Back to Journals » Cancer Management and Research » Volume 14

Fibrinogen–Albumin Ratio Index Exhibits Predictive Value of Neoadjuvant Chemotherapy in Osteosarcoma

Authors Li Z, Zhou C, Peng Q, Wang SG , Qian G, Tang L , Zhou X, Yang Q, Shen Z, Huang GZ, Wang Y, Li H 

Received 19 January 2022

Accepted for publication 15 April 2022

Published 5 May 2022 Volume 2022:14 Pages 1671—1682

DOI https://doi.org/10.2147/CMAR.S358310

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Dr Antonella D'Anneo



Zhendong Li,1,* Chenliang Zhou,2,* Qing Peng,3,* Suguo Wang,3 Guowei Qian,2 Lina Tang,2 Xin Zhou,4 Qingcheng Yang,1 Zan Shen,2 GaoZhong Huang,3 Yonggang Wang,2 Hongtao Li2

1Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China; 2Department of Oncology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China; 3Department of VIP Clinic, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China; 4Cancer Research Institute of Jilin University, the First Hospital of Jilin University, Changchun, Jilin, 130021, People’s Republic of China

*These authors contributed equally to this work

Correspondence: Yonggang Wang; Hongtao Li, Department of Oncology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, No. 600, Yishan Road, Xu Hui District, Shanghai, 200233, People’s Republic of China, Tel +086-021-24058408, Email [email protected]; [email protected]

Purpose: Inflammatory response and nutritional status are associated with cancer development and progression. The present study aimed to evaluate the predictive ability of the fibrinogen–albumin ratio index (FARI) to the efficacy of neoadjuvant chemotherapy (NAC) for osteosarcoma.
Patients and methods: A retrospective analysis involving 752 consecutive osteosarcoma patients between 2012 and 2020 was performed. Data on serum fibrinogen, albumin levels, white blood cell count, platelet count, and alkaline phosphatase (ALP) before and after NAC were collected. The predictive value of the NAC efficacy in osteosarcoma was assessed by constructing a receiver operating characteristic (ROC) curve and calculating the area under the curve (AUC). Prognosis and its predictive factors were analyzed by Kaplan–Meier method and COX regression analysis. Nomogram was established according to selected variables. The predictive performance of the nomogram model was assessed using C-statistics.
Results: A total of 203 patients were included. ROC analysis showed that both FARI before NAC (preFARI; AUC = 0.594, p = 0.032) and the change in FARI before and after NAC (dfFARI = preFARI-postFARI; AUC = 0.652, p = 0.001) exhibited more favorable predictive ability than ALP and other inflammation markers. The preFARI was divided into the high group (> 6.1%) and the low group (≤ 6.1%) based on the optimal cut-off value of 6.1%. Patients with a high preFARI showed significantly decreased metastasis-free survival (MFS) and disease-free survival (DFS) (all p< 0.01). In multivariable analysis, preFARI was an independent prognostic marker for patients with osteosarcoma. Predictive nomograms exhibited good ability to predict MFS (C-index = 0.748, se = 0.028) and DFS (C-index=0.727, se = 0.030).
Conclusion: Our findings indicated that FARI exhibits the favorable predictive ability for the efficacy of NAC for osteosarcoma, which could support clinicians and patients in clinical decision-making and treatment optimization.

Keywords: osteosarcoma, neoadjuvant chemotherapy, fibrinogen, albumin, fibrinogen–albumin ratio index, prognosis

Introduction

Osteosarcoma is the most common type of bone cancer in children and adolescents,1 with an annual incidence of approximately 8–11 per million.2,3 Although surgery combined with chemotherapy and other multidisciplinary therapies can significantly improve the prognosis of patients compared with that of the previous treatments alone, the 5-year survival rate for patients with osteosarcoma is still less than 70%.4 It is reported that neoadjuvant chemotherapy (NAC) has shown great efficacy for osteosarcoma, and the 5-year overall survival (OS) has astonishingly increased to 60–70%.5 NAC-induced tumor tissue necrosis rate based on pathological grading is the gold standard for assessing the effectiveness of NAC, which is also a prognostic factor for disease progression.6–8 However, this crucial prognostic factor becomes relevant only after surgery. Accumulating evidence has shown that serum markers have prognostic value and are useful in monitoring responses to therapies; nevertheless, there are no guidelines recommended in clinical practice.9,10

Significant differences in inflammatory proteins such as fibrinogen (FIB) and albumin (ALB) have been widely researched in relation to cancer survival and repeatedly proven to be an independent prognostic factor.11,12 ALB is an important acute-phase protein reflecting not only the inflammatory state but also the nutritional status.13 FIB, as an essential acute-phase protein, plays a significant regulatory role in both the systemic inflammatory response and cancer progression, including proliferation, angiogenesis and metastasis of tumor cells.14 The fibrinogen–albumin ratio index (FARI) has been proposed as a low-cost and widely used marker to predict cancer prognosis. Recent evidence showed that FARI has potential value in the early detection of cancer, and the significantly higher FARI was found in colorectal cancer patients compared with healthy controls.15 Moreover, increasing research suggests that FARI is also a good prognostic factor for various tumors, such as gastric tumor,16,17 non-small cell lung tumor,18 gallbladder tumor,19 prostate tumor,20 breast tumor,21 and hepatocellular tumor.22 In colorectal and gallbladder cancer patients, higher FARI scores are observed in patients with unfavorable overall survival.19,23,24 Thus, we speculate that FARI might be an effective prognostic indicator for cancer. Is there a certain correlation between FARI (fibrinogen and aluminum) and osteosarcoma? Although sarcoma and carcinoma are distinguishable in derivation, biological behavior, and microenvironment,25 the study reported that FARI showed a similar association in sarcoma patients as it is in carcinoma patients.26 Specifically, in operable sarcoma, patients with increased FARI had a shorter median survival than those with a low FARI.26 However, sarcomas are a heterogeneous group that arises in soft tissue and bones,27,28 resulting in remarkable differences in disease presentation, management, and survival. Therefore, FARI might have different roles in osteosarcoma patients in the context of NAC. Unfortunately, few studies have reported the role of FARI in prognosis and the prediction of response for NAC in osteosarcoma patients.

Herein, we conducted a retrospective study to assess the predictive value of FARI in terms of prognosis and NAC efficacy in osteosarcoma patients, and to compare it with alkaline phosphatase (ALP) and established systemic inflammation markers, including neutrophil–lymphocyte ratio (NLR), lymphocyte–monocyte ratio (LMR), platelet–lymphocyte ratio (PLR), and systemic immune–inflammation index (SII).

Methods

Study Population

In this retrospective study, patients with pathologically confirmed osteosarcoma and who have been treated NAC at the Shanghai Sixth People’s Hospital from August 2011 to April 2020 were screened for inclusion in this study. Patients were required to have the complete data of the blood chemical analysis before NAC (pre-) and after NAC (post-), including serum fibrinogen, albumin levels, white blood cell (WBC) count, platelet (PLT) count, and ALP levels. Patients with infection, liver disease, abnormal coagulation function, direct surgical treatment, other malignant tumors, infused albumin or received blood transfusion before collecting blood sample, mental illness or cognitive impairment were not ineligible for our study. Additionally, we excluded the patients who had a history of oral anticoagulants or acetylsalicylic acid preparations within 3 months before admission. Patients who did not follow-up data and complete medical records were also excluded. The study was approved by the Institutional Review Board of Shanghai Jiao Tong University Affiliated Sixth People’s Hospital (Approval no. 2021–080) and all participants signed a written informed consent in accordance with the Declaration of Helsinki.

Diagnosis and Grouping

The diagnosis of osteosarcoma was confirmed by a pathologic biopsy, and the disease was staged as per the Enneking stage.29 The tumor necrosis rate was categorized as two grades: a high tumor necrosis rate (≥90%) was considered as a good response, and a low tumor necrosis rate (<90%) as a poor response.30 Patients were classified as two subgroups (good response group and poor response) to evaluate the association between measurement indicators and response to NAC.

Measurement Indicators

The blood chemical analysis was performed using fully automatic biochemical analyzer (Hitachi 008AS) and hematology analyzer (Sysmex XN 9000) at the Department of Laboratory Science, Shanghai Sixth People’s Hospital. All data from the blood chemical analysis before the first NAC dose and after the last NAC dose were collected, including the levels of serum fibrinogen, albumin, WBC, PLT, and ALP. The specific reference value range of each indicator was listed in the Appendix table Inflammatory indicators were calculated using the following formulas: FARI = (fibrinogen/albumin) × 100%; NLR = (neutrophil count/lymphocyte count); LMR = (lymphocyte count/monocyte count); PLR = (platelet count/lymphocyte count); SII = (PLT count × NLR). Differences in FARI (dfFARI) and ALP (dfALP) between pre-NAC and post-NAC were calculated as pre-levels minus post-levels. The optimal cut-off values for these measurement indicators were determined by the X-tile software.31 Then, patients were stratified into several subgroups according to cut-off values of different measurement indicators.

Follow-Up

Follow-up appointments were made at Shanghai Sixth People’s Hospital. Follow-up visits were conducted every 6 weeks to 3 months during the first 2 years, and every 3 weeks to 4 months during the second 2 years, every 2 to 6 months during the subsequent 5 years, and every 6 to 12 months thereafter. The follow-up visit includes medical history, physical examination, radiological assessments (X-ray of the primary site and chest X-ray), etc. Metastasis-free survival (MFS) was defined as the time from the date of osteosarcoma excision to the date of tumor metastasis or the date of last follow-up visit. Disease-free survival (DFS) was defined as the time from the date of osteosarcoma excision to the date of tumor recurrence/metastasis or the date of last follow-up visit.

Statistical Analysis

Statistical analyses were carried out by using SPSS 26.0 software, GraphPad Prism 8.0.2, X-tile Software version 3.6.1 and R4.2 software. Continuous variables were expressed as median (interquartile range [IQR]) and compared using the Mann–Whitney U-test (age, preALP, preNLR, preLMR, prePLR, and preSII) or Students’t-tests according to the normality of data. Categorical variables were expressed as numbers (%) and compared using two-sided Chi-square (χ2) or Fisher’s exact tests. The predictive value of the NAC efficacy in osteosarcoma was assessed by constructing a receiver operating characteristic (ROC) curve and calculating the area under the curve (AUC). MFS and DFS rates were estimated using Kaplan–Meier method, and the between-group comparison was conducted using the Log rank test. The independent prognosis factors for the MFS and DFS of osteosarcoma were analyzed using COX regression analysis and presented by plotting forest diagrams. The independent influencing factors were used for creating a prediction model of prognosis in osteosarcoma patients; the nomogram was then constructed based on the selected variables visually, and the prediction model’s performance was further evaluated through Harrell’s C-statistic (C-index). P-values <0.05 were considered to be of statistical significance.

Results

Patient Characteristics

Between August 2011 and April 2020, 752 consecutive patients with osteosarcoma were screened, and a total of 203 patients meeting the inclusion criteria were included. Of the 203 patients included, the majority were male (62.1%) with a median age of 17 years (IQR 14–22 years). One hundred and thirty-three patients (65.5%) had primary tumors located in the femur, while the remaining 70 patients (34.5%) had primary tumors located in the tibia. The median baseline preFARI, preNLR, preLMR, prePLR, preSII, and preALP values were 6.2% (IQR, 5.3–7.5%), 1.9 (IQR, 1.5–2.4), 4.6 (IQR, 3.6–6.0), 133.5 (IQR, 104.7–170.0), 514.3 (IQR, 375.0–706.9), and 163 U/L (IQR, 104 U/L-279 U/L), respectively. At the last follow-up, 13 (13.5%) had recurrence, and 45 (46.9%) had tumor metastasis. The majority of patients (68.5%) had a necrosis rate of <90% and was determined as poor response. Additionally, 31.5% of patients had a necrosis rate of ≥90% and was determined to have a good response. The detailed baseline characteristics of the included patients are shown in Table 1.

Table 1 Baseline Characteristics of Included Patients

The FARI is Associated with Better Response

Prior to NAC, the mean FARI (p=0.038, Figure 1A) was significantly higher in the good response group than that in the poor response group. There were no significant differences between the two groups regarding NLR (p=0.653, Figure 1B), LMR (p=0.430, Figure 1C), PLR (p=0.059, Figure 1D), SII (p=0.480, Figure 1E), and ALP (p=0.072, Figure 1F).

Figure 1 The distribution of FARI (A), NLR (B), LMR (C), PLR (D), SII (E), and ALP (F) before NAC, FARI (I) and ALP (J) after NAC in the good and poor response groups. The change of distribution of FARI (G) and ALP (H) between before NAC and after NAC.

Abbreviations: ALP, alkaline phosphatase; FARI, the fibrinogen–albumin ratio index; NLR, neutrophil–lymphocyte ratio; LMR, lymphocyte–monocyte ratio; PLR, platelet–lymphocyte ratio; SII, systemic immune-inflammation index.

With regard to the change of FARI and ALP levels before and after NAC, we found that both the mean FARI (p=0.002, Figure 1G) and ALP (p<0.001, Figure 1H) levels significantly decreased after NAC. Similarly, the mean FARI (p=0.001, Figure 1I) was significantly higher in the good response group compared with the poor response group, while no significant differences in the mean ALP (p=0.265, Figure 1J) were found between the two groups. The results above indicate that preFARI and dfFARI are associated with better response, having the potential to be effective predictive indicators of NAC.

Cut-off Value

Through the X-tile software, cutoff values of preFARI, preALP, preNLR, preLMR, prePLR, and preSII were confirmed to be 6.1%, 393 U/L, 3.1, 3.1, 94, and 544, respectively. Meanwhile, the cutoff values of dfFARI and dfALP was −0.3 and 121 U/L, respectively. According to the cut-off value, patients were divided into two subgroups: high-preFARI group with an FARI of >6.1% (n = 107, 52.7%) and low-preFARI group with an FARI of ≤6.1% (n = 96, 47.3%).

With regard to tumor size and disease stage, both large-tumor (≥10; 42.0% vs 26.0%; p=0.017) and high-tumor necrosis rate (≥90%; 37.4% vs 25.0%; p=0.032) patients in the high-preFARI group were more than the low-preFARI group. Meanwhile, we also found that the high-preFARI group has more patients with no metastasis compared with the low-preFARI group (72.9% vs 53.1%; p=0.004) (Table 1).

Predictive Value of Markers to NAC Response

To verify the predictive potential of these markers to NAC response, the ROC analysis was performed and AUC was calculated. As shown in Figure 2, the AUC values of preFARI and dfFARI were 0.594 (95% CI: 0.511–0.676; p = 0.032) and 0.652 (95% CI: 0.571–0.733; p = 0.001), respectively, which were greater than that of preALP (AUC = 0.452; 95% CI: 0.366–0.537; p = 0.269), preNLR (AUC = 0.480; 95% CI: 0.397–0.563; p = 0.652), preLMR (AUC = 0.465; 95% CI: 0.385–0.546; p = 0.429), prePLR (AUC = 0.583; 95% CI: 0.499–0.666; p=0.059), or preSII (AUC = 0.531; 95% CI: 0.449–0.614; p=0.476). These results indicated that FARI had a better ability in preoperative prediction for the NAC response in osteosarcoma patients.

Figure 2 ROC curves for serum markers to discriminate patients’ response to NAC.

Kaplan-Meier Survival Analysis

First, we confirmed that patients with good response to NAC have favorable MFS and DFS than patients with poor response (Figure 3A and B). Therewith, we compared the survival outcomes between subgroups (high-preFARI vs low-preFARI; high-preALP vs low-preALP) to verify the predictive performance of FARI and preALP. The Log rank test demonstrated that patients with high preFARI have superior MFS and DFS (Figure 3C and D). With regard to the preALP, the analysis was conducted on 148 patient cohorts, which only included patients with elevated ALP before NAC. As a result, patients with high preALP (>224 U/L) have shorter MFS and DFS than those with low preALP (≤224 U/L) (Figure 3E and F).

Figure 3 Survival analysis of osteosarcoma patients according to different variables. Kaplan-Meier survival curves of MFS (A) and DFS (B) in the two response groups, MFS (C) and DFS (D) in the high and low FARI groups, MFS (E) and DFS (F) in the high and low ALP groups.

Abbreviations: MFS, metastasis-free survival; DFS, disease-free survival; FARI, the fibrinogen–albumin ratio index.

Univariable and Multivariable Analyses

In terms of MFS, univariate analysis showed that the gender (HR: 0.551, 95% CI: 0.327–0.929; p=0.025), tumor diameter (HR: 1.914, 95% CI: 1.211–3.025; p=0.005), necrosis rate (HR: 1.994, 95% CI: 1.157–3.437; p=0.013), preFARI (HR: 0.473, 95% CI: 0.296–0.756; p=0.002), preALP (HR: 3.323, 95% CI: 1.977–5.587; p<0.001), and preNLR (HR: 1.98, 95% CI: 1.039–3.775; p=0.038) were significantly associated with MFS. Further, we performed a multivariate analysis on the factors with p<0.1 in the univariate analysis. Multivariate analysis confirmed that tumor diameter (HR: 2.173, 95% CI: 1.314–3.592; p=0.002), necrosis rate (HR: 1.908, 95% CI: 1.087–3.348; p=0.024), preALP (HR: 1.997, 95% CI: 1.128–3.535; p=0.018) and preFARI (HR: 0.470, 95% CI: 0.285–0.777; p=0.003) were independent predictors of MFS (Figure 4A). The high preFARI level is associated with longer MFS. Similar findings were observed in the COX regression analysis for DFS (Figure 4B). The results showed that the high-preFARI (HR: 0.410, 95% CI: 0.258–0.652; p<0.001), good response (necrosis rate ≥90%; HR: 1.931, 95% CI: 1.128–3.306; p=0.016), female (HR: 0.565, 95% CI: 0.342–0.935; p=0.026), and small tumor diameter (<10; HR: 2.857, 95% CI: 1.185–4.498; p<0.001) were significantly associated with favorable DFS.

Figure 4 Univariate and multivariate analyses for metastasis-free survival (A) and disease-free survival (B).

Construction of the Nomogram Prediction Model

A nomogram model was constructed based on independent influencing factors (tumor response, preFARI, preALP, tumor diameter, and gender) selected in the above analysis to predict 1-, 3- and 5-year survival rates for osteosarcoma. By Harrell’s C-statistic test, the nomogram model showed significant performance for predicting MFS (C-index=0.748; se = 0.028, Figure 5A) and DFS (C-index = 0.727, se = 0.030; Figure 5B). Each level of each variable was assigned a grade score, and the total scores of individuals were calculated accordingly based on the scores corresponding to different values of each variable, and then the 1-, 3- and 5-year survival rates of individuals obtained. The higher total score represents longer MFS and DFS. For example, if the patient is a female, with tumor diameter <10 cm, poor response and preFARI ≤6.1%, the probability of 1-year, 3-year and 5-year DFS is estimated to be 75%, 47% and 35%, respectively (Figure 5C).

Figure 5 Nomogram to predict the probabilities of metastasis-free survival (A) and disease-free survival (B) of osteosarcoma. The example of nomogram in a female patient (C).

Discussion

In this study, the roles of FARI in patients with osteosarcoma around the knee were investigated. The results suggested that a higher level of FARI was observed in the large-tumor and good response group. And pretreatment FARI (preFARI) exhibited better performance in predicting tumor response to chemotherapy and showed a significant association with both MFS and DFS, indicating that it is an independent prognostic factor. To our knowledge, this was the first study to assess the predictive value of FARI for the effectiveness of NAC for osteosarcoma.

Consistent with previous studies reporting that the higher FARI was correlated with larger tumors,15,21,23,26,32 we also found a higher level of FARI in tumor size ≥10 cm group. Regarding tumor stage, high FARI levels were associated with advanced stage of colorectal, breast, and soft tissue sarcoma,15,21,23,26,32 but not related to hepatocellular cancer,22 indicating the association might vary depending on different cancer stage criteria. In the cohort of this study, no significant difference was observed between stage IIA and IIB. This might be due to unique Enneking stage criteria in bone cancer, which stratify disease by the pathological grade, the anatomic setting, and the presence of metastasis, without considering tumor size.33

The relationship between FARI and treatment response was investigated in a handful of reports,32,34 in which low pretreatment FARI levels were more likely to be observed in good-response patients with colorectal cancer.32,34 In contrast to these published results, we found the higher FARI levels in good-response patients than those in poor-response patients. This discrepancy might result from the following aspects: 1) age: the median age was 60 in other reports,32,34 while the median age was 17 in our cohort; 2) response evaluation system: tumor regression grade (TRG) was used in colorectal cancer,32 while tumor necrosis rate was applied in the present study.30

We found that preFARI exhibited more reliable and greater predictive ability than ALP and other markers like NLR, LMR, PLR, and SII. ALP is the most common marker in osteosarcoma, but it is elevated in 60% of patients.35–37 In our study, ROC analysis showed that FARI had a better performance than ALP in predicting tumor response to chemotherapy, suggesting that combined serum markers might be superior to a single serum item as a predictor.

In this study, the cut-off value for preFARI was set at 6.1%, while in previous studies, the cut-off value was 6 to 11,22,23,26,34,38,39 and reasons of this difference caused might be due to different study participants among these researches. Surprisingly, we found that the high-FARI (>6.1%) group showed a favorable MFS and DFS than the low-FARI group. These results were totally opposite to previous reports, in which the high-FARI group had unfavorable survival than the low-FARI group.22,23,26,34,38,39 To validate our findings, we provided comparisons of MFS and DFS based on gender, tumor size, tumor response, and ALP. These results were strongly consistent with large cohort studies in osteosarcoma6–8,40,41 and previous studies from our institute,42 indicating that data from the cohort of this study was reliable and replicable. Therefore, the opposing result of FARI might be due to different tumor types, even including operable soft tissue sarcoma.26

Finally, prognostic factors were evaluated using univariate and multivariate Cox regression analysis, which were applied to construct nomograms. The constructed nomograms were used to assess the 1-, 3- and 5-year MFS and DFS based on the identified prognostic factors. In our study, four independent prognostic factors for MFS (sex, tumor diameter, chemotherapy response, preFARI and preALP) were identified, which also significantly impact DFS, except for preALP. To test the reliability of the nomogram for prediction prognosis, we used Harrell’s C-statistic to test the accuracy of the prediction model. The C-index, index of concordance, evaluates the predictive accuracy of a model by assessing how well the model’s predictions match the actual observations.43,44 The C-indexes for MFS and DFS were 0.7478 (se = 0.02751) and 0.7265 (se = 0.02792), respectively, indicating good performances of the nomograms.

Several limitations associated with the present study warrant mention. First, this study is a single-center retrospective study with a limited sample size, leading to selection bias. Second, information on the operation approach and adjuvant therapy was incomplete, which could lead to potential biases, and inflammatory markers associated with C-reactive protein were not analyzed in this study as that data was not collected during routine preoperative examinations. Moreover, we did not perform an overall survival analysis due to the reluctance of families of deceased patients (the majority of the deceased were children, and the parents did not want to look back again and answer any questions at the time of telephone follow-up). Therefore, further studies are needed to confirm the results of this study.

Conclusions

In summary, preFARI and dfFARI have remarkable performance in predicting tumor response to neoadjuvant chemotherapy, and preFARI is an independent prognostic marker for metastasis-free and disease-free survival in osteosarcoma patients. Thus, FARI contributes to clinical decision-making and treatment optimization, serving as a promising predictive marker for NAC in osteosarcoma patients. Validation studies or large prospective studies are necessary to further validate our findings.

Data Sharing Statement

All data generated or analyzed during this study are included in this published article.

Ethics Approval and Informed Consent

The study was approved by the Institutional Review Board of Shanghai Jiao Tong University Affiliated Sixth People’s Hospital (Approval no. 2021-080). All participants were informed as to the purpose of this study and that this study complied with the Declaration of Helsinki.

Consent for Publication

All of the authors have agreed to the publication of the work.

Acknowledgments

We would like to thank Jun He for her valuable suggestions for our article and the anonymous reviewers at Professional Editors for reading and revising it.

Author Contributions

Conception and design: Zhendong Li and Chengliang Zhou.

Provision of materials and samples: Qing Peng and Suguo Wang.

Data collection and collation: Guowei Qian, Lina Tang, Xin Zhou and Qingcheng Yang.

Data analysis and interpretation: Zan Shen, GaoZhong Huang, Yonggang Wang and Hongtao Li. All authors contributed to data analysis, drafting or revising the article, have agreed on the journal to which the article will be submitted, gave final approval of the version to be published, and agree to be accountable for all aspects of the work.

Funding

This work was supported by China Medicine Education Association (CMEA) (No. 2020KTS012), the National Natural Scientific Foundation of China (NSFC) (No. 82002962; 81900189). The funder has no role in the study design, data collection and analysis, decision to publish, or manuscript preparation.

Disclosure

The authors declare that they have no competing interests in this work.

References

1. Harrison DJ, Geller DS, Gill JD, Lewis VO, Gorlick R. Current and future therapeutic approaches for osteosarcoma. Expert Rev Anticancer Ther. 2018;18:39–50.

2. Ritter J, Bielack SS. Osteosarcoma. Ann Oncol. 2010;21(Suppl 7):vii320–325.

3. Stiller CA, Bielack SS, Jundt G, Steliarova-Foucher E. Bone tumours in European children and adolescents, 1978-1997. Report from the Automated Childhood Cancer Information System project. Eur J Cancer. 2006;42:2124–2135.

4. Ji S-Y, Tang H-J, Luo X-T, et al. The Prognostic Value of A Preoperative Lymphocyte Count and C-Reactive Protein Ratio. Osteosarcoma. 2021;1:65.

5. Whelan J, Seddon B, Perisoglou M. Management of osteosarcoma. Curr Treat Options Oncol. 2006;7:444–455.

6. Whelan JS, Jinks RC, McTiernan A, et al. Survival from high-grade localised extremity osteosarcoma: combined results and prognostic factors from three European Osteosarcoma Intergroup randomised controlled trials. Ann Oncol. 2012;23:1607–v.

7. Bacci G, Longhi A, Versari M, et al. Prognostic factors for osteosarcoma of the extremity treated with neoadjuvant chemotherapy: 15-year experience in 789 patients treated at a single institution. Cancer. 2006;106:1154–1161.

8. Bielack SS, Kempf-Bielack B, Delling G, et al. Prognostic factors in high-grade osteosarcoma of the extremities or trunk: an analysis of 1702 patients treated on neoadjuvant cooperative osteosarcoma study group protocols. J Clin Oncol. 2002;20:776–790.

9. Evola FR, Costarella L, Pavone V, et al. Biomarkers of Osteosarcoma, Chondrosarcoma, and Ewing Sarcoma. Front Pharmacol. 2017;8:150.

10. Savitskaya YA, Rico-Martínez G, Linares-González LM, et al. Serum tumor markers in pediatric osteosarcoma: a summary review. Clin Sarcoma Res. 2012;2:9.

11. Cai W, Kong W, Dong B, et al. Pretreatment Serum Prealbumin as an Independent Prognostic Indicator in Patients With Metastatic Renal Cell Carcinoma Using Tyrosine Kinase Inhibitors as First-Line Target Therapy. Clin Genitourin Cancer. 2017;15:e437–e446.

12. Kelly P, Paulin F, Lamont D, et al. Pre-treatment plasma proteomic markers associated with survival in oesophageal cancer. Br J Cancer. 2012;106:955–961.

13. Gupta D, Lis CG. Pretreatment serum albumin as a predictor of cancer survival: a systematic review of the epidemiological literature. Nutr J. 2010;9:69.

14. Perisanidis C, Psyrri A, Cohen E, et al. Prognostic role of pretreatment plasma fibrinogen in patients with solid tumors: a systematic review and meta-analysis. Cancer Treat Rev. 2015;41:960–970.

15. Sun F, Tan YA, Gao QF, et al. Circulating fibrinogen to pre-albumin ratio is a promising biomarker for diagnosis of colorectal cancer. J Clin Lab Anal. 2019;33:e22635.

16. Zhang L, Wang Z, Xiao J, et al. Prognostic value of fibrinogen-to-albumin ratio in patients with gastric cancer receiving first-line chemotherapy. Oncol Lett. 2020;20:10.

17. Zhang J, Ruan J, Wang W, et al. Prognostic Value of the Combination of CEA and Fibrinogen/Albumin Ratio in Resectable Gastric Cancer. Cancer Manag Res. 2020;12:2767–2775.

18. Li SQ, Jiang YH, Lin J, et al. Albumin-to-fibrinogen ratio as a promising biomarker to predict clinical outcome of non-small cell lung cancer individuals. Cancer Med. 2018;7:1221–1231.

19. Xu WY, Zhang HH, Xiong JP, et al. Prognostic significance of the fibrinogen-to-albumin ratio in gallbladder cancer patients. World J Gastroenterol. 2018;24:3281–3292.

20. Man YN, Chen YF. Systemic immune-inflammation index, serum albumin, and fibrinogen impact prognosis in castration-resistant prostate cancer patients treated with first-line docetaxel. Int Urol Nephrol. 2019;51:2189–2199.

21. Hwang KT, Chung JK, Roh EY, et al. Prognostic Influence of Preoperative Fibrinogen to Albumin Ratio for Breast Cancer. J Breast Cancer. 2017;20:254–263.

22. Xu Q, Yan Y, Gu S, et al. A Novel Inflammation-Based Prognostic Score: the Fibrinogen/Albumin Ratio Predicts Prognoses of Patients after Curative Resection for Hepatocellular Carcinoma. J Immunol Res. 2018;2018:4925498.

23. Wang YY, Liu ZZ, Xu D, et al. Fibrinogen-Albumin Ratio Index (FARI): a More Promising Inflammation-Based Prognostic Marker for Patients Undergoing Hepatectomy for Colorectal Liver Metastases. Ann Surg Oncol. 2019;26:3682–3692.

24. Dolan RD, McSorley ST, Park JH, et al. The prognostic value of systemic inflammation in patients undergoing surgery for colon cancer: comparison of composite ratios and cumulative scores. Br J Cancer. 2018;119:40–51.

25. Saggioro M, D’Angelo E, Bisogno G, Agostini M, Pozzobon M. Carcinoma and Sarcoma Microenvironment at a Glance: where We Are. Front Oncol. 2020;10:76.

26. Liang Y, Wang W, Que Y, et al. Prognostic value of the fibrinogen/albumin ratio (FAR) in patients with operable soft tissue sarcoma. BMC Cancer. 2018;18:942.

27. Liegl-Atzwanger B. The role of pathology in sarcoma. Magazine Eur Med Oncol. 2020;13:159–163.

28. Ducimetière F, Lurkin A, Ranchère-Vince D, et al. Incidence of Sarcoma Histotypes and Molecular Subtypes in a Prospective Epidemiological Study with Central Pathology Review and Molecular Testing. PLoS One. 2011;6:e20294.

29. Cao S, Jiang L, Shen L, Xiong Z. Role of microRNA-92a in metastasis of osteosarcoma cells in vivo and in vitro by inhibiting expression of TCF21 with the transmission of bone marrow derived mesenchymal stem cells. Cancer Cell Int. 2019;19:31.

30. Raymond AK, Chawla S, Carrasco C, et al. Osteosarcoma chemotherapy effect: a prognostic factor. Semin Diagn Pathol. 1987;4:212–236.

31. Camp RL, Dolled-Filhart M, Rimm DL. X-tile: a new bio-informatics tool for biomarker assessment and outcome-based cut-point optimization. Clin Cancer Res. 2004;10:7252–7259.

32. Lu S, Liu Z, Zhou X, et al. Preoperative Fibrinogen-Albumin Ratio Index (FARI) is a Reliable Prognosis and Chemoradiotherapy Sensitivity Predictor in Locally Advanced Rectal Cancer Patients Undergoing Radical Surgery Following Neoadjuvant Chemoradiotherapy. Cancer Manag Res. 2020;12:8555–8568.

33. Enneking WF, Spanier SS, Goodman MA. A system for the surgical staging of musculoskeletal sarcoma. Clin Orthop Relat Res. 1980;1:106–120.

34. Zhang L, Zhang J, Wang Y, et al. Potential prognostic factors for predicting the chemotherapeutic outcomes and prognosis of patients with metastatic colorectal cancer. J Clin Lab Anal. 2019;33:e22958.

35. Shimose S, Kubo T, Fujimori J, Furuta T, Ochi M. A novel assessment method of serum alkaline phosphatase for the diagnosis of osteosarcoma in children and adolescents. J Orthop Sci. 2014;19:997–1003.

36. Khoury JF, Ben-Arush MW, Weintraub M, et al. Alkaline phosphatase level change in patients with osteosarcoma: its role as a predictive factor of tumor necrosis and clinical outcome. Isr Med Assoc J. 2014;16:26–32.

37. Ren HY, Sun LL, Li HY, Ye ZM. Prognostic Significance of Serum Alkaline Phosphatase Level in Osteosarcoma: a Meta-Analysis of Published Data. Biomed Res Int. 2015;2015:160835.

38. You X, Zhou Q, Song J, et al. Preoperative albumin-to-fibrinogen ratio predicts severe postoperative complications in elderly gastric cancer subjects after radical laparoscopic gastrectomy. BMC Cancer. 2019;19:931.

39. Wu M, Pan Y, Jia Z, et al. Preoperative Plasma Fibrinogen and Serum Albumin Score Is an Independent Prognostic Factor for Resectable Stage II-III Gastric Cancer. Dis Markers. 2019;2019:9060845.

40. Collins M, Wilhelm M, Conyers R, et al. Benefits and adverse events in younger versus older patients receiving neoadjuvant chemotherapy for osteosarcoma: findings from a meta-analysis. J Clin Oncol. 2013;31:2303–2312.

41. Ferrari S, Bertoni F, Mercuri M, et al. Predictive factors of disease-free survival for non-metastatic osteosarcoma of the extremity: an analysis of 300 patients treated at the Rizzoli Institute. Ann Oncol. 2001;12:1145–1150.

42. Li H, Wang Y, Liu Z, et al. Lack of association between platelet indices and disease stage in osteosarcoma at diagnosis. PLoS One. 2017;12:e0174668.

43. Peng B, Min R, Liao Y, Yu A. Development of Predictive Nomograms for Clinical Use to Quantify the Risk of Amputation in Patients with Diabetic Foot Ulcer. J Diabetes Res. 2021;2:6621035.

44. Qian C, Shu W, Jing-He L. Development and validation of Nomograms for predicting overall survival and Cancer-specific survival in patients with ovarian clear cell carcinoma. J Ovarian Res. 2020;13:123.

Creative Commons License © 2022 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.