Skip to main content

This is a preprint.

It has not yet been peer reviewed by a journal.

The National Library of Medicine is running a pilot to include preprints that result from research funded by NIH in PMC and PubMed.

Research Square logoLink to Research Square
[Preprint]. 2025 Feb 10:rs.3.rs-5690506. [Version 1] doi: 10.21203/rs.3.rs-5690506/v1

Race-based differences in serum biomarkers for cancer-associated cachexia in a diverse cohort of patients with pancreatic ductal adenocarcinoma

Jennifer Permuth 1, Margaret Park 2, Evan Davis 3, Solomon Alhassan 4, J Arnoletti 5, Toni Basinski 6, Ashley McKee 7, Mark Bloomston 8, Tiffany Carson 9, Tiago Biachi de Castria 10, Dung-Tsa Chen 11, Elena Cortizas 12, Sylvia Crowder 13, Maria Genilo Delgado 14, Wade Douglas 15, Jason Fleming 16, Pamela Hodul 17, Kevin Huguet 18, Kun Jiang 19, Dae Won Kim 20, John Koomen 21, Anjuli Luthra 22, Mokenge Malafa 23, Anjana Menon 24, Raiza Morales 25, Nipun Merchant 26, Kenneth Meredith 27, Qianxing Mo 28, Manual Molina-Vega 29, Lina Moreno-Urazan 30, Kayode Olumoyin 31, Nathan Parker 32, Jose Pimiento 33, Ghulam Rasool 34, Katarzyna Rejniak 35, Samer Sansil 36, Lauren Sparks 37, Paul Stewart 38, Alexandra Tassielli 39, Jamie Teer 40, Dan Viet Tran 41, Jose Trevino 42, Vic Velanovich 43, Christopher Whelan 44, Daniel Jeong 45, Sarah Judge 46, Andrew Judge 47
PMCID: PMC11844656  PMID: 39989973

Abstract

Pancreatic ductal adenocarcinoma is projected to become the second leading cause of cancer-related deaths by 2040, with the highest disease burden expected amongst Non-Hispanic Blacks. One of the most significant predictors of poor outcomes is the presence of cancer-associated cachexia (CCa). Yet, race- and ethnicity-specific biomarkers for early CCa diagnosis are lacking. Thus, evaluated a panel of candidate biomarkers of CCa in a diverse cohort of pre-treatment serum. Our study shows that GDF-15 was associated with cachexia severity, was superior to standard CCa-associated biomarkers at classifying cachexia, and differentiated between non-cachexia and pre-cachexia status, but only among Hispanic/Latinx and non-Hispanic Whites. Furthermore, high GDF-15 levels at diagnosis were associated with a ~ 2-fold increase in weight loss over the 6 months post-diagnosis. Thus, GDF-15 may be a potential biomarker for pre-cachexia (prior to weight loss) in the White and the Hispanic population, but not Black individuals. These findings underscore the fact that enrollment of minority individuals in clinical trials to evaluate treatments for CCa is of utmost importance.

Introduction

Pancreatic ductal adenocarcinoma (PDAC) is increasing in incidence in the United States (US) and globally and is projected to be the second leading cause of cancer-related deaths in the US by 20401. Up to 80% of PDAC cases develop cancer-associated cachexia (CCa), defined as a multifactorial wasting disorder characterized by loss of appetite, body weight, and/or lean body mass (i.e., skeletal muscle), leading to fatigue, functional impairment, treatment-related toxicities, poor quality of life, and reduced survival 25. Properly detecting CCa in its earliest stage and intervening to maintain muscle mass remain key goals in the care of patients with PDAC. Unfortunately, due to the lack of a well-established definition of CCa and heterogeneity of presentation, diagnosis of CCa typically does not occur until late in the disease course 69.

Blood-based biomarkers of CCa have the potential to aid in earlier diagnosis and in monitoring the effects of therapy. Laboratory values obtained as part of standard of care shown to have prognostic value for patients with CCa include serum C-reactive protein (CRP), albumin, hemoglobin (HgB), white blood cell (WBC) count, CRP:albumin ratio or the Glasgow Prognostic Score, neutrophil to lymphocyte ratio, platelet count, and bilirubin1018. Human studies conducted among patients with PDAC have also identified candidate circulating biomarkers of CCa which include pro-inflammatory cytokines such as interleukin-6 (IL-6), interleukin-8 (IL-8), tumor necrosis factor (TNF)-α, and monocyte chemoattractant protein-1 (MCP-1), transforming growth factor (TGF)-beta family members such as activin A and growth/differentiation factor (GDF)-15, growth factors including insulin-like growth factor binding protein 2 (IGFBP2), adipokines such as leptin and adiponectin, glycoproteins such as zinc-α2-glycoprotein (ZAG), apolipoproteins including apoC-II, apoC-III, and glucagon-like-peptide-1 (GLP-1), branched chain amino acids, ceramides (medium- or long-chain), and miRNAs1926. Sample sizes of PDAC cases in the aforementioned studies of novel blood-based biomarkers ranged from 60–99, with most but not all studies evaluating patients with metastatic disease at the time of blood collection. Because prior associations with blood-based biomarkers were generally made in patients with advanced disease (who oftentimes were undergoing treatment), these associations may be linked to disease progression rather than CCa. Several of these studies were conducted in Asia and may not be racially diverse or generalizable to other racial/ethnic populations18,23,27. Of those conducted in the US21,22,24,28, all primarily comprised of self-reported Non-Hispanic Whites (NHW), with only two of these studies 24,28 including self-reported Non-Hispanic Black (NHB)/African American individuals (n = 7 24 and n = 11 28), and only one study 28 including self-reported Hispanic/Latinx (H/L) individuals (n = 5).

Taken together, it is apparent that investigations of serum biomarkers of CCa in larger and more racially and ethnically diverse cohorts of newly-diagnosed PDAC cases are lacking. Given that PDAC incidence and mortality rates are highest in NHB followed by NHW and H/L29, it is prudent to focus research efforts on including historically underserved populations in studies of CCa. The goal of the current study was thus to investigate levels of candidate serum biomarkers in a diverse cohort of treatment-naïve PDAC cases and determine their association with CCa status, survival, and health related quality of life (HRQoL) overall and by race and ethnicity. Importantly, we assessed the added value of a race- and ethnicity-specific panel of biomarkers in predicting clinical outcomes above and beyond standard criteria obtained at the point of care.

Methods

Study Participants: Study Population:

The Florida Pancreas Collaborative (FPC) is a multi-institutional prospective cohort study and biobanking initiative established to advance PDAC research in racially/ethnically diverse populations 30. Participants in the FPC include 500 individuals with a suspected pancreatic mass presenting to one of the participating study sites. FPC participants meeting the following inclusion criteria were included in this analysis: 1) diagnosed with PDAC; 2) had available pre-treatment blood collections; and 3) were able to have their cachexia status ascertained. A total of 202 participants met these criteria (Figure S1). Importantly, systematic differences were not observed between characteristics of PDAC cases who did and did not donate blood (data not shown) 31,30. This study was approved by the Moffitt Cancer Center Scientific Review Committee (MCC19717, Pro00029598), and Advarra IRB (IRB00000971). All patients provided informed consent for participation30.

Data collection and Clinical Outcomes:

Demographic, clinical, and epidemiologic data was collected from enrolled participants at baseline and at a 6-month follow-up timepoint through: self-administered online- or teleform-based questionnaires; questions administered by the research coordinator at each participating study site; and abstraction from the electronic medical record30. Using available data, participants were first categorized using a schema that classifies the cachexia continuum into 4 stages (non-cachectic (NCa), pre-cachectic (PCa, i.e. those with abnormal labs or appetite who have not lost a significant amount of weight), cachectic (Ca), and refractory cachectic (RCa)) based on clinical, laboratory, functional, and nutritional criteria used by Vigano et al32. We also used a simplified method for CCa classification based on self-reported weight loss (WL) over the previous 6 months, with cases having >5% WL or an underweight body mass index (BMI) plus WL >2% categorized as cachectic, and those with <=5% WL as non-cachectic33). Due to small sample sizes for sub-groups of participants when using the categorization schema by Vigano et al32, the simplified dichotomized categorization of CCa based solely on WL was used for statistical analysis when stratifying by race and ethnicity33. As described previously30,31, HRQoL was assessed for cohort participants using the European Organization for Research and Treatment of Cancer (EORTC) QLQ-C30 instrument34.

Blood collection and processing:

Blood was collected via phlebotomy and processed into serum using previously described methods at baseline/pre-treatment and at follow-up timepoints30 and stored at −80°C. Table S1 lists all candidate biomarkers, the assay performed, and the % of samples falling within the detectable range. Detailed methods for assays performed are found in Supplemental Methods. Mean % coefficient of variation values (CV) (intra-assay variability) for all successful assays are found in Table S2, ranging from 1.16% (ENA-78/CXCL5) to 40.02% (Angiotensin II).

Statistical Analysis:

Associations among categorical variables such as demographic and clinical variables were examined using Pearson chi-squared test or Fisher’s exact test. For continuous variables, differences between two independent groups were examined by two sample t-test or Wilcoxon rank sum test; differences among three or more groups were examined by Analysis of Variance (ANOVA) or Kruskal-Wallis test followed by Tukey’s Honest Standard Differences or Dunn’s test for pairwise comparisons. Benjamini-Hochberg (BH) method was used for correction for multiple comparisons. Overall survival time was calculated in months from the date of diagnosis to the date of last contact or death. Survival functions were estimated by Kaplan-Meier methods and compared by log-rank test. Cox proportional hazards regression (‘survival’ R package) were used to examine associations between survival and multiple variables35. Linear or logistic regression models were used to assess relationships between variables of interest such as serum biomarker levels (predictor variables) and response variables such as HRQoL or cachexia status. Concordance probability estimate analysis was performed using the ‘CPE’ R package36. Receiver operator characteristic (ROC) analysis was performed using the ‘pROC’ R package37.

Results

Characteristics of the study population

Of 500 patients with a suspected pancreatic mass that were recruited to the Florida Pancreas Collaborative study, 318 were confirmed to have a diagnosis of PDAC 30. Select sociodemographic and clinical characteristics of PDAC cases who donated blood pre-treatment (n=206) are shown in Table S3 (stratified by race and ethnicity). The final analytic dataset includes 202 of the 206 PDAC cases with blood for whom cachexia status could be determined using established criteria 32,33, and comprised 132 NHW, 30 NHB, and 40 H/L cases (Table 1, Figure S1). At enrollment, males were significantly more likely than females to be cachectic (p=0.025). Other predictors of CCa included lower BMI (p=0.006), diabetes (p=0.027), and a worse ECOG functional score (p<0.001) (Table 1). The prevalence of cachexia was highest in H/L (67.5%) followed by NHB (63.3%) and NHW (55.2%). As expected, late-stage disease was also significantly associated with poorer survival (P<0.0001, see Figure 1A). However, cancer stage was not associated with cachexia status (Ca vs NCa) via a chi-squared test (P=0.36) or cachexia stage (NCa, PCa, Ca and RCa) via a Cochran-Armitage trend test (p=0.63). Cases classified as Ca (n=121) had significantly worse survival than those classified as NCa (n=81) (log-rank P=0.009, Figure 1B, Table 1, see Figure 1C for survival stratified by cachectic stage as described by Vigano et al32).

Table 1:

Sociodemographic and clinical characteristics of Florida Pancreas Collaborative participants with known cachexia status and baseline blood^ (n=202)*.

NCa (n=81) NCa % Ca (n=121) Ca % p-value
Age at diagnosis, mean (SD) 68.6 (11.1) 69.0 (10.2) 0.826
Sex Female 52 64.20 57 47.11 0.025
Male 29 35.80 64 52.89
Race and Ethnicity Non-Hispanic Black (NHB) 11 13.58 19 15.70 0.442
Hispanic/Latinx (H/L) 13 16.05 27 22.31
Non-Hispanic White (NHW) 57 70.37 75 61.98
Education Post-graduate 19 23.46 10 8.26 0.003
Up to college 29 35.80 31 25.62
Up to high school 9 11.11 28 23.14
Missing 24 29.63 52 42.98
Income $0–$60,000 17 20.99 33 27.27 0.298
$60,000–$100,000 15 18.52 14 11.57
Over $100,000 9 11.11 12 9.92
Missing 40 49.38 62 51.24
Insurance status Insured 52 64.20 69 57.02 0.618
Uninsured 0 0.00 2 1.65
Missing 29 35.80 50 41.32
Marital Status Married 42 51.85 46 38.02 0.138
Unmarried 12 14.81 26 21.49
Missing 27 33.33 49 40.50
Stage I/II 43 53.09 50 41.32 0.210
III/IV 26 32.10 44 36.36
Missing 12 14.81 27 22.31
Grade 1 (well-diff) 3 3.70 2 1.65 0.759
2 (moderately diff) 25 30.86 32 26.45
3 (poorly diff) 14 17.28 19 15.70
4 (undifferentiated) 0 0.00 0 0.00
Missing 39 48.15 68 56.20
BMI Category Underweight 5 6.17 16 13.22 0.021
Normal 20 24.69 42 34.71
Overweight 29 35.80 43 35.54
Obese 25 30.86 18 14.88
Missing 2 2.47 2 1.65
BMI, mean (SD) 27.8 (5.4) 25.7 (5.0) 0.006
Family history of pancreatic cancer No / Unknown 72 88.89 119 98.35 0.817
Yes 9 11.11 2 1.65
Smoking status Never 31 38.27 36 29.75 0.117
Former 26 32.10 37 30.58
Current 2 2.47 11 9.09
Missing 22 27.16 37 30.58
Age-adjusted CCI 0–5 13 10.74 18 22.22 1.000
>5 33 27.27 50 61.73
Missing 35 28.93 53 65.43
Diabetes Yes 20 24.69 46 38.02 0.027
No 35 43.21 53 43.80
Missing 26 32.10 22 18.18
% Weight loss, median (SD) 1.70 (2.8) 11.1 (5.3) <0.001
Waist circumf, median (SD) 36.56 (6.2) 37.80 (15.4) 0.731
Waist-Hip Ratio, median (SD) 0.92 (0.11) 0.92 (0.07) 0.360
ECOG functional score 0 53 65.43 38 31.40 <0.001
1 25 30.86 70 57.85
2 1 1.23 6 4.96
3–4 0 0.00 3 2.48
Missing 2 2.47 4 3.31
Survival in months, median (LCI;UCI) 19.7 (15.1;38.8) 15.1 (13.3;17.8) 0.009

Abbreviations: BMI=Body Mass Index; Ca=Cachectic; CCI= Charlson Comorbidity Index; NCa=Non-cachectic; LCI=Lower confidence interval; UCI=Upper confidence interval; SD=standard deviation; circumf = circumference

*

4 NHW patients with baseline blood (out of 206 total participants) could not be assessed for cachexia status due to missing weight values.

^

Cachexia staging according to Fearon et al (39) with the following modification: skeletal mass index (SMI) was not assessed.

Figure 1. Cachexia status and stage are linked to overall survival in a PDAC cohort with baseline blood available.

Figure 1

Survival was analyzed based on cachexia status using either weight loss alone (A), cachexia status based on weight loss and other blood biomarkers (B) stage (stage 1/2 vs 3/4, C) or R/E (D). Time to event (x-axis) is measured in months.

Compared to NHW, NHB and H/L tended to be diagnosed with PDAC at a younger age and were less likely to have private insurance or to be married. H/L patients in our cohort had a lower overall BMI (p=0.018) than other racial and ethnic groups (Table S3). Although NHB presented more frequently with late-stage disease (50.0%) than NHW and H/L (37.5% and 29.4% respectively), NHB did not have significantly poorer overall survival (OS) when compared to the NHW cohort (p= 0.192 based on a pairwise comparison of NHB vs NHW). H/L in our cohort demonstrated a significantly longer OS time than other groups (p=0.021, Table S3, Figure 1D).

Significant differences were observed between cachectic and non-cachectic PDAC cases overall for multiple serum biomarkers.

Using hierarchical clustering, biomarker clustering was not observed by CCa status (cachectic versus non-cachectic; Figure S2). Among PDAC cases with CCa versus without CCa, serum levels of 16 biomarkers were significantly altered (14 increased, 2 decreased) after adjustment for multiple comparisons, with Padj<0.05 (Table 2). TNF-α, GDF-15, IL-10, IL-22, MIP-1α, IL1-β, TIMP-1, IL-8, MIP-3α, IL-6, HGF, IFN-γ CRP, and MMP-2 were significantly higher in patients classified as having CCa compared with those without CCa, whereas leptin and HbA1c were significantly lower in the CCa group (Figure 2A, Table 2). The two most significantly increased markers were TNF-α (~1.4-fold increase) and GDF-15 (~1.4-fold increase), both with an adjusted p-value of <0.001. Separation of patients based on disease stage further revealed three biomarkers that were significantly elevated in cachectic patients with both early stage (stage I/II) and late stage (stage III/IV) disease: TNF-α (P=0.014 (early) and P=0.001 (late)), GDF-15 (P=0.003 (early) and P=0.018 (late)), and IL-22 (P=0.004 (early) and P=0.005 (late)), see Table S4 and Figure S3A). Among early (but not late) stage PDAC patients, IL-6 (P=0.001), MIP3-α (P=0.002) and IL-8 (P=0.009) were significantly higher in cachectic versus non-cachectic patients (Table S4 and Figure S3B).

Table 2:

Means and standard deviations for all biomarkers successfully tested, stratified by cancercachexia status and sorted by adjusted P-value*

Analyte NCa mean (n=81)^ Ca mean (n=121)^ NCa sd Ca sd p (Wilcoxon) padj (BH)
TNF-α 3.513 3.991 0.492 0.598 <0.001 <0.001
GDF-15 7.207 7.713 0.580 0.716 <0.001 <0.001
IL-10 1.482 2.236 1.107 1.100 <0.001 <0.001
IL-22 1.267 1.905 1.252 1.135 <0.001 <0.001
MIP-1α 5.120 5.485 0.943 0.865 <0.001 <0.001
IL-1B 0.024 0.999 1.098 1.271 <0.001 <0.001
Leptin 11.759 10.786 1.460 1.918 <0.001 0.002
TIMP-1 12.783 13.048 0.452 0.526 <0.001 0.002
IL-8 5.291 5.713 0.610 0.866 0.001 0.002
MIP-3α 4.896 5.481 1.079 1.271 0.001 0.002
IL-6 3.248 3.725 0.892 1.061 0.001 0.003
HbA1c 6.307 6.109 0.425 0.581 0.004 0.016
HGF 6.502 6.687 0.680 0.616 0.005 0.016
IFN-γ 4.330 4.700 0.992 0.864 0.005 0.016
CRP 14.927 15.623 1.611 1.972 0.014 0.038
MMP-2 10.273 10.436 0.565 0.523 0.019 0.050
Albumin 26.314 26.237 0.258 0.223 0.040 0.099
TGF-β2 4.689 4.871 0.798 0.904 0.050 0.116
Glucose 4.519 4.632 0.410 0.363 0.058 0.129
HDL 6.415 6.311 0.536 0.499 0.124 0.245
GRO- α 8.223 8.378 0.749 0.735 0.128 0.245
LDL 10.081 10.198 0.573 0.635 0.128 0.245
CCK 5.841 5.759 0.512 0.557 0.171 0.312
a-SMA 0.744 0.707 0.577 0.676 0.310 0.542
PPAR-γ 1.168 1.253 0.663 0.643 0.325 0.545
C peptide 9.797 9.838 0.747 0.757 0.380 0.614
ENA-78 9.127 9.109 0.811 0.802 0.581 0.875
MCP-1 7.925 7.960 0.370 0.437 0.583 0.875
Adiponectin 16.692 16.687 0.610 0.524 0.612 0.886
Activin A 6.900 6.985 1.396 1.298 0.677 0.908
Lumican 14.512 14.525 0.465 0.459 0.716 0.908
Triglyceride 3.545 3.573 0.546 0.556 0.717 0.908
Fibronectin 18.872 18.909 0.893 0.852 0.724 0.908
TGF- β1 11.020 11.054 0.413 0.434 0.751 0.908
HIF-1α 6.550 6.480 1.200 1.306 0.793 0.908
Insulin 4.076 4.043 1.060 0.891 0.830 0.908
Angiotensin II −0.218 0.023 1.951 2.427 0.833 0.908
G.CSF 4.794 4.864 0.629 0.831 0.842 0.908
Laminin 7.528 7.529 0.390 0.491 0.869 0.908
ZAG 15.379 15.355 0.383 0.457 0.871 0.908
MDC 9.399 9.373 0.338 0.415 0.887 0.908
CA19–9 3.371 3.227 2.158 2.458 0.949 0.949
*

All values are Log2 transformed.

^

Number of samples for analytes for which 100% of samples tested were in range.

Abbreviations: BH=Benjamini-Hochberg; Ca=Cachectic; NCa=Non-cachectic; p=p-value; padj =adjusted p-value; sd=standard deviation.

Figure 2. Cachectic and pre-cachectic PDAC patients demonstrate significant differences in stress-related markers at baseline and GDF-15 can be used to classify patients into cachexia categories:

Figure 2

Boxplots showing a log-scale value of all significantly different serum biomarkers between cachectic and non-cachectic PDAC patients (A) and when stratified into 4 stages as in Vigano et al. (B). NCa=non cachectic, PCa=pre cachectic, Ca=cachectic, RCa=refractory cachectic. All plots in A and B are significant based on a Wilcoxon rank sum or Kruskal-Wallis test and BH-adjusted p-value. Significant differences in serum biomarkers between groups (pairwise Dunn’s test) are denoted with a red line in B. Outliers are denoted by a red dot. C) ROC curves for percent self-reported weight loss (Pct Weightloss), GDF-15, TNF-alpha, white blood cell count (WBC), Hemoglobin and Albumin and a combined TNF-alpha * GDF-15 model (Combined Model) using cachexia status at baseline as the “ground truth”. Youden’s optimal thresholds are denoted with a black dot. D: Boxplot of percent weight loss (negative values indicate weight gain) from baseline to 6-month follow-up time point. Patients were dichotomized based on levels of GDF-15, TNF-alpha or combined GDF-15 * TNF-alpha levels (Low vs High based on the Youden’s threshold values calculated by the ROC in Figure 2C).

Patients who rated their health-related quality of life as 3 or less in the EORTC questionnaire at baseline31 also displayed significantly higher levels of GDF-15 (P<0.001), TNF-α (P=0.007), TIMP-1 (P=0.009), IL-8 (P<0.001), CRP (P=0.005), and IL-6 (P=0.01).

When stratified by sex, both males and females with Ca demonstrated significant increases in TNF-α, MIP-1α, IL-10, IL-22 and GDF-15 compared to NCa. A decrease in leptin was observed in Ca males and females compared to their non-cachectic counterparts (Table S5, Figure S4A). Intriguingly, for several biomarkers, (IL-1b, IL-6, IL-8, TIMP-1, and IFN-g) only females demonstrated significantly increased levels among Ca vs. NCa cases. In contrast, MIP-3α was significantly increased in males with Ca but not females (Figure S4B).

Pre-cachexia stage can be differentiated by GDF-15 and TIMP-1 levels.

When cachexia status was classified using a 4-stage system 32, we find that patients with PCa can be differentiated from NCa patients by CRP, TIMP-1 and GDF-15 (Figure 2B, Table S6). CRP was omitted from further analysis since it was used to stage cachexia32. Other analytes (TNF-α, HGF, IL-10, IL-22) were significantly increased only in patients with Ca or RCa. When TIMP-1 and GDF-15 levels were dichotomized using a median split, both TIMP-1 (P<0.001) and GDF-15 (P<0.001) were significantly increased over worsening cachexia status when subjected to a Cochrane-Armitage trend test. When ordinal regression was performed using cachexia status as the response variable and TIMP-1 and GDF-15 as predictor variables (dichotomized), GDF-15 remains significant as a predictor (P=0.021). Indeed, GDF-15 remains significantly increased over all four cachexia stages using ordinal regression even when adjusting for race and ethnicity, stage, age at diagnosis and BMI (P<0.001, Table 3).

Table 3:

High GDF-15 expression is associated with pancreatic cancer-associated cachexia stage*

Characteristic OR 95% CI p-value (global)
High GDF-15 4.01 2.38, 6.97 <0.001
Race/Ethnicity 0.12
 NHW
 NHB 2.45 0.93, 6.68
 H/L 1.87 0.75, 4.80
Age at diagnosis 0.99 0.96, 1.02 0.5
BMI 0.2
 Normal
 Underweight 2.18 0.65, 7.46
 Overweight 0.81 0.36, 1.81
 Obese 0.58 0.24, 1.39
Stage 0.11
 1/2
 3/4 1.68 0.88, 3.24

Abbreviations: OR=Odds ratio; CI=Confidence interval; NHW=Non-Hispanic white; NHB=Non-Hispanic black; H/L=Hispanic/Latinx; BMI=Body mass index.

*

Patients were separated into two groups based on their GDF-15 levels (via a median split). Ordinal regression was performed using cachexia stage (based on the Vigano et al classification (32)) as the response variable and GDF-15, race/ethnicity, age, BMI or stage as predictor variables.

GDF-15 and TNF-α perform better at cachexia classification than other commonly used classifiers of cachexia.

The area under the curve (AUC), optimal thresholds (using Youden Index), sensitivity and specificity were calculated for all analytes which were significantly different between NCa and Ca patient groups (Table S7). These calculations were also performed for other common blood-based biomarkers associated with cachexia status (WBC count, albumin, hemoglobin) 32 and for body composition metrics (BMI, waist circumference and waist to hip ratio). Percent WL was used for classification as a (positive) control. Both TNF-α and GDF-15 had AUC values of >0.7 and these analytes were thus used to generate a combined model by multiplying the two concentrations together (Figure 2C). The combined model did have significantly higher AUC than GDF-15 alone (P=0.009) but not better than TNF-α alone (P=0.76); AUCs for TNF-α and GDF-15 were not significantly different (P=0.730). Additional markers did not improve classification (data not shown).

When WL was calculated over time (i.e., from baseline to a 6-month follow-up timepoint), patients with high GDF-15 levels at baseline (based on the optimal ROC threshold) lost a mean of 8.0 pounds and patients with low GDF-15 lost a mean of 4.4 pounds (P=0.05, Figure 2D). TNF-α levels and combined levels of GDF-15 and TNF-α (combined model) did not significantly predict weight loss over the same time period (P=0.17 and P=0.06 for TNF-α and combined values respectively).

Stratification by race and ethnicity (R&E) reveals novel biomarkers of CCa for NHW and H/L participants.

Of the serum biomarkers significantly deregulated in Ca versus NCa PDAC cases overall (Table 2), several continued to show differences when stratified by R&E. For example, GDF-15, IL-22, MIP-1α, TIMP-1 and TNF-α levels were significantly higher among H/L and NHW PDAC cases categorized as having CCa than those without CCa (Figure 3, Table S8). Although statistically significant differences were not observed for any of these biomarkers when comparing NHB with and without CCa (Figure 3, Table S8), TNF-α and IL-22 levels did trend upwards for NHB cachectic patients, similar to the other R&E groups (Figure 3). In contrast, although GDF-15 was significantly higher in the cachexia group overall and for H/L and NHW subgroups, this biomarker did not show any difference in cachectic NHB compared to non-cachectic NHB PDAC patients (padj=0.95).

Figure 3. Both NHW and H/L participants demonstrate significant differences between non cachexia and cachexia status in stress-related markers at baseline:

Figure 3

Boxplots showing a log-scale value of serum biomarkers which are significantly different between cachectic and non-cachectic PDAC patients for at least two R/Es stratified into Non-Hispanic Black, Hispanic/Latinx and Non-Hispanic White patients. NHB=Non-Hispanic Black, H/L=Hispanic/Latinx, NHW=Non-Hispanic White, NCa=non cachectic, Ca=cachectic. Significant differences in serum biomarkers between groups are denoted with a red line. Outliers are denoted by a red dot.

Racial and ethnic variation is apparent for neutrophil- and stress-linked cytokine biomarkers ENA-78/CXCL5 and GRO-α/CXCL1, independent of CCa or pancreas tumor type.

We also assessed racial and ethnic differences at baseline for the panel of biomarkers studied. A total of 15 biomarkers had significantly different expression levels when comparing NHB, H/L, and NHW PDAC cases in the cohort (n=206) (Table S9, Figure S5). 3 of the 13 biomarkers were significantly elevated with a p<0.001 in NHB compared to both NHW and H/L cases and included GRO-α/CXCL1 (P<0.001) and ENA-78/CXCL5 (P<0.001) (Table S9, Figure 4A).

Figure 4. PDAC and other patients demonstrate wide racial variation in select serum biomarkers at baseline:

Figure 4

A) Boxplots showing a log-scale value of select significant biomarkers when PDAC R&E groups are compared (ENA-78/CXCL5, and GRO-α/CXCL1, IL-10 and TNF-α; GDF-15 was added as a control) and B) when stratified into cachexia groups (NHB=Non-Hispanic Black, HL=Hispanic/Latino, NHW=Non-Hispanic White, NCa=non cachexia, Ca=Cachexia). C) Boxplots showing ENA-78 and GRO-alpha levels after filtering the full FPC cohort to non-PDAC diagnoses. D) Boxplot of neutrophil to lymphocyte ratio (NLR) for PDAC patients in the study. Significant differences between relevant groups are denoted with a red line. Significance between NHBCa/NHBNCa and all other groups is denoted with a *. N/S = not significant. Outliers are denoted by a red dot.

Other analytes such as IL-10 (P=0.002) and TNF-α (P=0.002) showed significantly lower levels in NHW compared to other R&E groups (Table S9, Figure 4A). The TNF-α/IL-10 ratio38 was increased in NHW participants compared to NHB (P=0.02) or H/L (P=0.04) but was not significantly changed for R&E and cachexia status stratification. Other circulating biomarkers that were significantly decreased in NHW compared to the other R&E groups included IL-1β, TNF-α, and TGF-β2.

Further stratification by cachexia status for ENA-78/CXCL5, GRO-α /CXCL1 and GDF-15 (as a comparison) shows that ENA-78/CXCL5 and GRO-α/CXCL1 levels were unchanged for any R&E group when stratified by cachexia status (Figure 4B). When we performed a linear model with either ENA-78/CXCL5 or GRO-α as response variables and R&E, cachexia status, stage, sex, and BMI as predictor variables, NHB demonstrated significantly increased ENA-78/CXCL5 and GRO-α/CXCL1 compared to other R&E groups (Table S10). Furthermore, when ENA-78/CXCL5 and GRO-α/CXCL1 levels were compared by R&E in other diagnoses from the FPC cohort (including pancreatic neuroendocrine tumors (PNET), intraductal papillary mucinous neoplasms (IPMN), and pancreatitis), NHB individuals continued to demonstrate significantly elevated baseline levels of these biomarkers compared to other R&E groups (Figure 4C). IL-10 and TNF-α were not significantly changed when non-PDAC individuals were stratified by R&E.

Since both GRO-α/CXCL1 and ENA-78/CXCL5 are linked to neutrophil function, the serum neutrophil to lymphocyte ratio (NLR) was calculated using abstracted clinical data and plotted by R&E. We find that NHB PDAC patients demonstrated a small but significant decrease in NLR when compared to H/L and NHW (P<0.001 for both pairwise comparisons) (Figure 4D). Our data indicate that both higher levels in total lymphocyte count and lower levels in neutrophil count in NHB participants are responsible for this measure.

Survival is predicted by inflammatory and proliferative cytokine levels.

A univariate Cox proportional hazard analysis indicates that, among all biomarkers which were significantly different in Ca vs NCa PDAC patients, GRO-α, CRP, TNF-α, GDF-15, HGF, IL-10, IL-6, IL-8 and TIMP-1 are all independent predictors of OS in the full cohort as continuous variables (Table S11). However, after dichotomizing these analytes via a median split and controlling for BMI, R&E, stage, cachexia status, sex, and age at diagnosis (Figure 5A), a modified model including GDF-15, IL-6 and stage as significant predictors of survival was reached (Figure 5B). Furthermore, when dichotomized based on a median split, high GDF-15 and high IL-6 predict shorter survival times via the Kaplan-Meier estimation (Figure 5CD). To assess added value of GDF-15 for predicting overall survival, a Gonen and Heller concordance probability estimate (CPE)36,39 analysis was performed on Coxph models using GDF-15 alone (CPE=0.600, SE=0.022), IL-6 alone (CPE=0.575, SE=0.019), IL-6 and GDF-15 (CPE=0.608, SE=0.021) and GDF-15, IL-6 and WL (CPE=0.617, SE=0.022), all as continuous variables. All differences in calculated CPEs are thus within the standard error. A source file of all analyte levels, neutrophil and lymphocyte counts, longitudinal weight loss, cachexia status, diagnosis and survival data for all 275 patients with baseline blood available for analysis (for patients with any diagnosis) may be found in Table S12.

Figure 5. GDF-15 and IL-6 are linked to overall survival both CoxPH models and by Kaplan estimation:

Figure 5

A-B) Forest plots of dichotomized analytes found to be associates with survival as continuous variables in a univariate analysis when controlled for demographic and patho-clinical variables (A). B represents the final model. NHB=Non-Hispanic Black, HL=Hispanic/Latinx, NHW=Non-Hispanic White, NCa=non cachectic, Ca=Cachectic. C-D) Median levels of GDF-15 (C) or IL-6 (D) were calculated and participants were classed into either low (Low) or high (High) groups based on a median split. C) and D) represent Kaplan-Meier survival curves (time in months) for GDF-15 and IL-6, respectively. Time = time to event in months.

Discussion

The FPC cohort represents one of the most racially and ethnically diverse group of PDAC patients with CCa status and available serum biomarker data analyzed to date 1825,4044, and we find that racial disparities in outcomes are in general agreement with published outcomes from others by race or ethnicity45,46.

In the current investigation, growth differentiation factor-15 (GDF-15), also known as macrophage inhibitory cytokine 1 (MIC1), was identified as an important marker of pre-cachexia which continues to increase through the cachexia continuum, and which predicts survival among male and female H/L and NHW participants. GDF-15 is a pleiotropic molecule involved in the pathophysiology of cancers, cardiac disease, COVID-19, hyperemesis gravidarium and is tightly linked to stress pathways4751. It is known as a key biomarker of stress, dysfunctional metabolism and energy production and has emerged as an attractive therapeutic target for obesity/metabolic disorders, cachexia, and immunotherapy48,52,53. Anti-GDF-15 monoclonal antibody treatment is both well tolerated and may reverse anti-PD-L1 resistance in clinical trials54 thus this treatment modality may be useful in contexts other than CCa. In contrast to others26, we do not observe sexual dimorphism in cachexia-related GDF-15 upregulation55. Importantly, this molecule is found to be upregulated in the circulatory system of both early (I/II) and late (III/IV) stage patients, suggesting that the increase in GDF-15 is not secondary to cancer progression. Furthermore, our findings strongly suggest that this molecule should not be used as a marker for cachexia in NHB PDAC patients due to the possibility of false negatives. Importantly, GDF-15 was one of only two markers which was predictive of “pre-cachexia” status and this molecule predicted survival independently of weight loss based on both a Kaplan Meier estimate and CPE analysis. Early identification and intervention for cancer-related cachexia is key to alleviating the negative outcomes associated with this condition5,21,26,32,56,57. Ponsegromab (a monoclonal GDF-15 antibody) has shown promise in clinical trials53 but this trial importantly did not include any NHB individuals. Thus, these findings underscore the importance of minority inclusion in clinical trials but are valuable from an interventional standpoint for H/L and NHW populations.

One intriguing finding which emerged from this study is that, although TNF-α and IL-22 did not reach significance for the smaller NHB group in our cohort, the means did trend upwards for these two markers in cachectic NHB patients, in stark contrast to GDF-15 and TIMP-1. Thus, these molecules may warrant further investigation as racially agnostic indicators of metabolic stress. TNF-α may be of special interest, as this molecule has long been studied as a metabolic stress and inflammatory cytokine58,59. Our data demonstrate that NHB and H/L participants have slightly higher circulating TNF-α levels than NHW in line with previously published literature and secondary to increased chronic or acute social stressors minority groups experience that increase inflammation60,61. Higher baseline levels of TNF-α in minorities may thus serve to blunt increases in this cytokine related to cachectic stress. Nevertheless, with a larger sample size, increases in this cytokine may become apparent for NHB individuals with cachexia.

The role of GDF-15 and race is understudied. However, some studies include that of Rybicki et al62, who observed lower GDF-15 expression in male NHB participants in a benign biopsy prostate cancer risk study. Contrary to these findings, a study in 2012 identified Black race as being predictive of higher plasma GDF-15 levels in a study of atherosclerosis63. Hence, this work represents the first time that GDF-15 has been studied in the context of a response to metabolic stress (cachexia) and race. Taken together, these findings suggest that monitoring of weight/weight loss is of paramount importance in the diagnosis of CCa in NHB patients, as biomarkers of CCa for this population remain elusive.

Regarding our stratification based on sex, in general agreement with other literature55, we observe sexual dimorphism in circulating analytes for cachexia. Thus, these findings clearly demonstrate the need to include sex as a biological variable in studies of CCa.

Another molecule we found to be predictive of PCa, Ca, and RCa is the tissue inhibitor of metalloproteinases, TIMP-164. TIMP-1 is a pro-inflammatory cytokine associated with and proliferative processes64. Although TIMP-1 is not known to be associated with GDF-15 activity in cancer, TIMP-1 and GDF-15 were weakly correlated in a study of cardiovascular disease in aging patients, suggesting a prognostic use for these markers in a fingerprint65 to better predict cachexia status.

In contrast to others21, we did not observe increases in circulating monocyte chemoattractant protein 1 (MCP-1) in cachectic patients either in the full cohort or when we restricted our analysis to early stage PDAC. However, we did not stratify our cachectic cohort to those who lost > 10% of their body weight as in21. MCP-1 was a successful assay in our hands, and thus we are unable to explain this finding as a technical issue. However, we did observe that this analyte was decreased in minority PDAC patients at baseline (Figure S5) which may have masked CCa-induced increases observed in our diverse cohort. In general agreement with other studies40,51,66,67 however, we do observe increases in circulating GDF-15, TNF-α, IL-6 and decreases in leptin in cachectic PDAC patients.

Other main findings are the distinct racial differences in the TNF-α/IL-10 inflammatory/anti-inflammatory signaling axis67 and the aging/inflammatory biomarkers ENA-78/CXCL5 and GRO-α/CXCL120,40,68. ENA-78/CXCL5 and GRO-α/CXCL1 are of particular interest, as they are upregulated in NHB patients independent of CCa status and pancreas tumor type. ENA-78/CXCL5 is a member of the CXC chemokine family and is involved in neutrophil signaling and recruitment69. Similarly, the chemokine CXCL1 is an angiogenic inflammatory marker which has been linked to neutrophil recruitment and migration of adipose stromal cells into the tumor microenvironment70. ENA-78/CXCL5 and GRO-α/CXCL1 have been shown to have increased expression in tumors of patients with malignant versus non-malignant pancreas tumors, suggesting a role for these genes in pancreatic carcinogenesis. ENA-78/CXCL5 has been shown to promote pancreatic cancer cell growth, migration, and invasion, and to predict poor prognosis. Other findings indicate that GRO-α/CXCL1 and ENA-78/CXCL5 are associated with stress due to social disruption, adult adversity and/or psychological comorbidity in chronic pain disorders71,72. Moreover, these signaling molecules have been associated with PDAC immune evasion73,74, lending credence to a racially unbalanced CXCL1/5-signaling “tumor-friendly” environment. We find that a higher circulating CXCL1/ENA-78/CXCL5 signature is associated with the NHB race, but not with higher circulating neutrophils in NHB patients in agreement with published literature75, suggesting that this minority group may be desensitized to cytokine-mediated neutrophil mobilization or signaling. They may also suggest that NHB have increased formation of neutrophil extracellular traps either during cancer initiation or progression. The current literature indicates that both incidence and mortality for PDAC are increased in NHB patients compared to Whites in the US76, thus the current investigation may shed light on this disparity.

Interestingly, the cytokine in our panel with the strongest association with overall survival was IL-6. Consistent with other reports, IL-6 was found to be increased only in refractory cachectic patients in our cohort and was strongly associated with decreased survival66,77. IL-6 remained a significant predictor of overall survival in our large study, even when controlled for multiple covariates, suggesting a renewed interest in this prognostic marker may be warranted77. Indeed, for non-NHB patients, our findings suggest that GDF-15 is a marker for early cachexia and that IL-6 increases at a later stage in the cachectic continuum (i.e. when the patient has refractory disease). Thus, it is possible that a subset of patients with pre-cachexia and elevated GDF-15 do not go on to develop cachexia. Such patients could have significantly better outcomes than those with elevated IL-6 and refractory cachexia. Collectively, our findings suggest that GDF-15, while less predictive of survival outcomes, has utility as an early marker of metabolic stress. Thus, there may be a clinical benefit to evaluating GDF-15 levels as soon as possible after a probable PDAC diagnosis is made to alert clinicians of potential future weight loss and supportive care needs necessitating intervention (Fig. 6, created in biorender.com).

Figure 6.

Figure 6

Overview of IL-6 and GDF-15 levels throughout the cancer-associated cachexia continuum.

Overall, our findings suggest that, whereas non-Hispanic Whites and Hispanic/Latinx elevate circulatory GDF-15 levels in response to cachexia/metabolic stress, NHB do not. Furthermore, there may be an NHB-specific chronic and non-PDAC-influenced ENA-78/CXCL5/GRO-α/CXCL1 inflammatory phenotype for this minority, possibly due to social stress or allostatic load. Indeed, future studies by this group will investigate a possible link between adipose tissue deposition, cytokine signaling, race and PDAC progression.

Our cohort of patients was recruited not only from academic centers, but also from community hospitals in the area30,45. As such, this cohort represents a more “real-world” scenario regarding choice of therapy, outcomes and racial and ethnic diversity than many previous cohorts. Nevertheless, a limitation of the study is that none of the participants were “normal” healthy controls because all participants presented with a suspicion of a pancreatic mass. In addition, we note a wide range of mean %CV for our assays, suggesting that some biomarkers may be less amenable to assessment using antibody-based techniques. Nevertheless, all reliably dysregulated biomarkers chosen for further evaluation from our panel had an intra assay variability of < 20%. Finally, although our cohort was more diverse than others including clinical trials of cachexia therapies, there are only 30 NHB PDAC patients with baseline blood available in our cohort. Hence, trials aimed at specifically recruiting minority populations are crucial to fully understanding the nature of racial disparities in CCa and PDAC.

Conclusions

GDF-15 has emerged as a promising cachexia biomarker and is specifically increased prior to weight loss for non-NHB PDAC patients. Furthermore, although GDF-15 did not associate with cachexia status/weight loss in NHB patients, it was associated with survival even when controlled for R&E. Future work should endeavor to ensure that minority participants are well represented in PDAC cohorts so that the true biomarker landscape is evaluated from diverse populations. Furthermore, these studies suggest that sex should be included in any cachexia biomarker evaluation.

We also conclude that the inflammatory cytokine TNF-α warrants further investigation in a larger population of minority PDAC patients as the lack of increase in CCa-related TNF-α may be secondary to higher circulating levels of this cytokine in minority non-PDAC participants.

In addition to our GDF-15 findings, we report that the serum cytokine landscape for NHBs is decidedly neutrophil-linked and pro-inflammatory compared to either other minorities (H/L) or NHWs, but that circulating neutrophils are lower in the NHBs. Caution should be taken in interpreting these findings, however, to avoid over-simplifying the complex interplay between genetic background, social stressors, R&E and the metabolic stress response resulting from cancer-associated WL.

Acknowledgments

This work was supported in part by the James and Esther King Biomedical Research Program, Florida Department of Health (Grants #8JK02 awarded to JBP and JGT; #24K03 awarded to J.B.P. and A.R.J.), the Department of Defense Health Program Congressionally Directed Medical Research Program (W81XWH-22-1-1021 LOG#PA210192; awarded to JBP, JBF, and MM), the Tissue Core Facility, the Cancer Pharmacokinetics and Pharmacodynamics Core (PK/PD) Core Facility, and the Biostatistics and Bioinformatics Shared Resource at the H. Lee Moffitt Cancer Center & Research Institute, an NCI designated Comprehensive Cancer Center (P30-CA076292). Research reported in this publication was performed in part at the Biospecimen Shared Resource (BSSR) of the Sylvester Comprehensive Cancer Center at the University of Miami, RRID: SCR_022889 which is also supported by the National Cancer Institute (NCI) of the National Institutes of Health (NIH) under award number P30CA240139. The content is solely the responsibility of the authors and does not necessarily represent the official views of the sponsors or the participating institutions.

Footnotes

Conflicts of Interest

J.M.P is a consultant for ASTELLAS, J&J Worldwide, Ferranova, and ADVOCARE. All other authors noted that they have no conflicts or perceived conflicts of interest related to this work.

Contributor Information

Jennifer Permuth, H. Lee Moffitt Cancer Center and Research Institute.

Margaret Park, H. Lee Moffitt Cancer Center and Research Institute.

Evan Davis, H. Lee Moffitt Cancer Center and Research Institute.

Solomon Alhassan, H. Lee Moffitt Cancer Center and Research Institute.

J. Arnoletti, Orlando Health Cancer Institute

Toni Basinski, H. Lee Moffitt Cancer Center and Research Institute.

Ashley McKee, H. Lee Moffitt Cancer Center and Research Institute.

Mark Bloomston, Ohio State University.

Tiffany Carson, H. Lee Moffitt Cancer Center and Research Institute.

Tiago Biachi de Castria, H. Lee Moffitt Cancer Center and Research Institute.

Dung-Tsa Chen, Moffitt Cancer Center.

Elena Cortizas, University of Miami.

Sylvia Crowder, H. Lee Moffitt Cancer Center and Research Institute.

Maria Genilo Delgado, H. Lee Moffitt Cancer Center and Research Institute.

Wade Douglas, Florida State University College of Medicine.

Jason Fleming, University of Texas Southwestern Medical Center.

Pamela Hodul, H. Lee Moffitt Cancer Center and Research Institute.

Kevin Huguet, St. Anthony’s Hospital.

Kun Jiang, Moffitt Cancer Center.

Dae Won Kim, H. Lee Moffitt Cancer Center and Research Institute.

John Koomen, Moffitt Cancer Center.

Anjuli Luthra, H. Lee Moffitt Cancer Center and Research Institute.

Mokenge Malafa, H. Lee Moffitt Cancer Center and Research Institute.

Anjana Menon, H. Lee Moffitt Cancer Center and Research Institute.

Raiza Morales, H. Lee Moffitt Cancer Center and Research Institute.

Nipun Merchant, University of Miami.

Kenneth Meredith, Sarasota Memorial Hospital.

Qianxing Mo, H. Lee Moffitt Cancer Center and Research Institute.

Manual Molina-Vega, Lakeland Heath Regional Hospital.

Lina Moreno-Urazan, H. Lee Moffitt Cancer Center and Research Institute.

Kayode Olumoyin, H. Lee Moffitt Cancer Center and Research Institute.

Nathan Parker, Moffitt Cancer Center.

Jose Pimiento, H. Lee Moffitt Cancer Center and Research Institute.

Ghulam Rasool, H. Lee Moffitt Cancer Center and Research Institute.

Katarzyna Rejniak, H. Lee Moffitt Cancer Center and Research Institute.

Samer Sansil, H. Lee Moffitt Cancer Center and Research Institute.

Lauren Sparks, Translational Research Institute, AdventHealth.

Paul Stewart, Moffitt Cancer Center.

Alexandra Tassielli, H. Lee Moffitt Cancer Center & Research Institute.

Jamie Teer, Department of Biostatistics and Bioinformatics, Moffitt Cancer Center.

Dan Viet Tran, University of Miami Miller School of Medicine.

Jose Trevino, Virginia Commonwealth University.

Vic Velanovich, University of South Florida.

Christopher Whelan, Moffitt Cancer Center.

Daniel Jeong, Department of Radiology, Moffitt Cancer Center.

Sarah Judge, Department of Physical Therapy, College of Public Health and Health Professions, University of Florida.

Andrew Judge, Department of Physical Therapy, College of Public Health and Health Professions, University of Florida.

References

  • 1.Rahib L., Wehner M.R., Matrisian L.M. & Nead K.T. Estimated Projection of US Cancer Incidence and Death to 2040. JAMA Netw Open 4, e214708 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Poulia K.A. et al. Pancreatic Cancer and Cachexia-Metabolic Mechanisms and Novel Insights. Nutrients 12(2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Kordes M., Larsson L., Engstrand L. & Lohr J.M. Pancreatic cancer cachexia: three dimensions of a complex syndrome. Br J Cancer 124, 1623–1636 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Takeda T. et al. The impact of cachexia and sarcopenia in elderly pancreatic cancer patients receiving palliative chemotherapy. Int J Clin Oncol 26, 1293–1303 (2021). [DOI] [PubMed] [Google Scholar]
  • 5.Fearon K., Arends J. & Baracos V. Understanding the mechanisms and treatment options in cancer cachexia. Nat Rev Clin Oncol 10, 90–9 (2013). [DOI] [PubMed] [Google Scholar]
  • 6.O’Connell T.M. et al. Metabolic Biomarkers for the Early Detection of Cancer Cachexia. Front Cell Dev Biol 9, 720096 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Chen X.Y. et al. A comparison of four common malnutrition risk screening tools for detecting cachexia in patients with curable gastric cancer. Nutrition 70, 110498 (2020). [DOI] [PubMed] [Google Scholar]
  • 8.Penna F. et al. Autophagy Exacerbates Muscle Wasting in Cancer Cachexia and Impairs Mitochondrial Function. J Mol Biol 431, 2674–2686 (2019). [DOI] [PubMed] [Google Scholar]
  • 9.Han J., Meng Q., Shen L. & Wu G. Interleukin-6 induces fat loss in cancer cachexia by promoting white adipose tissue lipolysis and browning. Lipids Health Dis 17, 14 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.McMillan D.C. The systemic inflammation-based Glasgow Prognostic Score: a decade of experience in patients with cancer. Cancer Treat Rev 39, 534–40 (2013). [DOI] [PubMed] [Google Scholar]
  • 11.Pointer D.T. Jr. et al. Neutrophil to lymphocyte ratio, not platelet to lymphocyte or lymphocyte to monocyte ratio, is predictive of patient survival after resection of early-stage pancreatic ductal adenocarcinoma. BMC Cancer 20, 750 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Nakamura Y. et al. Neutrophil/lymphocyte ratio has a prognostic value for patients with terminal cancer. World J Surg Oncol 14, 148 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Menter D.G. et al. Platelet “first responders” in wound response, cancer, and metastasis. Cancer Metastasis Rev 36, 199–213 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Liu Y. et al. Association between Platelet Count with 1-year Survival in Patients with Cancer Cachexia. J Cancer 12, 7436–7444 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Klocker E.V. et al. Decreased Activity of Circulating Butyrylcholinesterase in Blood Is an Independent Prognostic Marker in Pancreatic Cancer Patients. Cancers (Basel) 12(2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Koh H.H. et al. Association of Albumin-Bilirubin Grade and Myosteatosis with its Prognostic Significance for Patients with Colorectal Cancer. Ann Surg Oncol 29, 3868–3876 (2022). [DOI] [PubMed] [Google Scholar]
  • 17.Li X.R. et al. Association of serum total bilirubin with survival outcomes in patients with cancer cachexia: A prospective, multicenter cohort study. Nutrition 102, 111711 (2022). [DOI] [PubMed] [Google Scholar]
  • 18.Sato K. et al. Pentraxin 3 is an adipose tissue-related serum marker for pancreatic cancer cachexia predicting subsequent muscle mass and visceral fat loss. Cancer Sci 113, 4311–4326 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Tsai V.W., Brown D.A. & Breit S.N. Targeting the divergent TGFbeta superfamily cytokine MIC1/GDF15 for therapy of anorexia/cachexia syndromes. Curr Opin Support Palliat Care 12, 404–409 (2018). [DOI] [PubMed] [Google Scholar]
  • 20.Paval D.R. et al. A systematic review examining the relationship between cytokines and cachexia in incurable cancer. J Cachexia Sarcopenia Muscle 13, 824–838 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Talbert E.E. et al. Circulating monocyte chemoattractant protein-1 (MCP-1) is associated with cachexia in treatment-naive pancreatic cancer patients. J Cachexia Sarcopenia Muscle 9, 358–368 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Lewis H.L. et al. Perioperative cytokine levels portend early death after pancreatectomy for ductal adenocarcinoma. J Surg Oncol 117, 1260–1266 (2018). [DOI] [PubMed] [Google Scholar]
  • 23.Dong J. et al. Serum insulin-like growth factor binding protein 2 levels as biomarker for pancreatic ductal adenocarcinoma-associated malnutrition and muscle wasting. J Cachexia Sarcopenia Muscle 12, 704–716 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Babic A. et al. Postdiagnosis Loss of Skeletal Muscle, but Not Adipose Tissue, Is Associated with Shorter Survival of Patients with Advanced Pancreatic Cancer. Cancer Epidemiol Biomarkers Prev 28, 2062–2069 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Cao Z., Zhao K., Jose I., Hoogenraad N.J. & Osellame L.D. Biomarkers for Cancer Cachexia: A Mini Review. Int J Mol Sci 22(2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Chakedis J.M. et al. Identification of circulating plasma ceramides as a potential sexually dimorphic biomarker of pancreatic cancer-induced cachexia. JCSM Rapid Commun 5, 254–265 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Suzuki R. et al. Diagnostic yield of endoscopic retrograde cholangiopancreatography-based cytology for distinguishing malignant and benign intraductal papillary mucinous neoplasm: systematic review and meta-analysis. Dig Endosc 26, 586–93 (2014). [DOI] [PubMed] [Google Scholar]
  • 28.Fogelman D.R. et al. A predictive model of inflammatory markers and patient-reported symptoms for cachexia in newly diagnosed pancreatic cancer patients. Support Care Cancer 25, 1809–1817 (2017). [DOI] [PubMed] [Google Scholar]
  • 29.Siegel R.L., Miller K.D., Wagle N.S. & Jemal A. Cancer statistics, 2023. CA Cancer J Clin 73, 17–48 (2023). [DOI] [PubMed] [Google Scholar]
  • 30.Permuth J.B. et al. The Florida Pancreas Collaborative Next-Generation Biobank: Infrastructure to Reduce Disparities and Improve Survival for a Diverse Cohort of Patients with Pancreatic Cancer. Cancers (Basel) 13(2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Permuth J.B. et al. Leveraging real-world data to predict cancer cachexia stage, quality of life, and survival in a racially and ethnically diverse multi-institutional cohort of treatment-naïve patients with pancreatic ductal adenocarcinoma. Frontiers in Oncology 14(2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Vigano A.A.L. et al. Use of routinely available clinical, nutritional, and functional criteria to classify cachexia in advanced cancer patients. Clin Nutr 36, 1378–1390 (2017). [DOI] [PubMed] [Google Scholar]
  • 33.Fearon K. et al. Definition and classification of cancer cachexia: an international consensus. Lancet Oncol 12, 489–95 (2011). [DOI] [PubMed] [Google Scholar]
  • 34.Fitzsimmons D. et al. Development of a disease specific quality of life (QoL) questionnaire module to supplement the EORTC core cancer QoL questionnaire, the QLQ-C30 in patients with pancreatic cancer. EORTC Study Group on Quality of Life. Eur J Cancer 35, 939–41 (1999). [DOI] [PubMed] [Google Scholar]
  • 35.Li J.C.A. Modeling survival data: Extending the Cox model. Sociological Methods & Research 32, 117–120 (2003). [Google Scholar]
  • 36.Heller G. & Mo Q. Estimating the concordance probability in a survival analysis with a discrete number of risk groups. Lifetime Data Anal 22, 263–79 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Robin X. et al. pROC: an open-source package for R and S+ to analyze and compare ROC curves. BMC Bioinformatics 12, 77 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Aroucha D.C. et al. High tumor necrosis factor-alpha/interleukin-10 ratio is associated with hepatocellular carcinoma in patients with chronic hepatitis C. Cytokine 62, 421–5 (2013). [DOI] [PubMed] [Google Scholar]
  • 39.Devlin S.M. & Heller G. Concordance probability as a meaningful contrast across disparate survival times. Stat Methods Med Res 30, 816–825 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Fong Y. et al. Cachectin/TNF or IL-1 alpha induces cachexia with redistribution of body proteins. Am J Physiol 256, R659–65 (1989). [DOI] [PubMed] [Google Scholar]
  • 41.Felix K. et al. Identification of serum proteins involved in pancreatic cancer cachexia. Life Sci 88, 218–25 (2011). [DOI] [PubMed] [Google Scholar]
  • 42.Sutandyo N. The role of microRNA in cancer cachexia and muscle wasting: A review article. Caspian J Intern Med 12, 124–128 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Loumaye A. & Thissen J.P. Biomarkers of cancer cachexia. Clin Biochem 50, 1281–1288 (2017). [DOI] [PubMed] [Google Scholar]
  • 44.Li X., Du L., Liu Q. & Lu Z. MicroRNAs: Novel players in the diagnosis and treatment of cancer cachexia (Review). Exp Ther Med 24, 446 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Permuth J.B. et al. Racial and ethnic disparities in a state-wide registry of patients with pancreatic cancer and an exploratory investigation of cancer cachexia as a contributor to observed inequities. Cancer Med 8, 3314–3324 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Riner A.N. et al. Disparities in Pancreatic Ductal Adenocarcinoma-The Significance of Hispanic Ethnicity, Subgroup Analysis, and Treatment Facility on Clinical Outcomes. Cancer Med 9, 4069–4082 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Alserawan L. et al. Growth Differentiation Factor 15 (GDF-15): A Novel Biomarker Associated with Poorer Respiratory Function in COVID-19. Diagnostics (Basel) 11(2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Haake M. et al. Tumor-derived GDF-15 blocks LFA-1 dependent T cell recruitment and suppresses responses to anti-PD-1 treatment. Nat Commun 14, 4253 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Lerner L. et al. Growth differentiating factor-15 (GDF-15): A potential biomarker and therapeutic target for cancer-associated weight loss. Oncol Lett 12, 4219–4223 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.May B.M. et al. Growth/differentiation factor-15 (GDF-15) as a predictor of serious arrhythmic events in patients with nonischemic dilated cardiomyopathy. J Electrocardiol 70, 19–23 (2022). [DOI] [PubMed] [Google Scholar]
  • 51.Molfino A. et al. Association between Growth Differentiation Factor-15 (GDF-15) Serum Levels, Anorexia and Low Muscle Mass among Cancer Patients. Cancers (Basel) 13(2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Varhaug K.N., Hikmat O., Nakkestad H.L., Vedeler C.A. & Bindoff L.A. Serum biomarkers in primary mitochondrial disorders. Brain Commun 3, fcaa222 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Groarke J.D. et al. Ponsegromab for the Treatment of Cancer Cachexia. N Engl J Med (2024). [DOI] [PubMed] [Google Scholar]
  • 54.https://www.clinicaltrials.gov/study/NCT04725474, accessed 07/23/2024.
  • 55.Zhong X. et al. Sex specificity of pancreatic cancer cachexia phenotypes, mechanisms, and treatment in mice and humans: role of Activin. J Cachexia Sarcopenia Muscle 13, 2146–2161 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Park M.A. et al. Defining and Addressing Research Priorities in Cancer Cachexia through Transdisciplinary Collaboration. Cancers (Basel) 16(2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Vigano A.L. et al. The abridged patient-generated subjective global assessment is a useful tool for early detection and characterization of cancer cachexia. J Acad Nutr Diet 114, 1088–1098 (2014). [DOI] [PubMed] [Google Scholar]
  • 58.Silva C.L. & Faccioli L.H. Tumor necrosis factor (cachectin) mediates induction of cachexia by cord factor from mycobacteria. Infect Immun 56, 3067–71 (1988). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Yehia R. et al. Impact of TNF-alpha Gene Polymorphisms on Pancreatic and Non-Small Cell Lung Cancer-Induced Cachexia in Adult Egyptian Patients: A Focus on Pathogenic Trajectories. Front Oncol 11, 783231 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Gillespie S.L. et al. Lifetime stressor exposure, systemic inflammation during pregnancy, and preterm birth among Black American women. Brain Behav Immun 101, 266–274 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Strahler J., Rohleder N. & Wolf J.M. Acute psychosocial stress induces differential short-term changes in catecholamine sensitivity of stimulated inflammatory cytokine production. Brain Behav Immun 43, 139–48 (2015). [DOI] [PubMed] [Google Scholar]
  • 62.Rybicki B.A. et al. Growth and differentiation factor 15 and NF-kappaB expression in benign prostatic biopsies and risk of subsequent prostate cancer detection. Cancer Med 10, 3013–3025 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Rohatgi A. et al. Association of growth differentiation factor-15 with coronary atherosclerosis and mortality in a young, multiethnic population: observations from the Dallas Heart Study. Clin Chem 58, 172–82 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Ma B. et al. TIMP1 promotes cell proliferation and invasion capability of right-sided colon cancers via the FAK/Akt signaling pathway. Cancer Sci 113, 4244–4257 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Eggers K.M. et al. Relations of growth-differentiation factor-15 to biomarkers reflecting vascular pathologies in a population-based sample of elderly subjects. Scand J Clin Lab Invest 72, 45–51 (2012). [DOI] [PubMed] [Google Scholar]
  • 66.Fan M. et al. Atractylenolide I ameliorates cancer cachexia through inhibiting biogenesis of IL-6 and tumour-derived extracellular vesicles. J Cachexia Sarcopenia Muscle 13, 2724–2739 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Shmarina G.V. et al. Tumor necrosis factor-alpha/interleukin-10 balance in normal and cystic fibrosis children. Mediators Inflamm 10, 191–7 (2001). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Zhu Z., Tang C., Xu T. & Zhao Z. Molecular Analysis of Prognosis and Immune Pathways of Pancreatic Cancer Based on TNF Family Members. J Oncol 2021 , 2676996 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Deng J. et al. DDR1-induced neutrophil extracellular traps drive pancreatic cancer metastasis. JCI Insight 6(2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Zhang T. et al. CXCL1 mediates obesity-associated adipose stromal cell trafficking and function in the tumour microenvironment. Nat Commun 7, 11674 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Lin J. et al. Proinflammatory phenotype in major depressive disorder with adulthood adversity: In line with social signal transduction theory of depression. J Affect Disord 341 , 275–282 (2023). [DOI] [PubMed] [Google Scholar]
  • 72.Karshikoff B. et al. Inflammatory Blood Signature Related to Common Psychological Comorbidity in Chronic Pain. Biomedicines 11(2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Kemp S.B. et al. Apolipoprotein E Promotes Immune Suppression in Pancreatic Cancer through NF-kappaB-Mediated Production of CXCL1. Cancer Res 81, 4305–4318 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Walsh R.M. et al. Adipose-Tumor Crosstalk contributes to CXCL5 Mediated Immune Evasion in PDAC. bioRxiv (2023). [Google Scholar]
  • 75.Hsieh M.M. et al. Neutrophil count in African Americans: lowering the target cutoff to initiate or resume chemotherapy? J Clin Oncol 28, 1633–7 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Tavakkoli A. et al. Racial Disparities and Trends in Pancreatic Cancer Incidence and Mortality in the United States. Clin Gastroenterol Hepatol 18, 171–178 e10 (2020). [DOI] [PubMed] [Google Scholar]
  • 77.Rupert J.E. et al. Tumor-derived IL-6 and trans-signaling among tumor, fat, and muscle mediate pancreatic cancer cachexia. J Exp Med 218(2021). [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Research Square are provided here courtesy of American Journal Experts

RESOURCES