, Muramatsu M, Sawabe MPolymorphisms of LTA, LGALS2, and PSMA6 genes and coronary atherosclerosis: a pathological study of 1503 consecutive autopsy cases. Atherosclerosis. 2012;221(2):458–460.[PubMed][Google Scholar]
  • 97. Kallio KA, Marchesani M, Vlachopoulou E, Mantyla P, Paju S, Buhlin K, Suominen AL, Contreras J, Knuuttila M, Hernandez M, Huumonen S, Nieminen MS, Perola M, Sinisalo J, Lokki ML, Pussinen PJGenetic variation on the BAT1-NFKBIL1-LTA region of major histocompatibility complex class III associates with periodontitis. Infect Immun. 2014;82(5):1939–1948.[Google Scholar]
  • 98. Upadhyay V, Fu YXLinking the microbiota and metabolic disease with lymphotoxin. Int Immunol. 2013;25(7):397–403.[Google Scholar]
  • 99. Lee KA, Gay C, Pullinger CR, Hennessy MD, Zak RS, Aouizerat BECytokine polymorphisms are associated with poor sleep maintenance in adults living with human immunodeficiency virus/acquired immunodeficiency syndrome. Sleep. 2014;37(3):453–463.[Google Scholar]
  • 100. Lee KA, Gay CL, Lerdal A, Pullinger CR, Aouizerat BECytokine polymorphisms are associated with fatigue in adults living with HIV/AIDS. Brain Behav Immun. 2014;40:95–103.[Google Scholar]
  • 101. Bower JE, Greendale G, Crosswell AD, Garet D, Sternlieb B, Ganz PA, Irwin MR, Olmstead R, Arevalo J, Cole SWYoga reduces inflammatory signaling in fatigued breast cancer survivors: a randomized controlled trial. Psychoneuroendocrinology. 2014;43:20–29.[Google Scholar]
  • 102. Zhang XJ, Huang W, Yang S, Sun LD, Zhang FY, Zhu QX, Zhang FR, Zhang C, Du WH, Pu XM, Li H, Xiao FL, Wang ZX, Cui Y, Hao F, Zheng J, Yang XQ, Cheng H, He CD, Liu XM, Xu LM, Zheng HF, Zhang SM, Zhang JZ, Wang HY, Cheng YL, Ji BH, Fang QY, Li YZ, Zhou FS, Han JW, Quan C, Chen B, Liu JL, Li D, Fan L, Zhang AP, Liu SX, Yang CJ, Wang PG, Zhou WM, Lin GS, Wu WD, Fan X, Gao M, Yang BQ, Lu WS, Zhang Z, Zhu KJ, Shen SK, Li M, Zhang XY, Cao TT, Ren W, Zhang X, He J, Tang XF, Lu S, Yang JQ, Zhang L, Wang DN, Yuan F, Yin XY, Huang HJ, Wang HF, Lin XY, Liu JJPsoriasis genome-wide association study identifies susceptibility variants within LCE gene cluster at 1q21. Nat Genet. 2009;41(2):205–210.[PubMed][Google Scholar]
  • 103. Howard Tripp N, Tarn J, Natasari A, Gillespie C, Mitchell S, Hackett KL, Bowman SJ, Price E, Pease CT, Emery P, Lanyon P, Hunter J, Gupta M, Bombardieri M, Sutcliffe N, Pitzalis C, McLaren J, Cooper A, Regan M, Giles I, Isenberg DA, Saravanan V, Coady D, Dasgupta B, McHugh N, Young-Min S, Moots R, Gendi N, Akil M, Griffiths B, Lendrem DW, Ng WFFatigue in primary Sjogren's syndrome is associated with lower levels of proinflammatory cytokines. RMD Open. 2016;2(2):e000282.[Google Scholar]
  • 104. Dantzer R, O'Connor JC, Freund GG, Johnson RW, Kelley KWFrom inflammation to sickness and depression: when the immune system subjugates the brain. Nat Rev Neurosci. 2008;9(1):46–56.[Google Scholar]
  • 105. Jha S, Pan-Yun Ting JHolding the inflammatory system in check: NLRs keep it cool. F1000Prime Rep. 2015;7:15.[Google Scholar]
  • 106. Martinon F, Mayor A, Tschopp JThe inflammasomes: guardians of the body. Annu Rev Immunol. 2009;27(1):229–265.[PubMed][Google Scholar]
  • 107. Latz E, Xiao TS, Stutz AActivation and regulation of the inflammasomes. Nat Rev Immunol. 2013;13(6):397–411.[Google Scholar]
  • 108. Borm ME, van Bodegraven AA, Mulder CJ, Kraal G, Bouma GThe effect of NOD2 activation on TLR2-mediated cytokine responses is dependent on activation dose and NOD2 genotype. Genes Immun. 2008;9(3):274–278.[PubMed][Google Scholar]
  • 109. Kutikhin AGRole of NOD1/CARD4 and NOD2/CARD15 gene polymorphisms in cancer etiology. Hum Immunol. 2011;72(10):955–968.[PubMed][Google Scholar]
  • 110. Glas J, Seiderer J, Tillack C, Pfennig S, Beigel F, Jurgens M, Olszak T, Laubender RP, Weidinger M, Muller-Myhsok B, Goke B, Ochsenkuhn T, Lohse P, Diegelmann J, Czamara D, Brand SThe NOD2 single nucleotide polymorphisms rs2066843 and rs2076756 are novel and common Crohn's disease susceptibility gene variants. PloS one. 2010;5(12):e14466.[Google Scholar]
  • 111. Yao Q, Shen M, McDonald C, Lacbawan F, Moran R, Shen BNOD2-associated autoinflammatory disease: a large cohort study. Rheumatology. 2015;54(10):1904–1912.[PubMed][Google Scholar]
  • 112. Connelly TM, Berg AS, Harris L, 3rd, Brinton D, Deiling S, Koltun WAGenetic determinants associated with early age of diagnosis of IBD. Dis Colon Rectum. 2015;58(3):321–327.[PubMed][Google Scholar]
  • 113. Anand PK, Kanneganti TDNLRP6 in infection and inflammation. Microbes Infect. 2013;15(10–11):661–668.[Google Scholar]
  • 114. Ydens E, Demon D, Lornet G, De Winter V, Timmerman V, Lamkanfi M, Janssens SNlrp6 promotes recovery after peripheral nerve injury independently of inflammasomes. J Neuroinflammation. 2015;12(1):143.[Google Scholar]
  • 115. Sharma D, Kanneganti TDThe cell biology of inflammasomes: Mechanisms of inflammasome activation and regulation. J Cell Biol. 2016;213(6):617–629.[Google Scholar]
  • 116. Wang A, Ling Z, Yang Z, Kiela PR, Wang T, Wang C, Cao L, Geng F, Shen M, Ran X, Su Y, Cheng T, Wang JGut microbial dysbiosis may predict diarrhea and fatigue in patients undergoing pelvic cancer radiotherapy: a pilot study. PloS one. 2015;10(5):e0126312.[Google Scholar]
  • 117. Docherty LE, Rezwan FI, Poole RL, Turner CL, Kivuva E, Maher ER, Smithson SF, Hamilton-Shield JP, Patalan M, Gizewska M, Peregud-Pogorzelski J, Beygo J, Buiting K, Horsthemke B, Soellner L, Begemann M, Eggermann T, Baple E, Mansour S, Temple IK, Mackay DJMutations in NLRP5 are associated with reproductive wastage and multilocus imprinting disorders in humans. Nat Commun. 2015;6:8086.[Google Scholar]
  • 118. Ebersole JL, Kirakodu S, Novak MJ, Exposto CR, Stromberg AJ, Shen S, Orraca L, Gonzalez-Martinez J, Gonzalez OAEffects of aging in the expression of NOD-like receptors and inflammasome-related genes in oral mucosa. Mol Oral Microbiol. 2016;31(1):18–32.[Google Scholar]
  • 119. Poli G, Brancorsini S, Cochetti G, Barillaro F, Egidi MG, Mearini EExpression of inflammasome-related genes in bladder cancer and their association with cytokeratin 20 messenger RNA. Urol Oncol. 2015;33(12):505 e1–507 e1.[PubMed][Google Scholar]
  • 120. Zhu L, Hu Z, Liu J, Gao J, Lin BGene expression profile analysis identifies metastasis and chemoresistance-associated genes in epithelial ovarian carcinoma cells. Med Oncol. 2015;32(1):426.[Google Scholar]
  • 121. Dufner A, Duncan GS, Wakeham A, Elford AR, Hall HT, Ohashi PS, Mak TWCARD6 is interferon inducible but not involved in nucleotide-binding oligomerization domain protein signaling leading to NF-kappaB activation. Mol Cell Biol. 2008;28(5):1541–1552.[Google Scholar]
  • 122. Kim SS, Ahn CH, Kang MR, Kim YR, Kim HS, Yoo NJ, Lee SHExpression of CARD6, an NF-kappaB activator, in gastric, colorectal and oesophageal cancers. Pathology. 2010;42(1):50–53.[PubMed][Google Scholar]
  • 123. Li L, Chen W, Zhu Y, Wang X, Jiang DS, Huang F, Wang L, Xiang F, Qin W, Wang Q, Zhang R, Zhu X, Li H, Chen XCaspase recruitment domain 6 protects against cardiac hypertrophy in response to pressure overload. Hypertension. 2014;64(1):94–102.[PubMed][Google Scholar]
  • 124. Iwata M, Ota KT, Duman RSThe inflammasome: pathways linking psychological stress, depression, and systemic illnesses. Brain Behav Immun. 2013;31:105–114.[Google Scholar]
  • 125. Roxburgh CS, McMillan DCTherapeutics targeting innate immune/inflammatory responses through the interleukin-6/JAK/STAT signal transduction pathway in patients with cancer. Transl Res. 2016;167(1):61–66.[PubMed][Google Scholar]
  • 126. Futosi K, Fodor S, Mocsai ANeutrophil cell surface receptors and their intracellular signal transduction pathways. Int Immunopharmacol. 2013;17(3):638–650.[Google Scholar]
  • 127. Sabio G, Davis RJTNF and MAP kinase signalling pathways. Semin Immunol. 2014;26(3):237–245.[Google Scholar]
  • 128. He Q, Gong Y, Gower L, Yang X, Friesel RESef regulates epithelial-mesenchymal transition in breast cancer cells. J Cell Biochem. 2016;117(10):2346–2356.[Google Scholar]
  • 129. Mellett M, Atzei P, Bergin R, Horgan A, Floss T, Wurst W, Callanan JJ, Moynagh PNOrphan receptor IL-17RD regulates Toll-like receptor signalling via SEFIR/TIR interactions. Nat Commun. 2015;6:6669.[PubMed][Google Scholar]
  • 130. Biswas R, Angshuman BNFkB pathway and inhibition: an overview. Computational Molecular Biology. 2016;6(1):1–20.[PubMed][Google Scholar]
  • 131. Hoesel B, Schmid JAThe complexity of NF-kappaB signaling in inflammation and cancer. Mol Cancer. 2013;12(1):86.[Google Scholar]
  • 132. Lawrence TThe nuclear factor NF-kappaB pathway in inflammation. Cold Spring Harb Perspect Biol. 2009;1(6):a001651.[Google Scholar]
  • 133. Sakoda Y, Nagai T, Murata S, Mizuno Y, Kurosawa H, Shoda H, Morishige N, Yanai R, Sonoda KH, Tamada KPathogenic function of herpesvirus entry mediator in experimental autoimmune uveitis by induction of Th1- and Th17-Type T cell responses. J Immunol. 2016;196(7):2947–2954.[PubMed][Google Scholar]
  • 134. Li D, Fu Z, Chen S, Yuan W, Liu Y, Li L, Pang D, Li DHVEM gene polymorphisms are associated with sporadic breast cancer in Chinese women. PloS one. 2013;8(8):e71040.[Google Scholar]
  • 135. Chen W, Tang WR, Zhang M, Chang K, Wei YLAssociation of DR4 (TRAIL-R1) polymorphisms with cancer risk in Caucasians: an updated meta-analysis. Asian Pac J Cancer Prev. 2014;15(6):2889–2892.[PubMed][Google Scholar]
  • 136. Webb E, Broderick P, Lubbe S, Chandler I, Tomlinson I, Houlston RSA genome-wide scan of 10 000 gene-centric variants and colorectal cancer risk. Eur J Hum Genet. 2009;17(11):1507–1514.[Google Scholar]
  • 137. Lee WH, Wu HH, Huang CKTargeting interleukin-17 receptors. Oncotarget. 2015;6(21):18244–18245.[Google Scholar]
  • 138. Huang CK, Yang CY, Jeng YM, Chen CL, Wu HH, Chang YC, Ma C, Kuo WH, Chang KJ, Shew JY, Lee WHAutocrine/paracrine mechanism of interleukin-17B receptor promotes breast tumorigenesis through NF-kappaB-mediated antiapoptotic pathway. Oncogene. 2014;33(23):2968–2977.[PubMed][Google Scholar]
  • 139. Kim YG, Ihm CG, Lee TW, Lee SH, Jeong KH, Moon JY, Chung JH, Kim SK, Kim YHAssociation of genetic polymorphisms of interleukins with new-onset diabetes after transplantation in renal transplantation. Transplantation. 2012;93(9):900–907.[PubMed][Google Scholar]
  • 140. Xu Y, Wang D, Luo Y, Li W, Shan Y, Tan X, Zhu CBeta amyloid-induced upregulation of death receptor 6 accelerates the toxic effect of N-terminal fragment of amyloid precursor protein. Neurobiology of aging. 2015;36(1):157–168.[PubMed][Google Scholar]
  • 141. Jang SW, Lim SG, Suk K, Lee WHActivation of lymphotoxin-beta receptor enhances the LPS-induced expression of IL-8 through NF-kappaB and IRF-1. Immunol Lett. 2015;165(2):63–69.[PubMed][Google Scholar]
Collaboration tool especially designed for Life Science professionals.Drag-and-drop any entity to your messages.