Skip to main content

The association between fibrinogen-to-albumin ratio (FAR) and adverse prognosis in patients with acute decompensated heart failure at different glucose metabolic states

Abstract

Background

Circulating fibrinogen-to-albumin ratio (FAR) has been proposed as a novel inflammatory biomarker and a cardiovascular disease risk predictor. However, its prognostic value in patients with acute decompensated heart failure (ADHF) and different glycemic metabolic states remains ambiguous.

Methods

A total of 1031 hospitalized patients with ADHF from January 2018 to May 2021 were included in the study. The primary endpoints were the major adverse cardiac and cerebral events (MACCEs). Patients were categorized into high-level FAR (FAR-H) and low-level FAR (FAR-L) groups based on the optimal cut-off value of FAR obtained from restricted cubic spline function analysis. The Kaplan–Meier plots and three multivariate-adjusted Cox proportional hazard models were used to determine the association between FAR and the risk of developing MACCEs in patients with ADHF at different glycemic metabolic states.

Results

MACCEs occurred in 483 (46.8%) patients during a median follow-up time of 520 days. The optimal FAR cut-off value was 0.079. Upon analyzing the Kaplan–Meier plots, the incidence of MACCEs was significantly different between the FAR groups in all patients and patients with diabetes mellitus (p < 0.05). After adjusting for the confounding factors, the hazard ratio (HR) for MACCEs in the FAR-H group was 1.29 compared with the FAR-L group in all patients (Model 3: 95% CI 1.07–1.56, p = 0.007). Additionally, high FAR was associated with MACCEs in three multivariate Cox models (Model 1, HR = 1.52, 95% CI 1.17–1.96, p = 0.002; Model 2, HR = 1.46, 95% CI 1.13–1.89, p = 0.004; Model 3, HR = 1.48, 95% CI 1.14–1.92, p = 0.003) in DM patients. But no significant differences were found between the FAR groups for prediabetes mellitus (Pre-DM) and normal glucose regulation (NGR) using the three Cox models (all p-values were > 0.05).

Conclusions

Elevated FAR was independently associated with poor prognosis in patients with ADHF and DM and thus could be used as a risk stratification tool and a potential therapeutic target in the future.

Introduction

Although systemic inflammation has been considered a common pathophysiological characteristic of acute and chronic heart failure (HF) [1, 2], the effect of inflammation in HF has long been controversial. Several clinical trials on anti-inflammation and HF yielded controversial outcomes [3,4,5,6]. However, the outcome of the CANTOS trial [7] published in 2019 revealed chronic inflammation as a critical process in the pathogenesis of HF, leading to the exploration of more biomarkers screening patients with HF that may benefit from anti-inflammatory strategies. Various biomarkers were elevated in response to the activation of systemic inflammation in patients with HF [8], and it is more evident in acute HF [2].

Fibrinogen is a glycoprotein complex synthesized in the liver that takes part in the processes of inflammation and thrombosis [9, 10]. Previous studies found associations of fibrinogen with the risk of mortality in patients with coronary artery disease (CAD) [11]. Albumin is a protein synthesized in the liver and influences nutrient absorption, colloidal pressure, and systemic inflammation [12, 13]. Fibrinogen-to-albumin ratio (FAR) is the proportion of these two indexes, which is associated with poor prognosis in a variety of cancers [14,15,16,17]. The FAR was also directly associated with the severity of coronary artery calcification and adverse prognosis of the acute coronary syndrome, especially in patients with diabetes mellitus (DM) [18,19,20]. Furthermore, FAR was more sensitive and specific in predicting major adverse cardiovascular events (MACE) than fibrinogen and albumin alone [21, 22].

As common comorbidity of heart failure, DM creates a chronic low-grade inflammatory environment, leading to further impairment of cardiac structure and function [4, 23]. Patients with HF and DM had a worse prognosis than patients with HF alone [24, 25]. Nevertheless, reports are lacking on the relationship of FAR with the prognosis of acute-decompensated heart failure (ADHF). Therefore, this retrospective study aimed to evaluate the relationship between FAR and major adverse cardiac and cerebrovascular events (MACCEs) in patients with ADHF under different glucose metabolism states.

Materials and methods

Study population

The single-center retrospective analysis of 1502 consecutive patients with ADHF admitted to Nanjing Drum Tower Hospital from January 2018 to May 2021 was carried out. The ADHF was defined using the 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure [26]. The exclusion criteria were: (1) patients with missing baseline data, (2) patients who were younger than 18 years, (3) patients with acute coronary syndrome, (4) patients with advanced cancer, and (5) patients who were lost to follow-up. The inclusion criteria are detailed in Fig. 1. This study was performed following the Declaration of Helsinki and with the approval of the Ethics Committee of the Nanjing Drum Tower Hospital.

Fig. 1
figure 1

Flow diagram of patient selection

Data collection and definitions

Trained physicians retrieved patient demographics, clinical history, anthropometric data, laboratory test results, echocardiographic data, and information on medication during hospitalization from the electronic medical records.

FAR was defined as the proportion of the plasma fibrinogen (g/L) to the plasma albumin level (g/L). Body mass index (BMI) was denoted as weight (kg)/(height [m])2. Based on the recommendations from the American Diabetes Association [27], DM was defined as fasting plasma glucose (FPG) of ≥ 7.0 mmol/L or glycated hemoglobin (HbA1c) of ≥ 6.5%, or the current use of hypoglycemic medications. Prediabetes mellitus (Pre-DM) was defined as 5.6 mmol/L ≤ FPG < 7.0 mmol/L or 5.7% ≤ HbA1c < 6.5%, and normal glucose regulation (NGR) was considered patients without pre-DM or DM. Hypertension was considered when there was a systolic blood pressure (SBP) of ≥ 140 mmHg and/or diastolic blood pressure (DBP) of ≥ 90 mmHg, any use of antihypertensive drugs, or having a history of hypertension. Hyperlipidemia was defined when the fasting total cholesterol (TC) was ≥ 240 mg/dL, low-density lipoprotein cholesterol (LDL-C) was > 160 mg/dL, TGs were ≥ 150 mg/dL, high-density lipoprotein cholesterol (HDL-C) was < 40 mg/dL, or lipid-lowering drugs were previously used.

The peripheral venous blood samples were initially collected after overnight fasting (> 8 h) and the different variables were determined in the laboratory. Fibrinogen, albumin (ALB), C-reactive protein (CRP), B-type natriuretic peptide (BNP), hemoglobin, hematocrit, uric acid (UA), creatinine, white blood cell count (WBC), FPG, HbA1c, total cholesterol, triglycerides, LDL-C, HDL-C, serum sodium, serum potassium, alanine aminotransferase (ALT), and aspartate transaminase (AST) were measured.

Endpoints and follow-up

The primary endpoint was MACCEs, such as all-cause death, cardiovascular death, non-fatal myocardial infarction, non-fatal ischemic stroke, and chronic HF worsening.

Patients were followed up by telephone and/or during clinic visits every six months.

Statistical analysis

Continuous variables were expressed as mean ± standard deviation (SD) or median [interquartile range (IQR)]. Categorical variables were expressed as numbers (percentages).

A restricted cubic spline regression analysis was performed to determine the optimal cut-off value of the FAR. Patients were categorized into two groups based on their FAR values [FAR-L (< 0.079) and FAR-H (≥ 0.079)]. Continuous variables were compared using analysis of variance (ANOVA) or the Kruskal–Wallis test. Categorical variables were compared using the χ2 test.

The cumulative event-free survival rate was calculated using the Kaplan–Meier plots and the log-rank test. Multivariate Cox proportional hazards models were applied to determine the associations of FAR with the incidence rates of MACCEs. Schoenfeld residuals were visually interpreted to validate the assumptions of proportional hazards before these analyses. The baseline variables with a p-value of < 0.1 or clinically significant were included in the Cox proportional hazards models. Multicollinearity was tested in the multivariate models with a threshold of variance inflation factor of < 5. Finally, three multivariate regression models remained: Model 1 with the adjustment for age, sex, BMI, SBP, DBP, and heart rate; Model 2 with the adjustment for variables included in Model 1 plus BNP, creatinine, WBC, sodium, UA, ALT, and left ventricular ejection fraction (LVEF); Model 3 with the adjustment for variables included in Model 2 plus history of hypertension, valvular heart disease, atrial fibrillation, and use of aldosterone antagonist, β-Blockers, ACEI/ARB/ARNI, or SGLT2i. Moreover, the prognostic value of FAR for MACCEs at different glucose metabolic states was also assessed by the models. Hazard ratios (HRs) were calculated and the results were reported as HRs and 95% confidence intervals (CIs). The low FAR was used as a reference in the three models. The correlation between FAR and cardiometabolic-related factors was analyzed using Spearman’s rank correlation. The subgroup analyses including age, sex, BMI, and glucose metabolism state were shown as the forest plot. All data were analyzed using R version 4.1.0, GraphPad Prism version 8.0.1, and SPSS for windows version 26. A P-value of < 0.05 was considered statistically significant.

Results

General characteristics of patients

The clinical characteristics of the participants relevant to the occurrence of MACCEs are listed in Table 1. The median follow-up time was 520 days. The proportions of patients who previously experienced non-ischemic cardiomyopathy (NIMD), pre-DM, NGR, hyperlipidemia, intake of angiotensin-converting enzyme inhibitor/angiotensin receptor inhibitor/ angiotensin receptor neprilysin inhibitor (ACEI/ARB/ARNI) were significantly lower in the MACCEs group. Further, DBP, ALB, hemoglobin, hematocrit, sodium, total cholesterol, FPG, and HDL-C were lower in the MACCEs group (all p-values were < 0.05). Additionally, the patients in the MACCEs group were older and had higher levels of FAR, HbA1C, CRP, BNP, UA, Creatinine, AST, the proportion of DM, and digoxin intake (all p-values were < 0.05).

Table 1 Baseline clinical characteristics and MACCEs

Comparison of clinical data among the two groups

Based on the restricted cubic spline analysis (Fig. 2), the optimal FAR cut-off value was 0.079 (p-value for non-linear was < 0.001). The patients were categorized into 2 groups based on the FAR value and the baseline characteristics of the two groups were analyzed (Table 2). In the FAR-H group, the fibrinogen, FPG, HbA1C, BNP, CRP, and WBC levels, the proportion of IMD, and occurrence of atrial fibrillation, hypertension, and the use of SGLT2i were significantly higher, compared to the FAR-L group. But the BMI, ALB, hemoglobin, hematocrit, sodium, HDL-C, AST, LVDD, LAD, the proportion of NIMD were relatively lower in the FAR-H group.

Fig. 2
figure 2

Hazard ratios for the MACCEs based on restricted cubic spine function for FAR. The red line represents the reference of the hazard ratios, and the red area represents 95% confidence intervals

Table 2 Clinical characteristics according to the level of FAR

Prediction of the incidence of MACCEs by FAR

The FAR was significantly associated with the incidence of MACCEs in all participants, as shown in Fig. 3A. Table 3 shows three multivariate Cox proportional hazard models to determine the correlation between the FAR groups and MACCEs. Consequently, high FAR was associated with higher incidence of MACCEs (Model 1, HR = 1.38, 95% CI 1.14–1.66, p = 0.001; Model 2, HR = 1.29, 95% CI 1.07–1.55, p = 0.008; Model 3, HR = 1.29, 95% CI 1.07–1.56, p = 0.007).

Fig. 3
figure 3

The Kaplan-Meier analysis for MACCEs according to different FAR levels for total patients (A), patients with DM (B), patients with Pre-DM (C) and patients with NGR (D)

Table 3 The HR (95% CI) of MACCEs according to FAR and different glucose metabolism states from the three models

Associations between FAR and MACCEs at different glucose metabolism states

Specific to different glucose metabolism states, the Kaplan–Meier curves (Fig. 3B–D) showed a significant difference in the incidence of MACCEs for DM between the FAR groups (P = 0.005), while no significant difference was found for pre-DM and NGR (all p-values were > 0.05). The high FAR was associated with MACCEs for DM in three multivariate Cox models (Model 1, HR = 1.52, 95% CI 1.17–1.96, p = 0.002; Model 2, HR = 1.46, 95% CI 1.13–1.89, p = 0.004; Model 3, HR = 1.48, 95% CI 1.14–1.92, p = 0.003; Table 3). But no significant differences were found between the FAR groups for pre-DM and NGR in three Cox models (all p-values were > 0.05). The FAR-H-DM combination group had the highest risk of developing MACCEs compared with the other groups (p = 0.005) (Fig. 4).

Fig. 4
figure 4

The Kaplan-Meier curve analysis for MACCEs according to status of FAR and glycemic metabolism

The correlation between FAR and other cardiometabolic factors

The Spearman’s rank correlation analysis outcomes are reported in Table 4. The FAR was positively correlated with age, CRP, WBC, BNP, FPG, and HbA1C, but negatively correlated with BMI and HDL-C (all p-values were < 0.05).

Table 4 The correlation coefficients between FAR and other cardiometabolic factors

Subgroup analysis

On post-hoc subgroup analysis, the FAR-H was associated with a high incidence of MACCEs, which was consistent across subgroups, age, sex, and BMI (Fig. 5). There were no interactions between the FAR and the above variables in the subgroup analyses (all p-values for interaction were > 0.05). But interaction between FAR and glucose metabolism state was seen (p-value for the interaction = 0.006). In the DM group, FAR-H was associated with the high risk of MACCEs, but not in the other groups.

Fig. 5
figure 5

The forest plot of MACCEs specific to different subgroups. Adjusted model included FAR, age, sex, body mass index, systolic blood pressure, diastolic blood pressure, heart rate, BNP, sodium,creatinine, UA, WBC, ALT, LVEF, history of hypertension, valvular heart disease, atrial fibrillation, aldosterone antagonist, β-blockers, ACEI/ARB/ARNI, and SGLT2i use

Discussion

In this retrospective study, the relationship between FAR, a simple proportion, and the risk of MACCEs in patients with ADHF was explored in a clinical setting. Upon analysis, the FAR was positively correlated with the indicators of glucose metabolism, such as HbA1C and FPG. Moreover, high FAR was significantly associated with the increased risk of MACCEs in patients with ADHF and DM. However, we did not find a significant correlation in patients with ADHF at pre-DM state.

The association between FAR and CVD risk

FAR, as a proportion of fibrinogen and albumin, has been elucidated in several diseases. Numerous studies demonstrated that preoperative FAR was associated with the adverse prognosis after surgery in many cancers, such as hepatocellular carcinoma, cholangiocarcinoma, and glioblastoma [28,29,30,31]. Additionally, FAR has been frequently studied in cardiovascular diseases (CVD), particularly CAD. FAR was a useful tool in predicting the severity of CAD in patients with the acute coronary syndrome (ACS) [32,33,34,35]. FAR was associated with the long-term prognosis in patients with ST-elevation myocardial infarction (STEMI), non-STEMI, and CAD who underwent percutaneous coronary intervention (PCI) [35,36,37,38]. Park et al. found FAR as a prognostic marker of high mortality in patients with CAD who underwent off-pump coronary artery bypass grafting [39].

Plausible association between FAR and ADHF with the DM state

Fibrinogen, an inflammatory marker [40], was increased in chronic inflammatory conditions such as diabetes, atherosclerosis, and HF [10, 41, 42]. The fibrinogen was related to the high risk of HF incidence [43, 44]. Meng et al. reported that high levels of fibrinogen displayed a higher risk of 90-day mortality in patients with ADHF [45]. Albumin was considered an important biomarker of malnutrition and inflammation in patients with HF [46]. Hypoalbuminemia was a predictor of short-term prognosis in patients with acute heart failure [46].

Wang et al. showed that a higher level of FAR with the DM state was associated with the worse 5-year outcome in patients with CAD undergoing PCI [20]. The role of chronic and low-grade inflammation in HF with DM was demonstrated, and DM as comorbidity contributed to the worse prognosis in patients with HF [4, 7, 23]. However, FAR has not yet been correlated with HF at the DM state, especially ADHF, which is a highly heterogeneous syndrome [47, 48]. In this study, the significant predictive value of FAR for developing MACCEs was demonstrated in patients with ADHF and DM, for the first time.

Potential mechanisms between FAR and ADHF

The underlying mechanism between FAR and ADHF needs to be elucidated. Acute phase reactants (APR) are inflammatory markers, which are produced in the liver in response to interleukin-6 (IL-6) [49]. APR can be upregulated or downregulated, with increasing or decreasing concentration during inflammation [49]. Positive APR includes procalcitonin, C-reactive protein, and fibrinogen, and negative APR includes albumin, prealbumin, and transferrin [49]. When acute exacerbation of chronic HF occurs, protein metabolism in the liver may synthesize pro-inflammatory proteins such as fibrinogen, emphasizing the role of a positive APR. Meanwhile, the hypoalbuminemia in the physiopathology of CVD may be from anti-inflammatory, antioxidant, anticoagulant, and anti-aggregation responses. Hypoalbuminemia was associated with long-term protein dystrophy in chronic HF [50], and albumin may have been further affected by the inflammatory status in patients with ADHF. Thus, when patients are with ADHF at the DM state, the higher the FAR level and the higher the degree of inflammation that may occur.

FAR as a potential therapeutic target

Previous research demonstrated that fibrinogen up-regulated the expression of proinflammatory cytokines, such as interleukin-1 (IL-1) and tumor necrosis factor-α (TNF-α) [51]. In the CANTOS study, IL-1β targeting Canakinumab significantly reduced the incidence of MACE in HF patients [7]. Inhibition of fibrinolysis by drugs, such as tranexamic acid, reduced inflammatory response in patients who underwent cardiopulmonary bypass [52]. However, no evidence currently exists that reducing fibrinogen or correcting hypoalbuminemia improves the prognosis of CVD. Therefore, the potential mechanisms and targeted therapeutic strategies of FAR in ADHF with DM state should be further investigated.

Limitations

This study also had several limitations. First, due to the retrospective design, the FAR was not dynamically measured in patients over the follow-up period. Second, as this was a single-center study with limited sample size, data bias could not be avoided despite correcting multiple confounding factors. Third, prospective cohort studies are required to validate these findings.

Conclusion

This study demonstrated that FAR was independently associated with the adverse prognosis in patients with ADHF at DM state, but this association was not observed in patients at pre-DM and NGR states.

Availability of data and materials

The information and data of the study population were extracted from the hospital information system. The datasets are not publicly available because the privacy of the participants should be protected. Data are however available from the corresponding author on a reasonable request.

Abbreviations

HF:

Heart failure

CAD:

Coronary artery disease

FAR:

Fibrinogen-to-albumin ratio

DM:

Diabetes mellitus

MACE:

Major adverse cardiovascular events

MACCEs:

Major adverse cardiac and cerebral events

ADHF:

Acute decompensated heart failure

BMI:

Body mass index

FPG:

Fasting plasma glucose

HbA1c:

Glycated hemoglobin

Pre-DM:

Prediabetes mellitus

NGR:

Normal glucose regulation

SBP:

Systolic blood pressure

DBP:

Diastolic blood pressure

TC:

Total cholesterol

LDL-C:

Low-density lipoprotein-C

HDL‑C:

High‑density lipoprotein cholesterol

ALB:

Albumin

CRP:

C-reactive protein

BNP:

B-type natriuretic peptide

UA:

Uric acid

WBC:

White blood cell

ALT:

Aspartate transaminase

AST:

Alanine aminotransferase

SD:

Standard deviation

IQR:

Interquartile range

ANOVA:

Analysis of variance

LVEF:

Left ventricular ejection fraction

HRs:

Hazard ratios

CIs:

95% Confidence intervals NIMD: none-ischemic cardiomyopathy

ACEI:

Angiotensin-converting enzyme inhibitor

ARB:

Angiotensin receptor inhibitor

ARNI:

Angiotensin receptor enkephalinase inhibitor

SGLT2i:

Sodium-glucose contrasporter-2 inhibitor

CVD:

Cardiovascular diseases

PCI:

Percutaneous coronary intervention

APR:

Acute phase reactants

IL-1:

Interleukin-1

TNF-α:

Tumor necrosis factor-α

References

  1. Pfisterer M, Buser P, Rickli H, Gutmann M, Erne P, Rickenbacher P, Vuillomenet A, Jeker U, Dubach P, Beer H, et al. BNP-guided vs symptom-guided heart failure therapy: the trial of intensified vs standard medical therapy in elderly patients with congestive heart failure (TIME-CHF) randomized trial. JAMA. 2009;301(4):383–92.

    Article  CAS  PubMed  Google Scholar 

  2. O’Connor CM, Starling RC, Hernandez AF, Armstrong PW, Dickstein K, Hasselblad V, Heizer GM, Komajda M, Massie BM, McMurray JJ, et al. Effect of nesiritide in patients with acute decompensated heart failure. N Engl J Med. 2011;365(1):32–43.

    Article  CAS  PubMed  Google Scholar 

  3. Adamo L, Rocha-Resende C, Prabhu SD, Mann DL. Reappraising the role of inflammation in heart failure. Nat Rev Cardiol. 2020;17(5):269–85.

    Article  PubMed  Google Scholar 

  4. Murphy SP, Kakkar R, McCarthy CP, Januzzi JL Jr. Inflammation in heart failure: JACC state-of-the-art review. J Am Coll Cardiol. 2020;75(11):1324–40.

    Article  PubMed  Google Scholar 

  5. Mann DL, McMurray JJ, Packer M, Swedberg K, Borer JS, Colucci WS, Djian J, Drexler H, Feldman A, Kober L, et al. Targeted anticytokine therapy in patients with chronic heart failure: results of the randomized etanercept worldwide evaluation (RENEWAL). Circulation. 2004;109(13):1594–602.

    Article  CAS  PubMed  Google Scholar 

  6. Chung ES, Packer M, Lo KH, Fasanmade AA, Willerson JT. Anti TNFTACHFI: Randomized, double-blind, placebo-controlled, pilot trial of infliximab, a chimeric monoclonal antibody to tumor necrosis factor-alpha, in patients with moderate-to-severe heart failure: results of the anti-TNF therapy against congestive heart failure (ATTACH) trial. Circulation. 2003;107(25):3133–40.

    Article  CAS  PubMed  Google Scholar 

  7. Everett BM, Cornel JH, Lainscak M, Anker SD, Abbate A, Thuren T, Libby P, Glynn RJ, Ridker PM. Anti-inflammatory therapy with canakinumab for the prevention of hospitalization for heart failure. Circulation. 2019;139(10):1289–99.

    Article  CAS  PubMed  Google Scholar 

  8. Redfield MM, Chen HH, Borlaug BA, Semigran MJ, Lee KL, Lewis G, LeWinter MM, Rouleau JL, Bull DA, Mann DL, et al. Effect of phosphodiesterase-5 inhibition on exercise capacity and clinical status in heart failure with preserved ejection fraction: a randomized clinical trial. JAMA. 2013;309(12):1268–77.

    Article  CAS  PubMed  Google Scholar 

  9. Tousoulis D, Papageorgiou N, Androulakis E, Briasoulis A, Antoniades C, Stefanadis C. Fibrinogen and cardiovascular disease: genetics and biomarkers. Blood Rev. 2011;25(6):239–45.

    Article  CAS  PubMed  Google Scholar 

  10. Barazzoni R, Zanetti M, Davanzo G, Kiwanuka E, Carraro P, Tiengo A, Tessari P. Increased fibrinogen production in type 2 diabetic patients without detectable vascular complications: correlation with plasma glucagon concentrations. J Clin Endocrinol Metab. 2000;85(9):3121–5.

    Article  CAS  PubMed  Google Scholar 

  11. Yuan D, Jiang P, Zhu P, Jia S, Zhang C, Liu Y, Liu R, Xu J, Tang X, Zhao X, et al. Prognostic value of fibrinogen in patients with coronary artery disease and prediabetes or diabetes following percutaneous coronary intervention: 5-year findings from a large cohort study. Cardiovasc Diabetol. 2021;20(1):143.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Mitch WE. Malnutrition: a frequent misdiagnosis for hemodialysis patients. J Clin Invest. 2002;110(4):437–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Don BR, Kaysen G. Serum albumin: relationship to inflammation and nutrition. Semin Dial. 2004;17(6):432–7.

    Article  PubMed  Google Scholar 

  14. Zhu H, Zhang W, Ye P, Wu M. Preoperative circulating fibrinogen to albumin ratio in predicting 5-year prognosis of oral cancer radical surgery. Neoplasma. 2022.

  15. Yang Q, Liang D, Yu Y, Lv F. The prognostic significance of the fibrinogen-to-albumin ratio in patients with triple-negative breast cancer: a retrospective study. Front Surg. 2022;9:916298.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Xu J, Li S, Feng Y, Zhang J, Peng Y, Wang X, Wang H. The fibrinogen/albumin ratio index as an independent prognostic biomarker for patients with combined hepatocellular cholangiocarcinoma after surgery. Cancer Manag Res. 2022;14:1795–806.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Wu W, Wang Q, Han D, Li J, Nie Y, Guo D, Yang L, Tao K, Zhang X, Dou K. Prognostic value of preoperative inflammatory markers in patients with hepatocellular carcinoma who underwent curative resection. Cancer Cell Int. 2021;21(1):500.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Zhu Y, Tao S, Zhang D, Xiao J, Wang X, Yuan L, Pan H, Wang D. Association between fibrinogen/albumin ratio and severity of coronary artery calcification in patients with chronic kidney disease: a retrospective study. PeerJ. 2022;10:e13550.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Celebi S, Ozcan Celebi O, Berkalp B, Amasyali B. The association between the fibrinogen-to-albumin ratio and coronary artery disease severity in patients with stable coronary artery disease. Coron Artery Dis. 2020;31(6):512–7.

    Article  PubMed  Google Scholar 

  20. Wang P, Yuan D, Zhang C, Zhu P, Jia S, Song Y, Tang X, Xu J, Li T, Zeng G, et al. High fibrinogen-to-albumin ratio with type 2 diabetes mellitus is associated with poor prognosis in patients undergoing percutaneous coronary intervention: 5-year findings from a large cohort. Cardiovasc Diabetol. 2022;21(1):46.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Karahan O, Acet H, Ertas F, Tezcan O, Caliskan A, Demir M, Kaya AF, Demirtas S, Cevik MU, Yavuz C. The relationship between fibrinogen to albumin ratio and severity of coronary artery disease in patients with STEMI. Am J Emerg Med. 2016;34(6):1037–42.

    Article  PubMed  Google Scholar 

  22. Xiao L, Jia Y, Wang X, Huang H. The impact of preoperative fibrinogen-albumin ratio on mortality in patients with acute ST-segment elevation myocardial infarction undergoing primary percutaneous coronary intervention. Clin Chim Acta. 2019;493:8–13.

    Article  CAS  PubMed  Google Scholar 

  23. Odegaard AO, Jacobs DR Jr, Sanchez OA, Goff DC Jr, Reiner AP, Gross MD. Oxidative stress, inflammation, endothelial dysfunction and incidence of type 2 diabetes. Cardiovasc Diabetol. 2016;15:51.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Kong MG, Jang SY, Jang J, Cho HJ, Lee S, Lee SE, Kim KH, Yoo BS, Kang SM, Baek SH, et al. Impact of diabetes mellitus on mortality in patients with acute heart failure: a prospective cohort study. Cardiovasc Diabetol. 2020;19(1):49.

    Article  PubMed  PubMed Central  Google Scholar 

  25. De Groote P, Lamblin N, Mouquet F, Plichon D, McFadden E, Van Belle E, Bauters C. Impact of diabetes mellitus on long-term survival in patients with congestive heart failure. Eur Heart J. 2004;25(8):656–62.

    Article  PubMed  Google Scholar 

  26. McDonagh TA, Metra M, Adamo M, Gardner RS, Baumbach A, Bohm M, Burri H, Butler J, Celutkiene J, Chioncel O, et al. 2021 ESC guidelines for the diagnosis and treatment of acute and chronic heart failure. developed by the task force for the diagnosis and treatment of acute and chronic heart failure of the European society of cardiology (ESC). With the special contribution of the heart failure association (HFA) of the ESC. G Ital Cardiol. 2022;23(4 Suppl 1):e1–127.

    Google Scholar 

  27. American Diabetes Association. 2. Classification and diagnosis of diabetes: standards of medical care in diabetes-2019. Diabetes Care. 2019;42(Suppl 1):S13–28.

    Article  Google Scholar 

  28. Li J, Zhou X, Xiang Y, Zhang S, Feng W, Yuan Y, Liu Y, Yin S. Clinical significance of preoperative fibrinogen to albumin ratio in patients with glioblastoma: a singe center experience. Cancer Manag Res. 2021;13:3259–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Mai RY, Bai T, Luo XL, Wu GB. Preoperative fibrinogen-to-albumin ratio predicts the prognosis of patients with hepatocellular carcinoma subjected to hepatectomy. BMC Gastroenterol. 2022;22(1):261.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Liu H, Qiu G, Hu F, Wu H. Fibrinogen/albumin ratio index is an independent predictor of recurrence-free survival in patients with intrahepatic cholangiocarcinoma following surgical resection. World J Surg Oncol. 2021;19(1):218.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Wang Y, Xu W, Wang Y. Prognostic role of preoperative fibrinogen to albumin ratio in breast cancer. Clin Chim Acta. 2020;510:360–2.

    Article  CAS  PubMed  Google Scholar 

  32. Binti NN, Ferdausi N, Anik MEK, Islam LN. Association of albumin, fibrinogen, and modified proteins with acute coronary syndrome. PLoS ONE. 2022;17(7):e0271882.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Duan Z, Luo C, Fu B, Han D. Association between fibrinogen-to-albumin ratio and the presence and severity of coronary artery disease in patients with acute coronary syndrome. BMC Cardiovasc Disord. 2021;21(1):588.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Erdogan G, Arslan U, Yenercag M, Durmus G, Tugrul S, Sahin I. Relationship between the fibrinogen-to-albumin ratio and syntax score in patients with non-ST-elevation myocardial infarction. Rev Invest Clin. 2021;10:11.

    Google Scholar 

  35. Li M, Tang C, Luo E, Qin Y, Wang D, Yan G. Relation of fibrinogen-to-albumin ratio to severity of coronary artery disease and long-term prognosis in patients with non-st elevation acute coronary syndrome. Biomed Res Int. 2020;2020:1860268.

    PubMed  PubMed Central  Google Scholar 

  36. Liu G, Fan CM, Guo H, Fan WN, Li ML, Cui GX. Fibrinogen-to-albumin ratio predicts long-term outcomes for patients with ST-elevation myocardial infarction and multivessel disease: a prospective observational cohort study. Exp Ther Med. 2021;21(5):465.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. He D, Jiao Y, Yu T, Song J, Wen Z, Wu J, Duan W, Sun N, Sun Z, Sun Z. Prognostic value of fibrinogen-to-albumin ratio in predicting 1-year clinical progression in patients with non-ST elevation acute coronary syndrome undergoing percutaneous coronary intervention. Exp Ther Med. 2019;18(4):2972–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Zhang DP, Mao XF, Wu TT, Chen Y, Hou XG, Yang Y, Ma X, Zhang JY, Ma YT, Xie X, et al. The fibrinogen-to-albumin ratio is associated with outcomes in patients with coronary artery disease who underwent percutaneous coronary intervention. Clin Appl Thromb Hemost. 2020;26:1076029620933008.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Park S, Nam K, Kim TK. Association between preoperative fibrinogen-to-albumin ratio and all-cause mortality after off-pump coronary artery bypass grafting: a retrospective observational study. Anesth Analg. 2022;134(5):1021–7.

    Article  CAS  PubMed  Google Scholar 

  40. Fibrinogen Studies C, Danesh J, Lewington S, Thompson SG, Lowe GD, Collins R, Kostis JB, Wilson AC, Folsom AR, Wu K, et al. Plasma fibrinogen level and the risk of major cardiovascular diseases and nonvascular mortality: an individual participant meta-analysis. JAMA. 2005;294(14):1799–809.

    Google Scholar 

  41. Witte KK, Ford SJ, Preston T, Parker JD, Clark AL. Fibrinogen synthesis is increased in cachectic patients with chronic heart failure. Int J Cardiol. 2008;129(3):363–7.

    Article  PubMed  Google Scholar 

  42. Back M, Yurdagul A Jr, Tabas I, Oorni K, Kovanen PT. Inflammation and its resolution in atherosclerosis: mediators and therapeutic opportunities. Nat Rev Cardiol. 2019;16(7):389–406.

    PubMed  PubMed Central  Google Scholar 

  43. Eisen A, Benderly M, Behar S, Goldbourt U, Haim M. Inflammation and future risk of symptomatic heart failure in patients with stable coronary artery disease. Am Heart J. 2014;167(5):707–14.

    Article  PubMed  Google Scholar 

  44. Suthahar N, Lau ES, Blaha MJ, Paniagua SM, Larson MG, Psaty BM, Benjamin EJ, Allison MA, Bartz TM, Januzzi JL Jr, et al. Sex-specific associations of cardiovascular risk factors and biomarkers with incident heart failure. J Am Coll Cardiol. 2020;76(12):1455–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Meng Z, Zhao Y, He Y. Fibrinogen level predicts outcomes in critically ill patients with acute exacerbation of chronic heart failure. Dis Markers. 2021;2021:6639393.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Ancion A, Allepaerts S, Oury C, Gori AS, Pierard LA, Lancellotti P. Serum albumin level and hospital mortality in acute non-ischemic heart failure. ESC Heart Fail. 2017;4(2):138–45.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Njoroge JN, Teerlink JR. Pathophysiology and therapeutic approaches to acute decompensated heart failure. Circ Res. 2021;128(10):1468–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Essay P, Balkan B, Subbian V. Decompensation in critical care: early prediction of acute heart failure onset. JMIR Med Inform. 2020;8(8): e19892.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Gulhar R, Ashraf MA, Jialal I: Physiology, Acute Phase Reactants. In: StatPearls. Florida: Treasure Island. 2022.

  50. Arques S, Ambrosi P. Human serum albumin in the clinical syndrome of heart failure. J Card Fail. 2011;17(6):451–8.

    Article  CAS  PubMed  Google Scholar 

  51. Jensen T, Kierulf P, Sandset PM, Klingenberg O, Joo GB, Godal HC, Skjonsberg OH. Fibrinogen and fibrin induce synthesis of proinflammatory cytokines from isolated peripheral blood mononuclear cells. Thromb Haemost. 2007;97(5):822–9.

    Article  CAS  PubMed  Google Scholar 

  52. Jimenez JJ, Iribarren JL, Brouard M, Hernandez D, Palmero S, Jimenez A, Lorente L, Machado P, Borreguero JM, Raya JM, et al. Safety and effectiveness of two treatment regimes with tranexamic acid to minimize inflammatory response in elective cardiopulmonary bypass patients: a randomized double-blind, dose-dependent, phase IV clinical trial. J Cardiothorac Surg. 2011;6:138.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the following funding: The Natural Science Foundation of China (81870204). Medical science and technology development foundation, Nanjing Department of Health (ZKX19017). Project of Modern Hospital Management and Development Institute, Nanjing University and Aid project of Nanjing Drum Tower Hospital Health Education & Research Foundation (NDYG2019018, NDYG2019014).

Author information

Authors and Affiliations

Authors

Contributions

LW, RH, and BX contributed to the conception and design of the study; RH contributed to manuscript writing; QD and LC contributed to data collection and management; LC, QD, and ZYW contributed to the statistical analysis; JZC, HYZ, and LH participated in the patient follow-up; LW and BX contributed to manuscript revision and data review. All authors have read and approved the manuscript.

Corresponding authors

Correspondence to Biao Xu or Lian Wang.

Ethics declarations

Ethics approval and consent to participate

The data collection for the study was approved by the Medical Ethics Committee of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School (2021–532-02). All participants verbally consented to participate. Our database is not open to the public since the privacy of the participants should be protected.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Huang, R., Dai, Q., Chang, L. et al. The association between fibrinogen-to-albumin ratio (FAR) and adverse prognosis in patients with acute decompensated heart failure at different glucose metabolic states. Cardiovasc Diabetol 21, 241 (2022). https://doi.org/10.1186/s12933-022-01662-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12933-022-01662-x

Keywords