Skip to main content

Comprehensive machine learning-based preoperative blood features predict the prognosis for ovarian cancer

Abstract

Purpose

Significant advancements in improving ovarian cancer (OC) outcomes have been limited over the past decade. To predict prognosis and improve outcomes of OC, we plan to develop and validate a robust prognosis signature based on blood features.

Methods

We screened age and 33 blood features from 331 OC patients. Using ten machine learning algorithms, 88 combinations were generated, from which one was selected to construct a blood risk score (BRS) according to the highest C-index in the test dataset.

Results

Stepcox (both) and Enet (alpha = 0.7) performed the best in the test dataset with a C-index of 0.711. Meanwhile, the low RBS group possessed observably prolonged survival in this model. Compared to traditional prognostic-related features such as age, stage, grade, and CA125, our combined model had the highest AUC values at 3, 5, and 7 years. According to the results of the model, BRS can provide accurate predictions of OC prognosis. BRS was also capable of identifying various prognostic stratifications in different stages and grades. Importantly, developing the nomogram may improve performance by combining BRS and stage.

Conclusion

This study provides a valuable combined machine-learning model that can be used for predicting the individualized prognosis of OC patients.

Peer Review reports

Introduction

Ovarian cancer(OC), an aggressive gynecological cancer, has a 5-year survival rate of less than 50% and ranks the first in tumor-related deaths among gynecologic cancers in the United States [1]. 75% of epithelial ovarian cancer (EOC) patients are already in advanced stages at the time of detection due to the sneaky clinical symptoms and the lack of early screening tools [2]. Traditional FIGO stage, grade, CA-125, and tumor residuals provide a relatively reliable reference for patient treatment selection and predicting prognosis [3,4,5,6,7]. Still, the high degree of heterogeneity in EOC, even among patients at the same stage, can lead to a wide range of outcomes [8].

In recent years, treatment for ovarian cancer is no longer a ‘one-size-fits-all’ fixed treatment proposition [9, 10]. Multiple clinical trials have categorized patients into high- and low-risk cohorts [11]. In many cases, the treatment decision with PARP inhibitors or bevacizumab may be influenced by the risk stratification predicted based on these clinical and genetic factors [11]. Therefore, the construction of predictive models for EOC prognosis is essential.

Through bioinformatics research, numerous prognostic models for gene signatures have been developed. These prognostic models, while generally achieving good predictive results, lacked integrated biological signatures because they were based on gene expression files for specific biological pathways such as immune [12], metabolism [13], m6A [14], and autophagy [15]. As a result, there is a need to take into account more usable and effective clinical biomarkers for prediction. Previous research successfully predicted prognosis using various preoperative blood indicators [16, 17]. According to certain research, blood indicators may represent the tumor microenvironment. However, screening using a few blood indicators resulted in the loss of key information and was inadequate for exploring the characteristic landscapes and survival prognosis of OC patients. Machine learning (ML) has shown enormous application value in evaluating prognosis and making clinical diagnoses. Besides, ML can adequately utilize large datasets for training, which avoids loss of data. Previous studies have demonstrated the superiority of machine learning algorithms over non-machine learning algorithms [18,19,20,21,22,23,24,25]. Using a decision tree algorithm, Feng et al. [25] constructed a prediction model for EOC based on preoperative blood markers and clinicopathologic parameters, but the prediction still has greater potential for improvement. Using multiple types of machine learning algorithms, the integrated program could provide a model with consensus output for OC prognosis. And the combination of algorithms can further reduce the dimensionality of the variables, making the model more simplified and increasing accuracy. Previously, Hansen et al. identified and quantified circRNAs expression by combining two (or more) algorithms and found that algorithm combinations could improve algorithm complementarity and resolve algorithm-specific false positives [26].

Here, we utilized 88 machine learning algorithm combinations to explore prognostic stratification based on blood features to guide individualized management of EOC patients.

Methods

Study population

Figure 1 depicted the research design process schematically. Retrospective screening was performed on 443 EOC patients from Jan.2010 to Dec.2020. Exclusion criteria were as described in previous articles [27]. In addition to this, patients with no follow-up records were excluded (n = 88). Finally, a total of 331 EOC patients were matched. The original dataset (n = 331) was randomly divided into training dataset (n = 231) and test dataset (n = 100) using a 7:3 ratio. The analysis has been approved by the Ethics Committee of Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine.

Fig. 1
figure 1

Study design process

HRD status assessments

Combining the homologous recombination deficiency (HRD) score and the status of BRCA1/2 mutations is necessary to determine the HRD status. The HRD score was calculated as the sum of the loss of heterozygosity (LOH) , telomeric allelic imbalance (TAI), and large-scale state transitions (LST) scores. HRD score was tested by BGI Genomics Co., Ltd., and HRD status was considered positive if the HRD score was greater than 42 or BRCA1/2 mutations.

Prognostic signature selection and development

Based on our previous study [27], a total of 33 blood features were selected from the cohort. Next, to develop the blood risk score (BRS), we incorporated these blood features and age into our program. The preoperative blood features contained Sodium (Na), Potassium (K), Chlorine (Cl), White blood cell (WBC), Neutrophil (Neu), Lymphocyte (Lym), Hematocrit (Hct), and Platelet (PLT), Red blood cell (RBC), Hemoglobin (Hb), Monocyte (Mono), Eosinophil (Eo), Basophil (Baso), Carcinoembryonic antigen (CEA), Alpha-fetoprotein (AFP), Carbohydrate antigen 19 − 9 (CA19-9), and Carbohydrate antigen 125 (CA-125), Blood urea nitrogen (BUN), Creatinine (Cr), Uric acid (UA), Alanine aminotransferase (ALT), Aspartate aminotransferase (AST), Alkaline phosphatase (ALP), Total protein (TP), Albumin (Alb), Prealbumin (PA), globulin (GLOB), glutamyl transpeptidase (GGT), Lactate dehydrogenase (LDH), Thrombin time (TT), Prothrombin time (PT), Fibrinogen (Fb) and Activated partial thromboplastin time (APTT).

To enhance the accuracy and robustness of comprehensive and systematic approaches, we integrated 10 ML algorithms and generated 88 algorithm combinations. We used the preceding procedure to create a reliable prognosis model for ovarian cancer [28, 29]. In this study, we combined 10 classical algorithms: random forest (RSF), survival support vector machine (Survival-SVM), least absolute shrinkage and selection operator (LASSO), elastic network (Enet), gradient boosting machine (GBM), supervised principal components (SuperPC), ridge regression, partial least squares regression for Cox (plsRcox), CoxBoost, and Stepwise Cox (StepCox). Variable screening was provided by RSF, LASSO, CoxBoost, and Stepwise Cox. We constructed 88 model combinations using the framework of LOOCV based on the approach of Liu et al. [30]. Next, we constructed prognostic signatures in the training dataset using a combination of 88 models. To train and tune the models, and reduce overfitting, the original training dataset was divided into a sub-training set and a validation set through LOOCV. Specifically, in each LOOCV trial, N-1 samples were used as the sub-training set to train the models, and the remaining single sample was used as a validation set to validate the models and optimize model parameters. This process was repeated N times until each sample was used as a validation set once. When the models were obtained, we evaluated the models using the test dataset. We used prognostic models to predict the overall survival of patients. The BRS was finally estimated using the signatures gathered from the training and test cohorts. More details were shown in the Supplementary Material.

Evaluating the clinical significance of BRS

The concordance index (C-index) and the integrated Brier score (IBS), two widely used assessment metrics, were employed in the prior papers to assess the efficacy of the survival prediction model [31]. C-index is defined as the proportion of patient pairs in which the predicted and observed survival outcomes were concordant [32]. A C-index of 0.5 indicates no predictive discrimination, and a C-index of 1 indicates perfect predictive accuracy. The IBS, which represents the mean squared discrepancies between observed survival status and anticipated survival probability at a specific time point, is used to assess the error of survival prediction. An IBS value of 0 suggests perfect prediction, whereas 1 shows completely wrong prediction. By taking into account the highest C-index of the test cohort, we were able to determine the best prognostic model for OC. In addition, the Mean Square Error (MSE) of the training dataset was calculated based on the predicted results generated by each iteration of LOOCV. The MSE of the test dataset was calculated based on the final model. The smaller the MSE value, the more accurate the predicted results.

Between high- and low-risk groups, clinical parameters such as age, FIGO stage, and grade were compared. A Kaplan-Meier (KM) analysis in clinical subgroups was also conducted. To evaluate the BRS’s predictive power, receiver-operator characteristic (ROC) curves were created for the test dataset. We conducted time-dependent ROC curves and areas under the curve (AUCs) analyses of the model predictive power at 3, 5 and 7 years using the R package timeROC. We used SHAP to interpret the output of the optimal machine learning combination [33].

Construction of nomogram

Multivariate and univariate analyses were carried out using Cox’s hazards regression model. Hazard ratios (HR) were determined from Cox proportional hazards regression models. And the prognostic risk factor is indicated by an HR more than 1, whereas the protective impact is shown by an HR less than 1. The “rms” package of the R software was used to create the nomogram. To assess the discrimination of the nomogram model, time-ROC and calibration curves were used.

Statistical analysis

The R software (v.4.1.3) was used for all statistical analysis. Categorical variables were analyzed using the chi-squared or Fisher exact tests, while continuous variables were studied using the Wilcoxon rank-sum or T tests. The ROC analysis was performed using the R package “survivalROC”, and the optimal cut-off value of BRS for predicting overall survival (OS) was determined. There was statistical significance at P < 0.05.

Results

Clinical characteristics

Table 1 listed the general clinical characteristics of the EOC patients. The mean age of datasets was 57.61 ± 10.39 years old. A total of 137 (41.4%) and 194 (58.6%) patients were in early (FIGO I or II) or late (FIGO III or IV) stages of the OC. Histology-proven serous subtypes were present in 229 (70.2%) of patients. A heat map was obtained to express the results of Pearson correlation analysis of selected features (Fig. 2).

Table 1 The baseline characteristics of the EOC patients
Fig. 2
figure 2

The correlation heat map. The correlation between the biomarkers was depicted in the heatmap

Univariate and multivariate Cox analysis

We conducted univariate and multivariate Cox regression analysis in all patients to further ascertain whether these chosen features acted as an independent risk factor for the survival outcome of ovarian cancer patients (Table 2). The UA (HR = 1.0031, P = 0.0279), TP (HR = 0.9568, P = 0.0018), Alb (HR = 0.9159, P = 0.0000), AST (HR = 1.0212, P = 0.0146), PA (HR = 0.9955, P = 0.0062), LDH (HR = 1.0015, P = 0.0112), Lym (HR = 0.6378, P = 0.0435), Hct (HR = 0.0036, P = 0.0344), TT (HR = 0.8264, P = 0.0001), Fb (HR = 1.0908, P = 0.0347), and CA-125 (HR = 1.0002, P = 0.0006) were determined as significantly prognostic factors for OS through the univariate analysis (Table 2). We used multivariate cox regression analysis to adjust for any potential confounding factors that may have existed in univariate cox’s regression. Finally, UA (HR = 1.0044, P = 0.013), Alb (HR = 0.7742, P = 0.0117), TT (HR = 0.7805, P = 0.0000), and CA-125 (HR = 1.0002, P = 0.01) were independent factors for survival according to multivariate cox regression analysis.

Table 2 Univariate and multivariate cox regression analysis

Integrated development of ovarian cancer prognosis model

ML with preoperative blood metrics as input was trained to export a risk score for survival, which was used to measure the level of risk for an individual. For our training cohort, we implemented 88 algorithm combinations to acquire prediction models, then, for our test cohort, we calculated the C-index and IBS of each algorithm. Considering there were fewer independent predictors and the model had a filtering function, we did not use the above independent risk factors to train models but instead used all the characteristics.

As shown in Fig. 3A and Table S1, the combination of Stepcox (both) and Enet (alpha = 0.7) with the most prominent C-index (0.711) and the low IBS (0.169) was chosen as the final model. The mean MSE in the training dataset was 0.188, and the test dataset was 0.192. Following final model evaluation, we calculated BRS for every sample in the test cohort. The characteristics used by each model were shown in Fig. 3B. The features selected for the optimal model were TP, Alb, and TT. In Figure S1, we presented the SHAP values of features for the optimal model. BRS was categorized based on its cut-off value (0.007) into high and low groups to evaluate its prognostic performance. A KM curve for OS and RFS shows that the high BRS group had significantly shorter survival times in the test cohort (p = 0.0015 for OS and p = 0.035 for RFS, Fig. 3C and D). To measure the discrimination of BRS, we conducted the analysis of time-ROC. In the test group, the 3-, 5-, and 7-year OS of BRS had respective AUCs of 0.738, 0.781, and 0.752. which was higher than other common prognostic predictors, such as FIGO stage, HRD status, grade, age, and CA-125 (Fig. 3E).

Fig. 3
figure 3

Construction and testing of the combination machine learning model-based blood features for prognosis of OC patients. A. The C-index values for 88 ML algorithms were calculated in the test dataset. B. Selection of blood features for developing machine-learning models. C, D. KM survival analysis for overall survival (C) and recurrence free survival (D) between the high and low BRS groups in the test dataset. E. Common clinical characteristics at 3,5,7 years in the test dataset were contrasted with the predict performance of BRS.

Predictive performance in different clinical features

In order to better understand BRS, we grouped patients of the test dataset based on several clinical traits, including stage, grade and pathology type. At the same time, subgroup analysis reduced the presence of heterogeneity and allowed for more reliable prediction results. With the later FIGO stage, we discovered that the BRS significantly increased (p = 0.022), but there were no significant differences in grade and pathology type (Fig. 4A-C). Interestingly, BRS also significantly improved the capacity to distinguish overall survival in several clinical subgroups, such as stage (early and late) and G3 group, although no differentiation was demonstrated for the RFS (Fig. 4D-G). In these subgroups, high BRS represented poorer overall survival.

Fig. 4
figure 4

Performance of BRS in different subgroups. A, B, C. The distribution of BRS in stage (A), grade (B), and histologic types (C). D, E. The KM analysis of overall survival and recurrence free survival in different stage subgroups. F, G. The KM analysis of overall survival and recurrence free survival in different grade subgroups

Nomogram based on BRS and clinical features

Using univariate Cox regression analysis, we identified BRS (HR = 4.808, P = 0.04) and stage (HR = 3.621, P = 0.006) as risk factors for OS (Fig. 5A). Furthermore, through multivariate cox regression, we found that BRS (HR = 4.475, P = 0.007) and stage (HR = 3.08, P = 0.021) were independent risk factors (Fig. 5B). Given the prospective therapeutic applicability of BRS, a predictive nomogram incorporating two independent predictors of mortality (BRS and stage) was constructed (Fig. 5C). Meanwhile, personalized patient scores were computed to predict the OS at three, five, and seven years. According to the calibration plot, our nomogram performed good in predicting OC patients’ prognoses (Fig. 5D). At 3-, 5-, 7-year, the nomogram’s AUCs were 0.773, 0.821, and 0.887 (Fig. 5E), which indicated its accuracy and stability. In addition, the nomogram model had an IBS of 0.153 (Table S2). whereas the BRS has an IBS of 0.169. Thus, the excellent predictive performance of nomogram model for long-term survival was validated.

Fig. 5
figure 5

The development of nomogram. A, B. Univariate (A) and multivariate (B) cox regression analysis. C. The nomogram integrated BRS and stage was constructed. D. Calibration curves used to compare the predicted and actual 3, 5,7 years survival probabilities. E. Time-dependent receiver-operator characteristic (ROC) analysis for predicting 3-, 5-, and 7-year OS.

Discussion

Conventional features have considerable limitations in prognostic management and hazard rate estimation when taken into account in the context of tumor heterogeneity and the varied clinical outcomes of patients at the same stage. Since many patients were not analyzed genomically, many prognosis markers or predictive models could not be directly used for clinical application. To ascertain the prognosis of OC, it may be beneficial to investigate the classification and risk stratification of tumors by making adequate use of clinically available blood tests.

A growing amount of research has shown that peripheral blood test was essential for determining the prognosis of ovarian cancer [34,35,36] and other malignant cancers [37,38,39]. Preoperative blood markers can be quickly identified using standard blood testing, which is more convenient and affordable. However, preoperative peripheral blood assessment systems of ovarian cancer are not yet complete. Since 2000, there has been a tremendous improvement in the accuracy of employing ML models to predict patient survival and diagnosis [40]. Our previous article established supervised diagnostic models and unsupervised prognosis models based on age and pre-operative blood indicators. To further increase the predictive power of the model, we constructed prognostic characteristics of OC patients by combining machine learning algorithms using age and 33 blood metrics. In order to prevent unsuitable model approaches owing to personal preferences, we combined 10 machine learning algorithms into 88 combinations and chose the best model. This combined ML model approach has been used to predict the prognosis of bladder cancer [41], muscle-invasive urothelial cancer [42], pancreatic cancer [29], and endometrial cancer [43], as well as validated in multiple datasets with good robust and AUC values. Importantly, the optimal model demonstrated strong and stable prediction performance by evaluating the C-index, IBS, and mean MSE. Three, five-, and seven-year OS in the test cohort had AUCs of 0.738, 0.781, and 0.752, respectively. This predictive efficacy was superior to our risk model based on unsupervised machine learning [27]. Surprisingly, the predictive efficacy of BRS exceeded that of models integrating multi-scale clinical imaging and genomic data [44]. Some traditional clinical characteristics have been shown to be useful in the prognostic assessment of OC patients. Therefore, we contrasted the effectiveness of BRS with these clinical characteristics. Apparently, the predictive efficacy of our model was preferable to these traditional predictors, including age, pathological grade, stage, HRD status, and CA-125. We compared BRS with currently recognized prognostic biomarkers in clinical practice and guidelines, which also increased the trust of physicians in our model.

The features identified by our optimal model included TP, Alb, TT. Zhong et al. found that thrombin could induce epithelial-mesenchymal transition and promote the invasion of ovarian cancer cells [45]. A recent study has revealed the connections between OC growth and coagulation [46]. Our study emphasized the importance of TT for the prognosis of OC, which may provide new insights into the biological mechanisms of coagulation in ovarian cancer. Serum Alb level is a crucial indicator for patients’ systemic inflammatory response and nutritional condition. The relationship between Alb level and the prognosis of patients has been found in many cancers, including ovarian, colorectal, and lung cancer [47]. The effect of albumin on ovarian cancer is complex, and additional approaches are needed to explore the mechanisms.

Importantly, the stage between the high- and low-risk groups varied significantly. We discovered that as FIGO stage was raised, risk scores considerably rose. Besides, BRS significantly improved the capacity to identify different clinical subgroups’ survival statuses. Our model exhibited independent predictive performance after adjusting for stage and grade. The nomogram was further modified to increase the clinical utility of BRS. It showed higher AUC values compared to BRS alone, and exceeding the predictive power of a nomogram also based on peripheral blood features constructed by Bai et al. [48], implying a higher predictive value for prognostic prediction in OC patients, which suggested that it may be a promising alternative metric for assessing prognostic risk in clinical OC.

However, BRS still has some limitations. First, all of the samples used in our investigation were retrospective, thus prospective samples should be used in the future for BRS corroboration. Second, we accept that our work will need external validation because it was only evaluated on a dataset from one institution. Finally, the lack of investigation into therapy efficacy needs more confirmation in the future, and exploring integrated genomes and imaging models with BRS could improve risk stratification’s ability to predict outcomes.

In summary, we combined various ML methods to predict risk stratification for EOC patients, and we found that the integrated algorithms increased the efficacy of the test dataset beyond common clinical factors. Our findings promoted clinical prognostic research by multiple combination machine learning.

Data availability

These findings’ data are now being used in another study, thus, they cannot be shared. Following the publication of the paper, requests for data will be taken into consideration by the corresponding author.

Code Availability

The corresponding author can provide the code used to produce the findings in this study upon request.

References

  1. Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA Cancer J Clin. 2023;73(1):17–48.

    Article  PubMed  Google Scholar 

  2. Berek JS, Kehoe ST, Kumar L, Friedlander M. Cancer of the ovary, fallopian tube, and peritoneum. Int J Gynaecol Obstet. 2018;143(Suppl 2):59–78.

    Article  PubMed  Google Scholar 

  3. Peres LC, Cushing-Haugen KL, Kobel M, Harris HR, Berchuck A, Rossing MA, Schildkraut JM, Doherty JA. Invasive epithelial ovarian Cancer survival by Histotype and Disease Stage. J Natl Cancer Inst. 2019;111(1):60–8.

    Article  PubMed  Google Scholar 

  4. Chi DS, Eisenhauer EL, Lang J, Huh J, Haddad L, Abu-Rustum NR, Sonoda Y, Levine DA, Hensley M, Barakat RR. What is the optimal goal of primary cytoreductive surgery for bulky stage IIIC epithelial ovarian carcinoma (EOC)? Gynecol Oncol. 2006;103(2):559–64.

    Article  CAS  PubMed  Google Scholar 

  5. Wimberger P, Lehmann N, Kimmig R, Burges A, Meier W, Du Bois A. Arbeitsgemeinschaft Gynaekologische Onkologie Ovarian Cancer Study G: prognostic factors for complete debulking in advanced ovarian cancer and its impact on survival. An exploratory analysis of a prospectively randomized phase III study of the Arbeitsgemeinschaft Gynaekologische Onkologie Ovarian Cancer Study Group (AGO-OVAR). Gynecol Oncol. 2007;106(1):69–74.

    Article  PubMed  Google Scholar 

  6. Chang SJ, Bristow RE, Ryu HS. Impact of complete cytoreduction leaving no gross residual disease associated with radical cytoreductive surgical procedures on survival in advanced ovarian cancer. Ann Surg Oncol. 2012;19(13):4059–67.

    Article  PubMed  Google Scholar 

  7. Jacobs I, Bast RC Jr. The CA 125 tumour-associated antigen: a review of the literature. Hum Reprod. 1989;4(1):1–12.

    Article  CAS  PubMed  Google Scholar 

  8. Kopper O, de Witte CJ, Lohmussaar K, Valle-Inclan JE, Hami N, Kester L, Balgobind AV, Korving J, Proost N, Begthel H, et al. An organoid platform for ovarian cancer captures intra- and interpatient heterogeneity. Nat Med. 2019;25(5):838–49.

    Article  CAS  PubMed  Google Scholar 

  9. Heintz AP, Odicino F, Maisonneuve P, Quinn MA, Benedet JL, Creasman WT, Ngan HY, Pecorelli S, Beller U. Carcinoma of the ovary. FIGO 26th Annual Report on the Results of Treatment in Gynecological Cancer. Int J Gynaecol Obstet 2006, 95 Suppl 1:S161-192.

  10. Rose PG, Java JJ, Salani R, Geller MA, Secord AA, Tewari KS, Bender DP, Mutch DG, Friedlander ML, Van Le L, et al. Nomogram for Predicting Individual Survival after recurrence of Advanced-Stage, High-Grade Ovarian Carcinoma. Obstet Gynecol. 2019;133(2):245–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Chambers LM, O’Malley DM, Coleman RL, Herzog TJ. Is there a low-risk patient population in advanced epithelial ovarian cancer? A critical analysis. Am J Obstet Gynecol 2022.

  12. Shen S, Wang G, Zhang R, Zhao Y, Yu H, Wei Y, Chen F. Development and validation of an immune gene-set based Prognostic signature in ovarian cancer. EBioMedicine. 2019;40:318–26.

    Article  PubMed  Google Scholar 

  13. Zhang H, Chi M, Su D, Xiong Y, Wei H, Yu Y, Zuo Y, Yang L. A random forest-based metabolic risk model to assess the prognosis and metabolism-related drug targets in ovarian cancer. Comput Biol Med. 2023;153:106432.

    Article  CAS  PubMed  Google Scholar 

  14. Tan W, Liu S, Deng Z, Dai F, Yuan M, Hu W, Li B, Cheng Y. Gene signature of m6A-related targets to predict prognosis and immunotherapy response in ovarian cancer. J Cancer Res Clin Oncol. 2023;149(2):593–608.

    Article  CAS  PubMed  Google Scholar 

  15. Ding J, Wang C, Sun Y, Guo J, Liu S, Cheng Z. Identification of an autophagy-related signature for prognosis and Immunotherapy Response Prediction in Ovarian Cancer. Biomolecules 2023, 13(2).

  16. Marchetti C, Romito A, Musella A, Santo G, Palaia I, Perniola G, Di Donato V, Muzii L, Benedetti Panici P. Combined plasma fibrinogen and neutrophil lymphocyte ratio in Ovarian Cancer Prognosis May play a role? Int J Gynecol Cancer. 2018;28(5):939–44.

    Article  PubMed  Google Scholar 

  17. Miao Y, Yan Q, Li S, Li B, Feng Y. Neutrophil to lymphocyte ratio and platelet to lymphocyte ratio are predictive of chemotherapeutic response and prognosis in epithelial ovarian cancer patients treated with platinum-based chemotherapy. Cancer Biomark. 2016;17(1):33–40.

    Article  CAS  PubMed  Google Scholar 

  18. Enshaei A, Robson CN, Edmondson RJ. Artificial Intelligence Systems as Prognostic and Predictive tools in Ovarian Cancer. Ann Surg Oncol. 2015;22(12):3970–5.

    Article  CAS  PubMed  Google Scholar 

  19. Ow GS, Kuznetsov VA. Big genomics and clinical data analytics strategies for precision cancer prognosis. Sci Rep. 2016;6:36493.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  20. Paik ES, Lee JW, Park JY, Kim JH, Kim M, Kim TJ, Choi CH, Kim BG, Bae DS, Seo SW. Prediction of survival outcomes in patients with epithelial ovarian cancer using machine learning methods. J Gynecol Oncol. 2019;30(4):e65.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Arezzo F, Cormio G, La Forgia D, Santarsiero CM, Mongelli M, Lombardi C, Cazzato G, Cicinelli E, Loizzi V. A machine learning approach applied to gynecological ultrasound to predict progression-free survival in ovarian cancer patients. Arch Gynecol Obstet. 2022;306(6):2143–54.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Avesani G, Tran HE, Cammarata G, Botta F, Raimondi S, Russo L, Persiani S, Bonatti M, Tagliaferri T, Dolciami M et al. CT-Based Radiomics and Deep learning for BRCA mutation and progression-free survival prediction in Ovarian Cancer using a Multicentric dataset. Cancers (Basel) 2022, 14(11).

  23. Belotti Y, Lim EH, Lim CT. The role of the Extracellular Matrix and Tumor-infiltrating Immune cells in the prognostication of high-Grade Serous Ovarian Cancer. Cancers (Basel) 2022, 14(2).

  24. Laios A, Katsenou A, Tan YS, Johnson R, Otify M, Kaufmann A, Munot S, Thangavelu A, Hutson R, Broadhead T, et al. Feature selection is critical for 2-Year prognosis in Advanced Stage High Grade Serous Ovarian Cancer by using machine learning. Cancer Control. 2021;28:10732748211044678.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Feng Y, Wang Z, Cui R, Xiao M, Gao H, Bai H, Delvoux B, Zhang Z, Dekker A, Romano A, et al. Clinical analysis and artificial intelligence survival prediction of serous ovarian cancer based on preoperative circulating leukocytes. J Ovarian Res. 2022;15(1):64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Hansen TB. Improved circRNA identification by combining prediction algorithms. Front Cell Dev Biol. 2018;6:20.

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  27. Wu M, Zhao Y, Dong X, Jin Y, Cheng S, Zhang N, Xu S, Gu S, Wu Y, Yang J, et al. Artificial intelligence-based preoperative prediction system for diagnosis and prognosis in epithelial ovarian cancer: a multicenter study. Front Oncol. 2022;12:975703.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Liu Z, Guo C, Dang Q, Wang L, Liu L, Weng S, Xu H, Lu T, Sun Z, Han X. Integrative analysis from multi-center studies identities a consensus machine learning-derived lncRNA signature for stage II/III colorectal cancer. EBioMedicine. 2022;75:103750.

    Article  CAS  PubMed  Google Scholar 

  29. Wang L, Liu Z, Liang R, Wang W, Zhu R, Li J, Xing Z, Weng S, Han X, Sun YL. Comprehensive machine-learning survival framework develops a consensus model in large-scale multicenter cohorts for pancreatic cancer. Elife 2022, 11.

  30. Liu Z, Liu L, Weng S, Guo C, Dang Q, Xu H, Wang L, Lu T, Zhang Y, Sun Z, et al. Machine learning-based integration develops an immune-derived lncRNA signature for improving outcomes in colorectal cancer. Nat Commun. 2022;13(1):816.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  31. Qiu YL, Zheng H, Devos A, Selby H, Gevaert O. A meta-learning approach for genomic survival analysis. Nat Commun. 2020;11(1):6350.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  32. Li J, Lai C, Peng S, Chen H, Zhou L, Chen Y, Chen S. The prognostic value of integration of pretreatment serum amyloid A (SAA)-EBV DNA (S-D) grade in patients with nasopharyngeal carcinoma. Clin Transl Med. 2020;9(1):2.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Lundberg SM, Nair B, Vavilala MS, Horibe M, Eisses MJ, Adams T, Liston DE, Low DK, Newman SF, Kim J, et al. Explainable machine-learning predictions for the prevention of hypoxaemia during surgery. Nat Biomed Eng. 2018;2(10):749–60.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Asher V, Lee J, Innamaa A, Bali A. Preoperative platelet lymphocyte ratio as an independent prognostic marker in ovarian cancer. Clin Transl Oncol. 2011;13(7):499–503.

    Article  PubMed  Google Scholar 

  35. Raungkaewmanee S, Tangjitgamol S, Manusirivithaya S, Srijaipracharoen S, Thavaramara T. Platelet to lymphocyte ratio as a prognostic factor for epithelial ovarian cancer. J Gynecol Oncol. 2012;23(4):265–73.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Williams KA, Labidi-Galy SI, Terry KL, Vitonis AF, Welch WR, Goodman A, Cramer DW. Prognostic significance and predictors of the neutrophil-to-lymphocyte ratio in ovarian cancer. Gynecol Oncol. 2014;132(3):542–50.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Dong L, Bai K, Cao Y, Huang Q, Lv L, Jiang Y. Prognostic value of pre-operative platelet to lymphocyte ratio in patients with Resected Primary Hepatocellular Carcinoma. Clin Lab. 2016;62(11):2191–6.

    PubMed  Google Scholar 

  38. Zhao C, Li LQ, Yang FD, Wei RL, Wang MK, Song DX, Guo XY, Du W, Wei XT. A hematological-related Prognostic Scoring System for patients with newly diagnosed Glioblastoma. Front Oncol. 2020;10:591352.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Arthur R, Williams R, Garmo H, Holmberg L, Stattin P, Malmstrom H, Lambe M, Hammar N, Walldius G, Robinsson D, et al. Serum inflammatory markers in relation to prostate cancer severity and death in the Swedish AMORIS study. Int J Cancer. 2018;142(11):2254–62.

    Article  CAS  PubMed  Google Scholar 

  40. Cruz JA, Wishart DS. Applications of machine learning in cancer prediction and prognosis. Cancer Inf. 2007;2:59–77.

    Google Scholar 

  41. Xu H, Liu Z, Weng S, Dang Q, Ge X, Zhang Y, Ren Y, Xing Z, Chen S, Zhou Y, et al. Artificial intelligence-driven consensus gene signatures for improving bladder cancer clinical outcomes identified by multi-center integration analysis. Mol Oncol. 2022;16(22):4023–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Chu G, Ji X, Wang Y, Niu H. Integrated multiomics analysis and machine learning refine molecular subtypes and prognosis for muscle-invasive urothelial cancer. Mol Ther Nucleic Acids. 2023;33:110–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Li Y, Niu JH, Wang Y. Machine learning-based neddylation landscape indicates different prognosis and immune microenvironment in endometrial cancer. Front Oncol. 2023;13:1084523.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Boehm KM, Aherne EA, Ellenson L, Nikolovski I, Alghamdi M, Vazquez-Garcia I, Zamarin D, Long Roche K, Liu Y, Patel D, et al. Multimodal data integration using machine learning improves risk stratification of high-grade serous ovarian cancer. Nat Cancer. 2022;3(6):723–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Zhong YC, Zhang T, Di W, Li WP. Thrombin promotes epithelial ovarian cancer cell invasion by inducing epithelial-mesenchymal transition. J Gynecol Oncol. 2013;24(3):265–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Swier N, Versteeg HH. Reciprocal links between venous thromboembolism, coagulation factors and ovarian cancer progression. Thromb Res. 2017;150:8–18.

    Article  CAS  PubMed  Google Scholar 

  47. Yuk HD, Ku JH. Role of systemic inflammatory response markers in Urothelial Carcinoma. Front Oncol. 2020;10:1473.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Bai G, Zhou Y, Rong Q, Qiao S, Mao H, Liu P. Development of Nomogram models based on peripheral blood score and clinicopathological parameters to Predict Preoperative Advanced Stage and Prognosis for epithelial ovarian Cancer patients. J Inflamm Res. 2023;16:1227–41.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not available.

Funding

National Natural Science Foundation of China: 82072866, 82272888, 82102856; Science and Technology Commission of Shanghai Municipality: 21S31903600, 21YF1425800; Shanghai Hospital Development Center: SHDC2022CRW013, SHDC12022106, SHDC2022CRT015, SHDC12021601, 2022SKLY-12; Shanghai Jiao Tong University: YG2022ZD005.

Author information

Authors and Affiliations

Authors

Contributions

M.W performed the study and wrote the manuscript. M.W. and J.Y. interpreted the data and carried out statistical analysis. S.C., Y.Z., S.X, S.G., and YS.W. collected patients’ clinical data, blood features, and follow-up information. M.M., X.L., and H.Z. performed data curation. Y.W., A.Z., and M.W. designed the study. M.W. drew the figures and produced tables. J.S checked the grammar. M.W., S.G., J.S., Y.W., and A.Z. revised the manuscript. All authors approved the final manuscript.

Corresponding authors

Correspondence to Yu Wang or Aimin Zhao.

Ethics declarations

Ethics approval and consent to participate

We performed the research in accordance with the Declaration of Helsinki. This study was approved by the Ethics Committee of Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine. All patients provided informed consent for the usage of their data for research purposes.

Consent for publication

This study doesn’t contain any individual person’s data.

Conflict of interest

The authors declare no conflict of interest.

Additional information

Publisher’s Note

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

Electronic supplementary material

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

Wu, M., Gu, S., Yang, J. et al. Comprehensive machine learning-based preoperative blood features predict the prognosis for ovarian cancer. BMC Cancer 24, 267 (2024). https://0-doi-org.brum.beds.ac.uk/10.1186/s12885-024-11989-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s12885-024-11989-1

Keywords