• Stacked ensemble of machine-learning algorithms could establish more accurate prediction model for survival analysis than existing methods.

  • Stacked ensemble model can be applied to personalized prediction of HSCT outcomes from pretransplant characteristics.

Graft-versus-host disease-free, relapse-free survival (GRFS) is a useful composite end point that measures survival without relapse or significant morbidity after allogeneic hematopoietic stem cell transplantation (allo-HSCT). We aimed to develop a novel analytical method that appropriately handles right-censored data and competing risks to understand the risk for GRFS and each component of GRFS. This study was a retrospective data-mining study on a cohort of 2207 adult patients who underwent their first allo-HSCT within the Kyoto Stem Cell Transplantation Group, a multi-institutional joint research group of 17 transplantation centers in Japan. The primary end point was GRFS. A stacked ensemble of Cox Proportional Hazard (Cox-PH) regression and 7 machine-learning algorithms was applied to develop a prediction model. The median age for the patients was 48 years. For GRFS, the stacked ensemble model achieved better predictive accuracy evaluated by C-index than other state-of-the-art competing risk models (ensemble model: 0.670; Cox-PH: 0.668; Random Survival Forest: 0.660; Dynamic DeepHit: 0.646). The probability of GRFS after 2 years was 30.54% for the high-risk group and 40.69% for the low-risk group (hazard ratio compared with the low-risk group: 2.127; 95% CI, 1.19-3.80). We developed a novel predictive model for survival analysis that showed superior risk stratification to existing methods using a stacked ensemble of multiple machine-learning algorithms.

Allogeneic hematopoietic stem cell transplantation (HSCT) is a potentially curative therapy for hematological malignancies, bone marrow (BM) failure syndromes, and immunodeficiency syndromes. Although the improvement in outcome has been confirmed by several studies, 2 serious risk factors remain for poor outcome: transplantation-related morbidity and mortality (TRM) and progression of diseases. Intensive treatment to overcome disease progression often leads to severe graft-versus-host disease (GVHD) and TRM; on the other hand, reduced-intensity conditioning can reduce TRM but increase relapse rate.1-4 

Age, disease stage, donor type, and donor recipient gender combinations were reported to influence survival, nonrelapse mortality (NRM), and relapse risk. These pretransplant risk factors were identified from hypothesis-driven variable selection and validation using conventional statistical analysis. Various predictive scoring systems have also been established based on these findings. Hematopoietic cell transplantation–specific comorbidity index is used for assessment of pretransplant comorbidities.5  The European Group for Blood and Marrow Transplantation risk score consists of 6 pretransplant risk factors associated with relapse and GVHD.6  These scoring systems are used for clinical decision-making for HSCT indication or donor selection.

Machine learning is a field of computer science in which computer algorithms that have the ability to improve from experience without being explicitly programmed are studied. Machine-learning methods provide statistical calculations without the assumptions needed for traditional statistical analysis, so machine-learning–based prediction gives novel insights into clinical medicine. To predict prognosis of HSCT patients, early studies demonstrated the feasibility of machine-learning algorithms for binary outcomes, such as decision tree–based learning, artificial neural networks, and support vector machines.7-11  Recent reports applied machine-learning methods developed for right-censored data with or without competing risks for HSCT outcomes.12-14  Machine-learning methods are a useful way to predict time-dependent outcomes without assumptions, but how to improve predictive accuracy is still an ongoing discussion.

In this study, we developed a novel prediction model that appropriately handles right-censored data and competing risks using ensemble learning to predict composite end point of GVHD-free, relapse-free survival (GRFS).15 

Population

All transplantation data in Japan are annually collected at the Japanese Data Center for Hematopoietic Cell Transplantation. The Kyoto Stem Cell Transplantation Group (KSCTG), which is a multi-center group of 17 transplantation centers in Japan, received transplant data from the Japanese Data Center for Hematopoietic Cell Transplantation. From the registry database of KSCTG, we extracted clinical data for 2207 patients who underwent their first HSCT for hematologic malignancies between 1996 and 2016 in KSCTG hospitals. The study was conducted according to the Declaration of Helsinki and was approved by the institutional review boards at Kyoto University Hospital and all other participating centers.

End point and definitions

The primary end point was GRFS, and the secondary end points were overall survival (OS), relapse, NRM, and GVHD. GRFS was defined as the time from transplant to the last date of follow-up or event of grade III-IV acute GVHD, extensive chronic GVHD, relapse, or death.16  Relapse was defined based on the morphological and clinical evidence of disease activity, and NRM was defined as the time to death without relapse. Acute and chronic GVHD were diagnosed and graded using standard criteria.17,18  The intensity of the conditioning regimen was classified as myeloablative if total body irradiation >8 Gy, oral busulfan ≥9 mg/kg, IV busulfan ≥7.2 mg/kg, melphalan >140 mg/m2, or thiotepa ≥10 mg/kg was used in the conditioning regimen; otherwise it was classified as reduced intensity.19  We assessed disease risk using the refined disease risk index (DRI) established by the Center for International Blood and Marrow Transplant Research.20  The refined DRI does not establish adult T-cell leukemia/lymphoma (ATL) as an individual risk group. We regarded complete remission or partial remission ATL as intermediate risk and advanced ATL as very high risk. We categorized high-grade B-cell lymphoma with MYC and BCL2 and/or BCL6 rearrangements into the same group with Burkitt lymphoma due to its poor prognosis. DRI was retrospectively calculated based on the available registry data about diagnosis, chromosomal alterations, and staging before transplantation. Disease stage was defined as previously described.21 

The following is a summary of the analysis outline: (1) preprocessing-data quality assurance and imputations of missing values, (2) construction of each prediction model, (3) development of a stacked ensemble model, and (4) assessment of the predictive performance of a stacked ensemble model in accordance with the Type 2a (random split-sample development and validation) of prediction model studies covered by the Transparent Reporting of a prediction model for Individual Prognosis Or Diagnosis statement.22  Missing values are imputed with median value of the nonmissing data for categorical variables and with mean value of the nonmissing data for continuous variables. A dummy variable is also generated, indicating whether the data were missing for that particular patient.

Establishment of stacked ensemble model

A stacked ensemble of multiple machine-learning algorithms for right-censored data was applied to develop a model. Formally, an ensemble model is a model that combines the predictions from multiple trained models. A stacked ensemble model is a variation of the ensemble method and uses an algorithm that takes the outputs of submodels as inputs and learns the optimal way to combine the input predictions.23  Predictions are first generated using different algorithms, including cause-specific Cox Proportional Hazard (Cox-PH), Random Survival Forest, Dynamic DeepHit, ADABoost, XGBoost, Extra Tree Classifier, Bagging Classifier, and Gradient Boosting Classifier.12,24 -31  A meta-model is subsequently trained, using these predictions as inputs, to generate the final prediction. Data were randomly split into the training set (70% of the dataset) and the validation set (30%). The model was trained and tested using a fivefold cross-validation on the training set. All models were trained using 26 input variables available from the registry data containing information on a patient’s underlying disease, donor source, and patient and donor’s demographic characteristics. Patient sex, source of stem cells, and diagnosis (Table 1) and variables (supplemental Table 1) were treated as categorical variables. Patient age and time from diagnosis to transplant (Table 1) and variables (supplemental Table 2) were treated as continuous variables.

Table 1.

Patient characteristics

VariableTotalTraining setValidation setP
n = 2207 (%)n = 1765 (%)n = 442 (%)
Age group at transplant, y .327 
 ≤30 339 (15.4) 265 (15.0) 74 (16.7)  
 >30-40 340 (15.4) 277 (15.7) 63 (14.3)  
 >40-50 480 (21.7) 376 (21.3) 104 (23.5)  
 >50-60 631 (28.6) 509 (28.8) 122 (27.6)  
 >60 417 (18.9) 338 (19.2) 79 (17.9)  
Sex .572 
 Male 925 (41.9) 734 (41.6) 191 (43.2)  
 Female 1282 (58.1) 1031 (58.4) 251 (56.8)  
Source of stem cells .279 
 BM 1349 (61.1) 1061 (60.1) 288 (65.2)  
 Peripheral blood 356 (16.1) 292 (16.5) 64 (14.5)  
 BM + peripheral blood 7 (0.3) 6 (0.3) 1 (0.2)  
 Cord blood 495 (22.4) 406 (23.0) 89 (20.1)  
Time from diagnosis to transplant .707 
 ≤6 mo 793 (35.9) 638 (36.1) 155 (35.1)  
 >6 mo 1392 (63.1) 1108 (62.8) 284 (64.3)  
 Uncertain/missing 22 (1.0) 19 (1.1) 3 (0.7)  
Year of transplant .441 
 1996-2006 718 (32.5) 581 (32.9) 137 (31.0)  
 2007-2016 1489 (67.5) 1184 (67.1) 305 (69.0)  
Diagnosis .652 
 AML 868 (39.3) 703 (39.8) 165 (37.3)  
 ALL 371 (16.8) 296 (16.8) 75 (17.0)  
 ATL 130 (5.9) 102 (5.8) 28 (6.3)  
 CML 124 (5.6) 94 (5.3) 30 (6.8)  
 MDS 342 (15.5) 274 (15.5) 68 (15.4)  
 Other leukemia 31 (1.4) 23 (1.3) 8 (1.8)  
 MPN 38 (1.7) 28 (1.6) 10 (2.3)  
 NHL/HL/other lymphoma 294 (13.3) 236 (13.4) 58 (13.1)  
 MM/PCD 9 (0.4) 9 (0.5) 0 (0.0)  
Follow-up of survivors 
 Median time, month (range) 52.5 (0.5-244.6) 57.6 (0.5-244.6) 58.6 (0.7-235.4) .743 
VariableTotalTraining setValidation setP
n = 2207 (%)n = 1765 (%)n = 442 (%)
Age group at transplant, y .327 
 ≤30 339 (15.4) 265 (15.0) 74 (16.7)  
 >30-40 340 (15.4) 277 (15.7) 63 (14.3)  
 >40-50 480 (21.7) 376 (21.3) 104 (23.5)  
 >50-60 631 (28.6) 509 (28.8) 122 (27.6)  
 >60 417 (18.9) 338 (19.2) 79 (17.9)  
Sex .572 
 Male 925 (41.9) 734 (41.6) 191 (43.2)  
 Female 1282 (58.1) 1031 (58.4) 251 (56.8)  
Source of stem cells .279 
 BM 1349 (61.1) 1061 (60.1) 288 (65.2)  
 Peripheral blood 356 (16.1) 292 (16.5) 64 (14.5)  
 BM + peripheral blood 7 (0.3) 6 (0.3) 1 (0.2)  
 Cord blood 495 (22.4) 406 (23.0) 89 (20.1)  
Time from diagnosis to transplant .707 
 ≤6 mo 793 (35.9) 638 (36.1) 155 (35.1)  
 >6 mo 1392 (63.1) 1108 (62.8) 284 (64.3)  
 Uncertain/missing 22 (1.0) 19 (1.1) 3 (0.7)  
Year of transplant .441 
 1996-2006 718 (32.5) 581 (32.9) 137 (31.0)  
 2007-2016 1489 (67.5) 1184 (67.1) 305 (69.0)  
Diagnosis .652 
 AML 868 (39.3) 703 (39.8) 165 (37.3)  
 ALL 371 (16.8) 296 (16.8) 75 (17.0)  
 ATL 130 (5.9) 102 (5.8) 28 (6.3)  
 CML 124 (5.6) 94 (5.3) 30 (6.8)  
 MDS 342 (15.5) 274 (15.5) 68 (15.4)  
 Other leukemia 31 (1.4) 23 (1.3) 8 (1.8)  
 MPN 38 (1.7) 28 (1.6) 10 (2.3)  
 NHL/HL/other lymphoma 294 (13.3) 236 (13.4) 58 (13.1)  
 MM/PCD 9 (0.4) 9 (0.5) 0 (0.0)  
Follow-up of survivors 
 Median time, month (range) 52.5 (0.5-244.6) 57.6 (0.5-244.6) 58.6 (0.7-235.4) .743 

ALL, acute lymphoblastic leukemia; AML, acute myeloid leukemia; CML, chronic myeloid leukemia; HL, Hodgkin lymphoma; MDS, myelodysplastic syndrome; MM, multiple myeloma; MPN, myeloproliferative neoplasm; NHL, non-Hodgkin lymphoma; PCD, plasma cell disease.

Cox model, Random Survival Forest, and Dynamic DeepHit can directly handle competing risks. ADABoost, XGBoost, Extra Tree Classifier, Bagging Classifier, and Gradient Boosting Classifier belong to the class of multi-output tree-based ensemble algorithms. To allow these models to handle competing risks, we use the First Hitting Time model, which assumes that the individual hazard function is a form-fixed stochastic process.32  We then use these multi-output tree-based algorithms to estimate the probability density function of the first hitting time. In particular, for each patient, the predicted value y^ is a vector: y^=[y^1,,y^Tmax,y^Tmax+1], where Tmax is the longest observed time and the time unit used in our study is 30 days (duration from y^t to y^t+1). Given an individual with the covariate x, these models estimate y^t with the estimated probability P^(t,δix), where δi denotes the occurrence of the event of interest.

SHapley Additive exPlanations (SHAP) values were calculated for the stacked ensemble model. First proposed by Lundberg and Lee,33  SHAP is a united approach to explain the output of any machine-learning or deep-learning model. SHAP is based on Shapley values, a concept from game theory that measures the average contribution of a feature value to the prediction across all possible combinations (or coalitions) of other features. The pseudocode to calculate the SHAP value for feature X can be described as follows: (1) Get all subsets of features S that do not contain X. (2) Compute the effects of adding X to all those subsets on the predictions. (3) Average over all the contributions to compute the marginal contribution.

We used the Cox-PH model as the benchmark case and evaluated the models’ performance using inverse probability censoring weighted version of the C statistic (C-index) for single-risk models and the truncated C-index for competing-risk models.34,35  We used the restricted cubic splines approach to calculate the smoothed calibration curves and compute the integrated calibration index (ICI) and the median of the absolute difference between the predicted survival probabilities and smoothed survival frequencies (E50) to assess the calibration of different survival models.36  We first used the model to calculate the risk score for each patient in the validation set and get the median risk score. The high-risk group was defined as patients with a risk score above this median risk score whereas the low-risk group was defined as patients with a risk score below this median risk score. We also validated the final risk scores in the validation set using the Cox regression model or Fine-Gray competing risk model.

Other statistical considerations

Descriptive statistics were used to summarize patient characteristics. Comparisons of intergroup distributions were performed with the χ-square test or Fisher’s exact test for categorical variables and the Kruskal-Wallis test for continuous variables. The probability of GRFS and OS was estimated using the Kaplan-Meier method. Cumulative incidences for relapse, NRM, and GVHD were calculated using the cumulative incidence function to account for competing risks.37  Competing events were death without relapse for relapse, relapse for NRM, and death without GVHD for acute and chronic GVHD. P values < .05 were considered significant. All statistical analyses were performed with Python version 3.7 (Python Software Foundation, Fredricksburg, VA) and R version 3.6.1 (The R Foundation for Statistical Computing, Vienna, Austria).

Patient characteristics

We included and evaluated 2207 adult patients who underwent their first allogeneic HSCT for hematologic malignancies (Table 1; supplemental Tables 1 and 2). The median follow-up period for survivors was 52.5 months (range 0.5-244.6) after HSCT. The most common indication of HSCT was acute myeloid leukemia (n = 868; 39.3%) followed by acute lymphoblastic leukemia (n = 371; 16.8%), myelodysplastic syndrome (n = 342; 15.5%), and mature lymphoid neoplasms (lymphoma/myeloma; n = 294; 13.3%). The graft source was mainly BM (61.1%) followed by cord blood (22.4%) and peripheral blood (16.1%). Frequency of patients transplanted with HLA 1 antigen-mismatched patients was higher in the validation cohort than in the training cohort (22.2% vs 15.8%; P = .011). There was no significant difference between the training cohort and the validation cohort other than the number of HLA antigen mismatches.

Comparison of predictive models

Prediction models were generated using a stacked ensemble containing 1 classical statistical analysis, 1 deep-learning model, and 6 machine-learning algorithms (Figure 1). The novel stacked ensemble model achieved a higher C-index for GRFS (C-index: 0.670) than other competing risk models in the validation dataset (Table 2; Cox-PH: 0.668; Random Survival Forest: 0.660; XGBoost: 0.602; Gradient Boosting: 0.630; Component-wise Gradient Boosting: 0.663; Dynamic DeepHit: 0.646). This model also showed the highest C-index for OS (C-index: 0.763), relapse (C-index: 0.793), NRM (C-index: 0.777), grade II-IV acute GVHD (C-index: 0.656), and chronic GVHD (C-index: 0.583). We also calculated C-index for patients who received transplant between 2007 and 2018 in validation dataset and confirmed that the stacked ensemble model showed the highest C-index (supplemental Table 3; GRFS: 0.844; OS: 0.716; relapse: 0.819; NRM: 0.770; grade II-IV acute GVHD: 0.536; chronic GVHD: 0.606). The stacked ensemble model showed the smallest ICI for GRFS (0.023), OS (0.210), relapse (0.044), grade II-IV acute GVHD (0.017), and chronic GVHD (0.258) and achieved the smallest E50 other than for GRFS and chronic GVHD, for which the stacked ensemble model showed the second smallest E50 (Table 3; supplemental Figure 1). Compared with other state-of-the-art competing risk models, the stacked ensemble model achieves higher C-index and smaller ICI for GRFS, OS, relapse, NRM, and GVHD, and we used this model for feature extraction and prediction.

Figure 1.

Stacked ensemble model of machine-learning algorithms. Scheme of meta-model construction using stacking as an ensemble method.

Figure 1.

Stacked ensemble model of machine-learning algorithms. Scheme of meta-model construction using stacking as an ensemble method.

Close modal
Table 2.

Performance of each prediction model according to C-index in the validation cohort

Risk categoryGRFSOSRelapseNRMaGVHDcGVHD
Cox-PH 0.668 0.740 0.770 0.664 0.651 0.564 
Fine-Gray competing risk model NA NA 0.719 0.577 0.582 0.516 
Random Survival Forest 0.660 0.745 0.788 0.761 0.580 0.577 
XGBoost 0.602 0.712 0.756 0.543 0.540 0.573 
Gradient Boosting 0.630 0.602 0.754 0.453 0.590 0.505 
Component-wise Gradient Boosting 0.663 0.652 0.774 0.585 0.464 0.570 
Dynamic DeepHit 0.646 0.710 0.730 0.691 0.537 0.555 
Stacked Ensemble Model 0.670 0.763 0.793 0.777 0.656 0.583 
Risk categoryGRFSOSRelapseNRMaGVHDcGVHD
Cox-PH 0.668 0.740 0.770 0.664 0.651 0.564 
Fine-Gray competing risk model NA NA 0.719 0.577 0.582 0.516 
Random Survival Forest 0.660 0.745 0.788 0.761 0.580 0.577 
XGBoost 0.602 0.712 0.756 0.543 0.540 0.573 
Gradient Boosting 0.630 0.602 0.754 0.453 0.590 0.505 
Component-wise Gradient Boosting 0.663 0.652 0.774 0.585 0.464 0.570 
Dynamic DeepHit 0.646 0.710 0.730 0.691 0.537 0.555 
Stacked Ensemble Model 0.670 0.763 0.793 0.777 0.656 0.583 

aGVHD, grade II-IV acute GVHD; cGVHD, chronic GVHD; NA, not applicable.

Table 3.

Comparison of the Integrated Calibration Index and the median of the absolute difference between the predicted survival probabilities and smoothed survival frequencies for each prediction model

Integrated calibration index (EC50)
Risk categoryGRFSOSRelapseaGVHDcGVHD
Cox-PH 0.139 (0.151) 0.283 (0.248) 0.055 (0.029) 0.218 (0.212) 0.263 (0.208) 
Random Survival Forest 0.142 (0.147) 0.365 (0.372) 0.048 (0.029) 0.173 (0.178) 0.345 (0.346) 
XGBoost 0.027 (0.007) 0.393 (0.381) 0.176 (0.163) 0.265 (0.264) 0.306 (0.265) 
Gradient Boosting 0.050 (0.047) 0.438 (0.449) 0.159 (0.129) 0.254 (0.256) 0.309 (0.275) 
Component-wise Gradient Boosting 0.061 (0.068) 0.397 (0.395) 0.171 (0.145) 0.261 (0.264) 0.324 (0.318) 
Dynamic DeepHit 0.054 (0.059) 0.405 (0.409) 0.152 (0.153) 0.106 (0.108) 0.319 (0.320) 
Stacked Ensemble Model 0.023 (0.017) 0.210 (0.194) 0.044 (0.018) 0.017 (0.018) 0.258 (0.226) 
Integrated calibration index (EC50)
Risk categoryGRFSOSRelapseaGVHDcGVHD
Cox-PH 0.139 (0.151) 0.283 (0.248) 0.055 (0.029) 0.218 (0.212) 0.263 (0.208) 
Random Survival Forest 0.142 (0.147) 0.365 (0.372) 0.048 (0.029) 0.173 (0.178) 0.345 (0.346) 
XGBoost 0.027 (0.007) 0.393 (0.381) 0.176 (0.163) 0.265 (0.264) 0.306 (0.265) 
Gradient Boosting 0.050 (0.047) 0.438 (0.449) 0.159 (0.129) 0.254 (0.256) 0.309 (0.275) 
Component-wise Gradient Boosting 0.061 (0.068) 0.397 (0.395) 0.171 (0.145) 0.261 (0.264) 0.324 (0.318) 
Dynamic DeepHit 0.054 (0.059) 0.405 (0.409) 0.152 (0.153) 0.106 (0.108) 0.319 (0.320) 
Stacked Ensemble Model 0.023 (0.017) 0.210 (0.194) 0.044 (0.018) 0.017 (0.018) 0.258 (0.226) 

EC50, the median of the absolute difference between the predicted survival probabilities and smoothed survival frequencies.

Feature extraction and explanation

Using the stacked ensemble model, we calculated SHAP feature importance values for 26 variables that are used for model construction and ranked them according to their ability to discriminate between high- and low-risk patients. The SHAP value could explain the contribution of each variable to the estimate of GRFS for each patient (Figure 2A). Mean absolute value of the SHAP values for each variable could show the overall influence of each variable to model construction and their importance (Figure 2B). Characteristics about donors, including cell source, related donors or unrelated donors, and siblings or nonsibling relatives, were the most influential factors for GRFS. On the other hand, disease status before transplantation was the most influential factor for OS, as shown by high mean absolute SHAP value of DRI and disease stage.

Figure 2.

SHapley Additive exPlanations feature importance value for GRFS and OS. (A) Representative patients in GRFS (left) and OS (right) model. Red and blue bars indicate positive and negative feature contributions, respectively. (B) SHAP feature importance measured as the mean absolute Shapley values for GRFS (left) and OS (right). Variables having top 10 highest impact on model outputs are shown.

Figure 2.

SHapley Additive exPlanations feature importance value for GRFS and OS. (A) Representative patients in GRFS (left) and OS (right) model. Red and blue bars indicate positive and negative feature contributions, respectively. (B) SHAP feature importance measured as the mean absolute Shapley values for GRFS (left) and OS (right). Variables having top 10 highest impact on model outputs are shown.

Close modal

Validation

To validate the risk scores, we applied this model to the validation set. Based on the prediction score for each patient derived from the stacked ensemble model, we classified the final risk scores in the validation set into 2 groups: (1) high risk (above median) and (2) low risk (below median) for each of the risk categories. The probability of GRFS after 1 year and 2 years was 44.80% and 30.54% for the high-risk group and 57.47% and 40.69% for the low-risk group, respectively (Figure 3; hazard ratio [HR] compared with the low risk: 2.127; 95% CI, 1.19-3.80). The OS at 5 years was 52.58% for the high-risk group and 80.54% for the low-risk group (HR: 2.67; 95% CI, 2.02-3.52). Cumulative incidence of relapse at 5 years was 34.78% for the high-risk group and 13.61% for the low-risk group (Figure 4; HR compared with the low risk: 2.72; 95% CI, 1.85-3.99). The cumulative incidence of NRM at 5 years was 22.13% for the high-risk group and 12.92% for the low-risk group (HR compared with the low risk: 1.947; 95% CI, 1.24-2.74). The cumulative incidence of grade II-IV acute GVHD at 100 days was 46.61% for the high-risk group and 30.02% for the low-risk group (HR compared with the low risk: 1.66, 95% CI, 1.22-2.27). The cumulative incidence of chronic GVHD at 5 years was 35.20% for the high-risk group and 22.50% for the low-risk group (HR: 1.97; 95% CI, 1.44-2.70).

Figure 3.

Kaplan-Meier estimates for GRFS and OS in the validation set. Estimates for GRFS (A) and OS (B) are shown. Patients are stratified based on stacked ensemble meta-model score.

Figure 3.

Kaplan-Meier estimates for GRFS and OS in the validation set. Estimates for GRFS (A) and OS (B) are shown. Patients are stratified based on stacked ensemble meta-model score.

Close modal
Figure 4.

Cumulative incidence of relapse, NRM, and GVHD. (A) Relapse. (B) NRM. (C) Grade II-IV acute GVHD. (D) Chronic GVHD. Patients are stratified based on stacked ensemble meta-model score.

Figure 4.

Cumulative incidence of relapse, NRM, and GVHD. (A) Relapse. (B) NRM. (C) Grade II-IV acute GVHD. (D) Chronic GVHD. Patients are stratified based on stacked ensemble meta-model score.

Close modal

We successfully developed a novel prediction model for GRFS using the stacked ensemble of classical statistical analysis and multiple machine-learning algorithms. This study showed for the first time the improvement of predictive accuracy for GRFS using ensemble learning applicable for right-censored medical record data.

Our ensemble model is designed to handle right-censored data, a form of missing data problem specific for survival analysis. As a result, its outputs can be directly compared with classical algorithms such as Cox-PH or Fine-Gray model. Ignoring censored patients could potentially give a bias to the outcome. The longer the follow-up time, the larger this bias due to an increasing number of censored patients. Shouval et al13  used Random Survival Forest to analyze right-censored data and successfully established an umbilical cord blood transplantation risk score that could predict OS and RFS at 2 years in patients with acute leukemia who received cord blood transplantation. In addition, our model can analyze data with multiple competing risks and can be modified to include time-varying variables. Finally, by combining the outputs of various machine-learning methods, our ensemble model outperforms not only classical algorithms but also the most cutting-edge machine-learning models for survival analysis.

Stacking is an ensemble method that combines multiple heterogenous algorithms into single better predictive model. Sachs et al developed a stacking method with a pseudo-observation approach for ensemble of various machine-learning algorithms handling right-censored data.38  A diverse set of initial classifiers is the fundamental aspect for establishing an accurate ensemble model, so our ensemble model used 8 algorithms, including statistical analysis and tree-based and neural-network–based learning for meta-model construction.

One of the fundamental advantages of the machine-learning–based data mining approach is unbiased feature selection and prediction; on the other hand, its hidden nature of model construction and prediction makes it difficult for us to interpret the contribution of variables toward end points.9  Although this novel ensemble method provides superior prediction to conventional statistical analysis and each machine-learning algorithm, stacked-ensemble lost the interpretable tree-like structure of decision tree–based learning. Therefore, we introduced SHAP value for explanation of model prediction. In addition to the influence of each variable on model construction, SHAP value could extract and clearly visualize the contribution of each variable for personalized prediction of individual patients.

In addition to the hidden nature of model construction, the criteria for selection of machine-learning algorithms were often unclarified. A previous report using Japanese Transplant Unified Management Program created a prediction model derived from alternating decision tree based on its highest value of area under the curve.11  Our ensemble model showed higher C-index value than Cox-PH or state-of-the-art machine-learning algorithms for GRFS and OS. Validated by these findings, we used the stacked ensemble meta-model for further analysis.

GRFS was initially developed by Holtan et al to incorporate 2 major posttransplantation complications, relapse and GVHD, into a single end point.15  GRFS is useful for understanding ongoing morbidity due to GVHD that could not be interpreted by OS or RFS. For example, previous studies found that BM donors showed higher GRFS than peripheral blood stem cell donors in matched-sibling donors although BM and peripheral blood stem cell did not show difference in terms of OS or RFS. However, composite end points using right-censored data only measure the time to the first event, and GRFS cannot replace OS, RFS, or GVHD. Magenau et al reported that chronic GVHD had less modulating effect on OS than grade III-IV acute GVHD or relapse.39  This is partly explained by the association of graft-versus-leukemia effect with chronic GVHD.40  On the other hand, chronic GVHD has tremendous negative impact on quality of daily life after transplantation even under mild to moderate symptoms.41  To understand the efficacy of the stacked ensemble model on different end points after transplantation, we also analyzed OS, relapse, NRM, grade II-IV GVHD, and chronic GVHD, respectively. For all end points, the stacked ensemble model showed better predictive accuracy than other models, which validated the versatility and robustness of this model.

Several study limitations should be noted. One of the limitations is that this model is not completely free from hypothesis in terms of feature selection. We used 26 variables accessible in the registry for establishment of prediction models but might still ignore unknown or unavailable risk factors. For example, center effect was reported to be associated with HSCT outcome, but existence of unknown factors was suggested.42  Okamura et al14  developed a web application tool for OS, progression-free survival, relapse, and NRM after HSCT in a single center and provided their source code for in-house prognosis prediction using random survival forest. A center-specific prediction model might be able to overcome institution-associated bias, but standardization of prediction algorithms is still required. In addition to limited number of variables, some of the variables were categorized into subgroups based on clinically established criteria. For example, although HLA mismatch is categorized based on the number of mismatched antigens or alleles, each mismatch causes different immunological interaction between donor cells and recipient cells.21,43-46  Because we categorized patients into 6 groups according to the number of mismatched antigens or alleles, we could not consider their mismatched locus. Another example is DRI. We stratified pretransplant disease condition using DRI or disease status, which does not reflect recent progress in clinical implementation of genome sequencing technology.47,48  Nazha et al established prediction model that could integrate clinical and mutational variables into a single model using Random Survival Forest.49  Gandelman et al successfully stratified chronic GVHD severity using computational workflow made of visualization of high-dimensional single-cell data based on the t-Distributed Stochastic Neighbor Embedding algorithm, self-organizing maps, and marker enrichment modeling.50  Wider range of data collection and categorization of variables using machine learning–based clustering methods might contribute to unbiased variable selection and calculation.

Sample size also limited risk stratification of our model. In this study, we used C-index for evaluation and comparison of the predictive accuracy of prediction models, and the novel meta-model showed highest value for C-index. However, due to the small size of the validation set, we only classified the final risk scores into 2 groups. Moreover, these findings should be interpreted carefully from the viewpoint of bias in this registry data derived from known and unknown factors. Our dataset did not include the information about posttransplant cyclophosphamide usage, especially in haploidentical HSCT, maintenance therapy, and prophylaxis for fungal or viral infection. Other unknown factors, including center effect and different distribution of HLA alleles and haplotypes, might be influential in transplant outcomes. Further validation of the stacked ensemble model using different registry data is warranted.

In conclusion, we improved machine-learning predictive accuracy of GRFS using a stacked ensemble meta-model feasible for right-censored medical record data. This model provides direct and versatile application of machine-learning algorithms for time-to-event analysis. A user-friendly Web tool for personalized pretransplant prediction of HSCT outcome is now being constructed. Although external validation using larger data with more detailed patient information is required for individualized prediction and treatment, this ensemble-learning model will be useful for risk stratification of morbidity and mortality after HSCT from pretransplant characteristics.

The authors are grateful to Emi Furusaka, Miki Shirasu, Tomoko Okuda, and Megumi Oka for their expert data management and secretarial assistance and all members of the transplant centers in KSCTG for their dedicated care of patients and donors.

This work was supported by grants from AMED (JP18pc0101031), JSPS KAKENHI (18K08325), and Takeda Science Foundation (J.K.).

Contribution: J.K., I.M., and M.M. designed the study; M. Iwasaki, J.K., I.M., and L.M.A. conducted the study and drafted the manuscript; I.M. and L.M.A. analyzed the data; Y.A., T. Kondo, T.I., Y.U., K.I., T.A., A.Y., K.Y., M.N., N. Anzai, T.M., T. Kitano, M. Itoh, N. Arima, T.T., M.W., H.H., K.A., and A.T.-K. contributed to the data collection and the final draft of the study; and all authors read and approved the final manuscript.

Conflict-of-interest disclosure: J.K. received honoraria from Takeda Phamaceutical, Celgene, Novartis Pharma, Astellas Pharma, Chugai Pharmaceutical, Kyowa Kirin, Otsuka Pharmaceutical, Bristol Myers Squibb, JCR Pharmaceuticals, MSD, and Daiichi Sankyo outside the submitted work. J.K. has a patent blood disease prognosis prediction information generation system, information processing device, server, program, or method (patent number: JP2020-119383) pending to NextGeM Inc., Kyoto University. T. Kondo reported grants and honoraria from Asahi Kasei Pharma and honoraria from Otsuka Pharmaceutical, Sumitomo Dainippon Pharma, Astellas Pharma, MSD Pharma, Pfizer, Kyowa Kirin, and Chugai Pharmaceutical outside the submitted work. T.I. reported a grant from Ono Pharmaceutical outside the submitted work. K.I. reported honoraria from Ono Pharmaceutical, Celgene, Bristol Myers Squibb, Takeda Pharmaceutical, Novartis Pharma, Nippon Shinyaku, Chugai Pharmaceutical, Kyowa Kirin, Otsuka Pharmaceutical, and Astellas Pharma outside the submitted work. T. Kitano reported grants and honoraria from Kyowa Kirin, Chugai Pharmaceutical, and Eizai, grants from Teijin Pharma, and honoraria from Bristol Myers Squibb, Celgene, and Ono Pharmaceutical outside the submitted work. I.M., L.M.A., and M.M. have a patent blood disease prognosis prediction information generation system, information processing device, server, program, or method (patent number: JP2020-119383) pending to NextGeM Inc., Kyoto University. A.T.-K. reported grants and honoraria from Celgene, Bristol Myers Squibb, Asteras Pharma, and Kyowa Kirin, grants from Ono Pharmaceutical, Thyas, Takeda Pharmaceutical, Chugai Pharmaceutical, Eizai, Nippon Shinyaku, Otsuka Pharmaceutical, Pfizer, Ohara Pharmaceutical, and Sanofi, and honoraria from Novartis Pharma, and MSD outside the submitted work. The remaining authors declare no competing financial interests.

A full list of study group members appears in “ Appendix.”

Correspondence: Junya Kanda, Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, Japan, 606-8507; e-mail: jkanda16@kuhp.kyoto-u.ac.jp.

Appendix: study group members

Investigators of the Kyoto Stem Cell Transplantation Group have 2 different statuses: authors and collaborators. Authors: Makoto Iwasaki, Junya Kanda, Yasuyuki Arai, Tadakazu Kondo, Takayuki Ishikawa, Yasunori Ueda, Kazunori Imada, Takashi Akasaka, Akihito Yonezawa, Kazuhiro Yago, Masaharu Nohgawa, Naoyuki Anzai, Toshinori Moriguchi, Toshiyuki Kitano, Mitsuru Itoh, Nobuyoshi Arima, Tomoharu Takeoka, Mitsumasa Watanabe, Hirokazu Hirata, Kosuke Asagoe, and Akifumi Takaori-Kondo. Collaborators: Masakatsu Hishizawa, Rie Tabata, Takashi Ikeda, Yoshitomo Maesako, Noboru Yonetani, Satoshi Oka, Satoshi Yoshioka, Nobuhiro Hiramoto, Fumiya Wada, Yutaka Shimazu, Chisaki Mizumoto, Yusuke Tashiro, Naoki Tamura, Takuto Mori, Tomohiro Taya, Tomoki Iemura, Hiroyuki Matsui, Yoshinobu Konishi, Kiyotaka Izumi, Hiroyuki Muranushi, Mizuki Watanabe, Suguru Takeuchi, Kensuke Nakao, and Mari Morita-Fujita.

1.
Martino
R
,
de Wreede
L
,
Fiocco
M
, et al;
Acute Leukemia Working Party the subcommittee for Myelodysplastic Syndromes of the Chronic Malignancies Working Party of the European group for Blood Marrow Transplantation Group (EBMT)
.
Comparison of conditioning regimens of various intensities for allogeneic hematopoietic SCT using HLA-identical sibling donors in AML and MDS with <10% BM blasts: a report from EBMT
.
Bone Marrow Transplant.
2013
;
48
(
6
):
761
-
770
.
2.
Bornhäuser
M
,
Kienast
J
,
Trenschel
R
, et al
.
Reduced-intensity conditioning versus standard conditioning before allogeneic haemopoietic cell transplantation in patients with acute myeloid leukaemia in first complete remission: a prospective, open-label randomised phase 3 trial
.
Lancet Oncol.
2012
;
13
(
10
):
1035
-
1044
.
3.
Ossenkoppele
GJ
,
Janssen
JJWM
,
van de Loosdrecht
AA
.
Risk factors for relapse after allogeneic transplantation in acute myeloid leukemia
.
Haematologica.
2016
;
101
(
1
):
20
-
25
.
4.
Arai
Y
,
Takeda
J
,
Aoki
K
, et al;
AML and MDS Working Group of the Japan Society for Hematopoietic Cell Transplantation
.
Efficiency of high-dose cytarabine added to CY/TBI in cord blood transplantation for myeloid malignancy
.
Blood.
2015
;
126
(
3
):
415
-
422
.
5.
Sorror
ML
,
Maris
MB
,
Storb
R
, et al
.
Hematopoietic cell transplantation (HCT)-specific comorbidity index: a new tool for risk assessment before allogeneic HCT
.
Blood.
2005
;
106
(
8
):
2912
-
2919
.
6.
Gratwohl
A
,
Hermans
J
,
Goldman
JM
, et al;
Chronic Leukemia Working Party of the European Group for Blood and Marrow Transplantation
.
Risk assessment for patients with chronic myeloid leukaemia before allogeneic blood or marrow transplantation
.
Lancet.
1998
;
352
(
9134
):
1087
-
1092
.
7.
Shouval
R
,
Bondi
O
,
Mishan
H
,
Shimoni
A
,
Unger
R
,
Nagler
A
.
Application of machine learning algorithms for clinical predictive modeling: a data-mining approach in SCT
.
Bone Marrow Transplant.
2014
;
49
(
3
):
332
-
337
.
8.
Shouval
R
,
Labopin
M
,
Bondi
O
, et al
.
Prediction of allogeneic hematopoietic stem-cell transplantation mortality 100 days after transplantation using a machine learning algorithm: A European Group for Blood and Marrow Transplantation Acute Leukemia Working Party retrospective data mining study
.
J Clin Oncol.
2015
;
33
(
28
):
3144
-
3151
.
9.
Shouval
R
,
Labopin
M
,
Unger
R
, et al
.
Prediction of hematopoietic stem cell transplantation related mortality: lessons learned from the in-silico approach. A European Society for Blood and Marrow Transplantation Acute Leukemia Working Party data mining study
.
PLoS One.
2016
;
11
(
3
):
e0150637
.
10.
Buturovic
L
,
Shelton
J
,
Spellman
SR
, et al
.
Evaluation of a machine learning-based prognostic model for unrelated hematopoietic cell transplantation donor selection
.
Biol Blood Marrow Transplant.
2018
;
24
(
6
):
1299
-
1306
.
11.
Arai
Y
,
Kondo
T
,
Fuse
K
, et al
.
Using a machine learning algorithm to predict acute graft-versus-host disease following allogeneic transplantation
.
Blood Adv.
2019
;
3
(
22
):
3626
-
3634
.
12.
Ishwaran
H
,
Gerds
TA
,
Kogalur
UB
,
Moore
RD
,
Gange
SJ
,
Lau
BM
.
Random survival forests for competing risks
.
Biostatistics.
2014
;
15
(
4
):
757
-
773
.
13.
Shouval
R
,
Ruggeri
A
,
Labopin
M
, et al
.
An integrative scoring system for survival prediction following umbilical cord blood transplantation in acute leukemia
.
Clin Cancer Res.
2017
;
23
(
21
):
6478
-
6486
.
14.
Okamura
H
,
Nakamae
M
,
Koh
S
, et al
Interactive web application for plotting personalized prognosis prediction curves in allogeneic hematopoietic cell transplantation using machine learning
.
Transplantation.
2021
;
105
(
5
):
1090
-
1096
.
15.
Holtan
SG
,
DeFor
TE
,
Lazaryan
A
, et al
.
Composite end point of graft-versus-host disease-free, relapse-free survival after allogeneic hematopoietic cell transplantation
.
Blood.
2015
;
125
(
8
):
1333
-
1338
.
16.
Ruggeri
A
,
Labopin
M
,
Ciceri
F
,
Mohty
M
,
Nagler
A
.
Definition of GvHD-free, relapse-free survival for registry-based studies: an ALWP-EBMT analysis on patients with AML in remission
.
Bone Marrow Transplant.
2016
;
51
(
4
):
610
-
611
.
17.
Przepiorka
D
,
Weisdorf
D
,
Martin
P
, et al
.
1994 consensus conference on acute GVHD grading
.
Bone Marrow Transplant.
1995
;
15
(
6
):
825
-
828
.
18.
Sullivan
KM
,
Agura
E
,
Anasetti
C
, et al
.
Chronic graft-versus-host disease and other late complications of bone marrow transplantation
.
Semin Hematol.
1991
;
28
(
3
):
250
-
259
.
19.
Giralt
S
,
Ballen
K
,
Rizzo
D
, et al
.
Reduced-intensity conditioning regimen workshop: defining the dose spectrum. Report of a workshop convened by the Center for International Blood and Marrow Transplant Research
.
Biol Blood Marrow Transplant.
2009
;
15
(
3
):
367
-
369
.
20.
Armand
P
,
Kim
HT
,
Logan
BR
, et al
.
Validation and refinement of the disease risk index for allogeneic stem cell transplantation
.
Blood.
2014
;
123
(
23
):
3664
-
3671
.
21.
Kanda
J
,
Kawase
T
,
Tanaka
H
, et al;
HLA Working Group of the Japan Society for Hematopoietic Cell Transplantation
.
Effects of haplotype matching on outcomes after adult single-cord blood transplantation
.
Biol Blood Marrow Transplant.
2020
;
26
(
3
):
509
-
518
.
22.
Collins
GS
,
Reitsma
JB
,
Altman
DG
,
Moons
KGM
.
Transparent Reporting of a multivariable prediction model for Individual Prognosis Or Diagnosis (TRIPOD): the TRIPOD statement
.
Ann Intern Med.
2015
;
162
(
1
):
55
-
63
.
23.
Opitz
D
,
Maclin
R
.
Popular ensemble methods: an empirical study
.
J Artif Intell Res.
1999
;
11
:
169
-
198
.
24.
Reid
N
:
The statistical analysis of failure time data [book review]
.
Can J Stat.
1982
;
10
(
1
):
64
-
66
.
25.
Ishwaran
H
,
Kogalur
UB
,
Blackstone
EH
,
Lauer
MS
.
Random survival forests
.
Ann Appl Stat.
2008
;
2
(
3
):
841
-
860
.
26.
Lee
C
,
Yoon
J
,
Schaar
MV
.
Dynamic-DeepHit: a deep learning approach for dynamic survival analysis with competing risks based on longitudinal data
.
IEEE Trans Biomed Eng.
2020
;
67
(
1
):
122
-
133
.
27.
Thongkam
J
,
Xu
G
,
Zhang
Y
,
Huang
F
.
Breast cancer survivability via AdaBoost algorithms
.
Proc. Second Australas. Workshop Health Data Knowl. Manag.
2008
;
80
:
55
-
64
.
28.
Chen
T
,
Guestrin
C
. XGBoost: a scalable tree boosting system. Proc. 22nd ACM SIGKDD Int. Conf. Knowl. Discov. Data Min.
2016
;
785
-
794
.
29.
Geurts
P
,
Ernst
D
,
Wehenkel
L
.
Extremely randomized trees
.
Mach Learn.
2006
;
63
(
1
):
3
-
42
.
30.
Hothorn
T
,
Lausen
B
,
Benner
A
,
Radespiel-Tröger
M
.
Bagging survival trees
.
Stat Med.
2004
;
23
(
1
):
77
-
91
.
31.
Chen
Y
,
Jia
Z
,
Mercola
D
,
Xie
X
.
A gradient boosting algorithm for survival analysis via direct optimization of concordance index
.
Comput Math Methods Med.
2013
;
2013
:
873595
.
32.
Liu
P
,
Fu
B
,
Yang
SX
.
HitBoost: survival analysis via a multi-output gradient boosting decision tree method
.
IEEE Access.
2019
;
7
:
56785
-
56795
.
33.
Lundberg
SM
,
Lee
S-I
.
A unified approach to interpreting model predictions
.
Adv Neural Inf Process Syst.
2017
;
30
:
4765
-
4774
.
34.
Uno
H
,
Cai
T
,
Pencina
MJ
,
D’Agostino
RB
,
Wei
LJ
.
On the C-statistics for evaluating overall adequacy of risk prediction procedures with censored survival data
.
Stat Med.
2011
;
30
(
10
):
1105
-
1117
.
35.
Wolbers
M
,
Blanche
P
,
Koller
MT
,
Witteman
JCM
,
Gerds
TA
.
Concordance for prognostic models with competing risks
.
Biostatistics.
2014
;
15
(
3
):
526
-
539
.
36.
Austin
PC
,
Harrell
FE
Jr
,
van Klaveren
D
.
Graphical calibration curves and the integrated calibration index (ICI) for survival models
.
Stat Med.
2020
;
39
(
21
):
2714
-
2742
.
37.
Gooley
TA
,
Leisenring
W
,
Crowley
J
,
Storer
BE
.
Estimation of failure probabilities in the presence of competing risks: new representations of old estimators
.
Stat Med.
1999
;
18
(
6
):
695
-
706
.
38.
Sachs
MC
,
Discacciati
A
,
Everhov
ÅH
,
Olén
O
,
Gabriel
EE
.
Ensemble prediction of time‐to‐event outcomes with competing risks: a case‐study of surgical complications in Crohn’s disease
.
J R Stat Soc Ser C Appl Stat.
2019
;
68
(
5
):
1431
-
1446
.
39.
Magenau
J
,
Braun
T
,
Gatza
E
, et al
.
Assessment of individual versus composite endpoints of acute graft-versus-host disease in determining long-term survival after allogeneic transplantation
.
Biol Blood Marrow Transplant.
2019
;
25
(
8
):
1682
-
1688
.
40.
Storb
R
,
Gyurkocza
B
,
Storer
BE
, et al
.
Graft-versus-host disease and graft-versus-tumor effects after allogeneic hematopoietic cell transplantation
.
J Clin Oncol.
2013
;
31
(
12
):
1530
-
1538
.
41.
Kurosawa
S
,
Oshima
K
,
Yamaguchi
T
, et al
.
Quality of life after allogeneic hematopoietic cell transplantation according to affected organ and severity of chronic graft-versus-host disease
.
Biol Blood Marrow Transplant.
2017
;
23
(
10
):
1749
-
1758
.
42.
Schetelig
J
,
de Wreede
LC
,
Andersen
NS
, et al;
CLL subcommittee, Chronic Malignancies Working Party
.
Centre characteristics and procedure-related factors have an impact on outcomes of allogeneic transplantation for patients with CLL: a retrospective analysis from the European Society for Blood and Marrow Transplantation (EBMT)
.
Br J Haematol.
2017
;
178
(
4
):
521
-
533
.
43.
Kanda
J
,
Saji
H
,
Fukuda
T
, et al
.
Related transplantation with HLA-1 Ag mismatch in the GVH direction and HLA-8/8 allele-matched unrelated transplantation: a nationwide retrospective study
.
Blood.
2012
;
119
(
10
):
2409
-
2416
.
44.
Kanda
Y
,
Kanda
J
,
Atsuta
Y
, et al
.
Impact of a single human leucocyte antigen (HLA) allele mismatch on the outcome of unrelated bone marrow transplantation over two time periods. A retrospective analysis of 3003 patients from the HLA Working Group of the Japan Society for Blood and Marrow Transplantation
.
Br J Haematol.
2013
;
161
(
4
):
566
-
577
.
45.
Morishima
Y
,
Kashiwase
K
,
Matsuo
K
, et al;
Japan Marrow Donor Program
.
Biological significance of HLA locus matching in unrelated donor bone marrow transplantation
.
Blood.
2015
;
125
(
7
):
1189
-
1197
.
46.
Morishima
S
,
Shiina
T
,
Suzuki
S
, et al;
Japan Marrow Donor Program
.
Evolutionary basis of HLA-DPB1 alleles affects acute GVHD in unrelated donor stem cell transplantation
.
Blood.
2018
;
131
(
7
):
808
-
817
.
47.
Papaemmanuil
E
,
Gerstung
M
,
Bullinger
L
, et al
.
Genomic classification and prognosis in acute myeloid leukemia
.
N Engl J Med.
2016
;
374
(
23
):
2209
-
2221
.
48.
Lindsley
RC
,
Saber
W
,
Mar
BG
, et al
.
Prognostic mutations in myelodysplastic syndrome after stem-cell transplantation
.
N Engl J Med.
2017
;
376
(
6
):
536
-
547
.
49.
Nazha
A
,
Hu
Z-H
,
Wang
T
, et al
.
A personalized prediction model for outcomes after allogeneic hematopoietic cell transplant in patients with myelodysplastic syndromes
.
Biol Blood Marrow Transplant.
2020
;
26
(
11
):
2139
-
2146
.
50.
Gandelman
JS
,
Byrne
MT
,
Mistry
AM
, et al
.
Machine learning reveals chronic graft-versus-host disease phenotypes and stratifies survival after stem cell transplant for hematologic malignancies
.
Haematologica.
2019
;
104
(
1
):
189
-
196
.

Author notes

Requests for data sharing may be submitted to Junya Kanda (jkanda16@kuhp.kyoto-u.ac.jp).

The full-text version of this article contains a data supplement.

Supplemental data