Purpose: Apoptosis is essential for chemotherapy responses. In this discovery and validation study, we evaluated the suitability of a mathematical model of apoptosis execution (APOPTO-CELL) as a stand-alone signature and as a constituent of further refined prognostic stratification tools.

Experimental Design: Apoptosis competency of primary tumor samples from patients with stage III colorectal cancer (n = 120) was calculated by APOPTO-CELL from measured protein concentrations of Procaspase-3, Procaspase-9, SMAC, and XIAP. An enriched APOPTO-CELL signature (APOPTO-CELL-PC3) was synthesized to capture apoptosome-independent effects of Caspase-3. Furthermore, a machine learning Random Forest approach was applied to APOPTO-CELL-PC3 and available molecular and clinicopathologic data to identify a further enhanced signature. Association of the signature with prognosis was evaluated in an independent colon adenocarcinoma cohort (TCGA COAD, n = 136).

Results: We identified 3 prognostic biomarkers (P = 0.04, P = 0.006, and P = 0.0004 for APOPTO-CELL, APOPTO-CELL-PC3, and Random Forest signatures, respectively) with increasing stratification accuracy for patients with stage III colorectal cancer.

The APOPTO-CELL-PC3 signature ranked highest among all features. The prognostic value of the signatures was independently validated in stage III TCGA COAD patients (P = 0.01, P = 0.04, and P = 0.02 for APOPTO-CELL, APOPTO-CELL-PC3, and Random Forest signatures, respectively). The signatures provided further stratification for patients with CMS1-3 molecular subtype.

Conclusions: The integration of a systems-biology–based biomarker for apoptosis competency with machine learning approaches is an appealing and innovative strategy toward refined patient stratification. The prognostic value of apoptosis competency is independent of other available clinicopathologic and molecular factors, with tangible potential of being introduced in the clinical management of patients with stage III colorectal cancer. Clin Cancer Res; 23(5); 1200–12. ©2016 AACR.

This article is featured in Highlights of This Issue, p. 1121

Translational Relevance

Despite extensive research efforts, powerful biomarkers capable of identifying patients with stage III colorectal cancer at high risk for chemotherapy resistance, and thus relapse, remain an unmet clinical demand. Lack of response to chemotherapeutics has been ascribed to defective apoptosis. Using an integrative stepwise approach, we investigated the potential clinical utility of a mathematical model of caspase activation for identifying patient-specific risk of unfavorable outcomes. Among all clinicopathologic, demographic, and molecular predictors analyzed, enriched-apoptosis systems modeling delivered the highest-ranking independent prognostic biomarker. Furthermore, apoptosis modeling can be combined with molecular tumor subtyping to further refine risk predictions. We report the clinical validation of diagnostic tools for patients with stage III colorectal cancer that could deliver superior quality of care by personalizing cancer treatment.

Colorectal cancer is a leading contributor to morbidity and mortality worldwide (1). Colorectal cancer incidence is expected to increase due to longer life expectancy and lifestyle changes, such as poor diet and decreased physical activity (1). The current standard-of-care for patients with stage III colorectal cancer prescribes surgical resection followed by 5-fluorouracil (5-FU)-based adjuvant chemotherapy (2). Response to treatment varies greatly, and numerous studies have attempted to reconcile differential response to chemotherapy with the underlying molecular characteristics of the primary tumor (3). The mechanism of action of chemotherapeutic agents, such as 5-FU, is centered on their DNA-damaging effects and ability to induce cell-cycle arrest and apoptosis (4). Alterations in apoptotic signaling pathways foster tumor progression and contribute to insufficient response to treatment (5, 6). The major pathway of apoptosis activation by 5-FU and other genotoxic drugs is mitochondrial outer membrane permeabilization (MOMP), which triggers a complex signaling network of pro- and anti-apoptotic proteins controlling the activation of effector caspases (7). Cytochrome c and SMAC are released into the cytosol following MOMP and activate Apaf-1 and Procaspase-9 or antagonize the major caspase inhibitor XIAP. Caspase-9 can activate Caspase-3, which drives multiple positive feedback loops that ensure an efficient execution of apoptotic cell death (7).

Systems analysis of apoptosis signaling has the potential to deliver superior prognostic markers by accounting for the relative abundance of key proteins, network connectivity, and regulations of the pathways involved (8, 9). In previous work, our group developed a mathematical model, APOPTO-CELL, on the basis of ordinary differential equations that reliably simulates signaling kinetics within the caspase activation network downstream of MOMP (10, 11). As input, APOPTO-CELL requires protein concentrations of the key downstream apoptosis regulators Apaf-1, Procaspase-9, XIAP, SMAC, and Procaspase-3 to calculate cellular apoptosis competency. Apoptosis competency is calculated as the ability of Caspase-3 to cleave sufficient amounts of its substrates to ensure efficient execution of apoptosis. The mathematical model has been extensively validated in-house using single cell- and population-based approaches in cervical, colorectal, and glioblastoma cell line models (10, 12, 13). In initial proof-of-concept work, high apoptosis competency was associated with better outcome among chemotherapy-treated patients with colorectal cancer and glioblastoma (13–15).

Here, we performed a discovery and validation study aimed at developing stratification signatures to predict personalized risk in stage III colorectal cancer (Fig. 1). We investigated the potential of APOPTO-CELL as a stand-alone tool. Moreover, we synthetized a systems-biology signature by linking APOPTO-CELL with protein expression data and harnessed a machine learning approach to examine the prognostic relevance of ordinary differential equation (ODE)-based modeling, proteomics, and clinicopathologic data. We validated our signatures in an independent external cohort and assessed their prognostic value for distinct molecular subtypes of colorectal cancer.

Figure 1.

Workflow for the stepwise development and validation of personalized risk signatures in patients with stage III colorectal cancer. Three signatures with increasing prognostic accuracy were developed in a stepwise integrative approach on the basis of ODEs modeling, proteomics, and clinicopathologic features in the discovery cohort. The prognostic value of the apoptosis-based signatures was assessed in the context of microsatellite instability and BRAF mutational status using an expansion cohort. Validation was performed in an external cohort, including the evaluation of the prognostic value for colorectal cancer molecular subtypes.

Figure 1.

Workflow for the stepwise development and validation of personalized risk signatures in patients with stage III colorectal cancer. Three signatures with increasing prognostic accuracy were developed in a stepwise integrative approach on the basis of ODEs modeling, proteomics, and clinicopathologic features in the discovery cohort. The prognostic value of the apoptosis-based signatures was assessed in the context of microsatellite instability and BRAF mutational status using an expansion cohort. Validation was performed in an external cohort, including the evaluation of the prognostic value for colorectal cancer molecular subtypes.

Close modal

Patient cohorts

We developed prognostic signatures for patients with stage III colorectal cancer from 3 distinct and independent collections: discovery, expansion, and validation cohorts. The discovery cohort was an in-house multicenter study of patients with stage III colorectal cancer (n = 120). The expansion dataset included stage III colon cancer participants (n = 157) with known microsatellite instability (MSI) and BRAF status (GEO repository under accession 39582; ref. 16). The validation cohort was composed of patients with stage I–IV colon cancer (n = 136; COAD) from The Cancer Genome Atlas (TCGA) project.

A detailed description of the patients data handling and inclusion criteria for downstream analyses is presented in Supplementary Methods SM1.

Prognostic signatures

We developed 3 prognostic signatures: APOPTO-CELL, APOPTO-CELL-PC3, and RF.

APOPTO-CELL signature

APOPTO-CELL is a mathematical model of caspase activation resulting in apoptosis (10, 11). The model is composed of 53 ODEs, 19 state variables, and 75 kinetic parameters. It calculates apoptosis execution kinetics from the input proteins Apaf-1, Procaspase-9, XIAP, SMAC, and Procaspase-3. Previous quantitative studies found that Apaf-1 protein levels were not rate-limiting for apoptosome formation in colon cancer cells (14). Thus, Apaf-1 patient-specific protein levels were replaced by the median expression (0.123 μmol/L) previously determined in stage II/III colorectal cancer tumors (14). An amount of more than 25% substrate cleavage (SC) by active Caspase-3 served as the APOPTO-CELL prediction for apoptosis competency in line with previous single-cell imaging findings (10). While APOPTO-CELL was designed to use protein concentrations as inputs, surrogates such as RNA transcripts or gene expression can be used when protein expression is not available.

Molar protein concentrations were estimated from the normalized signal intensities via a novel pipeline illustrated in Fig. 2A.

Figure 2.

The APOPTO-CELL signature derived from ODE-based computations of absolute protein concentrations in tumor resections is a prognostic marker for patients with stage III colorectal cancer. A, Workflow of the pipeline developed to estimate absolute protein concentrations from the normalized signal intensities determined by RPPA (see Materials and Methods). B, Normalized RPPA signal intensities (i), reference distribution (ii), transformation function (iii), and estimated concentrations (iv) for Procaspase-9, XIAP, SMAC, and Procaspase-3. Data are color-coded on the basis of data density. Further details on these procedures are provided in the Materials and Methods. C, APOPTO-CELL–based simulations for the cleavage of effector caspase substrates are shown for patients with stage III colorectal cancer (n = 120). Time zero represents the event of MOMP. The dashed line at 25% substrate cleavage represents the decision threshold beyond which tumor cells are committed to die. D, Simulation results from (C) were binned using 10 percentage points increments in substrate cleavage. The majority of patient tumors presented with very low or very high apoptosis competency. E and F, Kaplan–Meier plots of DFS (E) and OS (F) are shown. Patients were stratified according to the APOPTO-CELL signature [substrate cleavage reached at 300 minutes in C and D being above (n = 77) or below (n = 43) the threshold of 25%]. P values were determined by log-rank tests.

Figure 2.

The APOPTO-CELL signature derived from ODE-based computations of absolute protein concentrations in tumor resections is a prognostic marker for patients with stage III colorectal cancer. A, Workflow of the pipeline developed to estimate absolute protein concentrations from the normalized signal intensities determined by RPPA (see Materials and Methods). B, Normalized RPPA signal intensities (i), reference distribution (ii), transformation function (iii), and estimated concentrations (iv) for Procaspase-9, XIAP, SMAC, and Procaspase-3. Data are color-coded on the basis of data density. Further details on these procedures are provided in the Materials and Methods. C, APOPTO-CELL–based simulations for the cleavage of effector caspase substrates are shown for patients with stage III colorectal cancer (n = 120). Time zero represents the event of MOMP. The dashed line at 25% substrate cleavage represents the decision threshold beyond which tumor cells are committed to die. D, Simulation results from (C) were binned using 10 percentage points increments in substrate cleavage. The majority of patient tumors presented with very low or very high apoptosis competency. E and F, Kaplan–Meier plots of DFS (E) and OS (F) are shown. Patients were stratified according to the APOPTO-CELL signature [substrate cleavage reached at 300 minutes in C and D being above (n = 77) or below (n = 43) the threshold of 25%]. P values were determined by log-rank tests.

Close modal

First, a kernel distribution object was constructed from the normalized protein intensities (or surrogate transcript amounts) with the MATLAB function fitdist (Statistics Toolbox) with a normal kernel smoothing, unbounded kernel support, and default bandwidth (steps 1–2). This step resulted in a smooth nonparametric representation of the distribution of the protein (or surrogate) concentrations (step 3). Second, a kernel probability distribution object was constructed from the reference distribution with the same method as for the protein (or surrogate) distribution, except with positive kernel support (right-hand side, steps 1–3). Third, the kernel distribution of the protein (or surrogate) intensities was used to calculate the smoothed cumulative distribution corresponding to the protein (or surrogate) intensity of each patient (left-hand side, step 4). Finally, the protein (or surrogate) cumulative distribution for each patient was converted into a corresponding absolute concentration using the inverse cumulative distribution of the reference distribution kernel (MATLAB function icdf from the Statistics Toolbox, step 5). Step 6 shows the transformation function from protein (or surrogate) signal intensities to estimated protein concentration.

We estimated APOPTO-CELL inputs from reverse-phase protein arrays (RPPAs), gene expression, and RNA transcripts for the discovery, expansion, and validation cohorts, respectively (Supplementary Methods SM2).

APOPTO-CELL-PC3 signature

Procaspase-3 exerts pivotal roles in multiple aspects of apoptosis both via apoptosome-dependent and -independent pathways. Thus, we generated a new signature (APOPTO-CELL-PC3) by combining the APOPTO-CELL model output with the expression of Procaspase-3 (Results and Fig. 4A). As with the APOPTO-CELL signature, Procaspase-3 concentration was estimated from RPPA, gene expression, and RNA transcripts for the discovery, expansion, and validation cohorts, respectively.

Random Forest signature

The Random Forest (RF) machine learning approach was conducted in MATLAB [built-in TreeBagger class (Statistics Toolbox) with 100.001 trees, surrogate decision split flags “on”; other options were set to “default”] and was used to calculate the RF classifier as a predictor for disease recurrence (17). We selected 36 months as a cutoff, as most recurrence events (87.9%) occurred within this time. For training purposes, we only considered patients known to have a recurrence or to be recurrence-free at 36 months (only 12.5% of patients were lost to follow-up). The training set consisted of two thirds of the patients, randomly sampled for each decision tree in the RF. For validation, recurrence predictions averaged from RF decision trees were analyzed for accuracy on patients that did not contribute to defining these trees (“out of bag” predictions). This allowed an unbiased validation without a separate validation cohort. The importance of each variable as a predictor was assessed by calculating the Permuted Variable Delta Error, a measurement for the increase in the prediction error upon random perturbation of the predictor values. The predictors that contributed positively to the accuracy of the RF (permuted variable delta > 0.05) were used to define the reduced RF classifier. Classifier performance was assessed by the area under the receiver operating characteristic (ROC) curve computed from the validation results.

We generated a synthetic cohort to further understand how the RF classifier makes predictions. This synthetic cohort had one entry for each unique hypothetical patient that each of the decision trees included in the RF ensemble was learned from. The complete set of hypothetical patients was compiled from all permutations of the unique values of each predictor. For each categorical predictor, values were given by each level (e.g., low-, medium-, and high-risk for the APOPTO-CELL-PC3 signature). For discrete numerical predictors (age and nodal count), the full set of values was taken from the complete list of unique cutoff points extracted from all the trees included in the RF ensemble.

We used the distribution of the patient-specific recurrence probabilities determined by the RF classifier in the discovery cohort to define risk groups (Fig. 5E). Survival analysis was performed using all patients (n = 120), including patients lost to follow-up who did not contribute to the RF classifier identification. The probability of recurrence, and thus the risk group, was computed on the basis of the out-of-bag predictions for patients included in the training set and de novo predictions for patients lost to follow-up within the first 36 months.

Statistical analysis

Kaplan–Meier estimates were used to compare disease-free (DFS) and overall survival (OS) curves between groups and statistically significant differences were determined by log-rank tests. We used univariate and multivariate Cox proportional hazard models to estimate the relative risks of outcome associated with the signatures (APOPTO-CELL, APOPTO-CELL-PC3, and RF) and clinical factors. We computed HRs and 95% confidence intervals (CIs) and we evaluated statistical significance with likelihood ratio tests. An in-depth account of the survival analyses is provided in Supplementary Methods SM3. Association between APOPTO-CELL and APOPTO-CELL-PC3 signatures with T stage and lymphovascular invasion was assessed by χ2 test.

Data processing and statistical analysis were performed in MATLAB (MATLAB and Statistics Toolbox Release 2014b, The MathWorks, Inc.) unless stated otherwise. Median follow-up time among censored patients, log-rank tests, Cox regression modeling, and assessment of proportional hazards assumptions were conducted in R (version 3.3.0) employing the functions Surv, coxph, and cox.zph from the package “survival” (version 2.39-2) and Anova from the package “car” (version 2.1-2). All tests were 2-sided and P < 0.05 was considered statistically significant.

Quantitative profiling of apoptosis execution proteins in a discovery cohort of patients with colorectal cancer

We explored whether mathematical modeling of effector caspase activation in the mitochondrial apoptosis pathway could identify high-risk patients in a multicenter cohort of patients with stage III colorectal cancer (n = 120; Fig. 1). Supplementary Table ST1 presents clinicopathologic and demographic characteristics.

Protein amounts of Procaspase-9, XIAP, SMAC, and Procaspase-3 were quantified in primary tumor samples by RPPAs (Supplementary Methods SM2).

We established a workflow to determine the molar concentrations required by APOPTO-CELL as input from normalized RPPA data (Fig. 2A and Materials and Methods). For each protein, absolute concentrations (Fig. 2B, iv) were estimated from normalized RPPA signal intensities (Fig. 2B, i) via a transformation function (Fig. 2B, iii) determined from a reference distribution (Fig. 2B, ii). We previously determined these reference distributions in fresh tumor tissues from a colorectal cancer cohort, with analogous demographic and clinicopathologic characteristics, using quantitative Western blotting (14). Figure 2B (i–iv) presents the normalized RPPA signal intensities, reference distributions, results from the alignments, and final estimated concentrations for all proteins from patient tumor samples included in the discovery cohort.

APOPTO-CELL delivers a stratification signature for patients with stage III colorectal cancer of the discovery cohort treated with 5-FU–based adjuvant chemotherapy

Tumor concentrations of Procaspase-9, XIAP, SMAC, and Procaspase-3 were input into the APOPTO-CELL mathematical model. We calculated apoptosis execution kinetics for a duration of 300 minutes (14) and computed substrate cleavage (SC) over time as a surrogate for the efficiency of apoptosis execution. Substrate cleavage profiles demonstrated that tumor apoptosis competency was variable between patients (Fig. 2C). We observed 2 major subpopulations displaying ≥ 90% or ≤ 10% substrate cleavage (58% and 32% of patient tumors, Fig. 2D), reflecting very high and low apoptosis competency, respectively. Thus, apoptosis execution functions as a biologic all-or-nothing switch between apoptosis competency and resistance, emanating from the systems-level interplay of key proteins (18, 19).

We investigated whether substrate cleavage was associated with clinical outcome among patients with stage III colorectal cancer treated with adjuvant chemotherapy. As 25% substrate cleavage serves as an optimal decision threshold for apoptotic cell death in cancer cell lines (10, 12) and colorectal cancer patient tumors (14), patients were dichotomized as those with SC > 25% or SC ≤ 25% at 300 minutes (Fig. 2C and D). We observed statistically significant differences between OS and DFS curves when comparing these groups (log-rank P < 0.05; Fig. 2E and F). Patients with SC ≤ 25% had increased risks of relapse (HR, 2.05; 95% CI, 1.03–4.05; P = 0.04) and death (HR, 3.78; 95% CI, 1.42–10.08; P = 0.006) compared with those with SC > 25%.

Sensitivity analyses demonstrate high robustness of the APOPTO-CELL signature

We examined whether APOPTO-CELL predictions of apoptosis susceptibility would notably change when accounting for noise in the protein concentrations (Fig. 3A). For each patient, we built a normal distribution centered on the reference (unperturbed) value with a given SD for each of the input proteins (including Apaf-1). Next, for each patient, we ran 1,000 simulations with values randomly drawn from these distributions (bootstrapping; ref. 20) and computed the predictions for apoptosis competency. We then computed a robustness index (RI) as the fraction of simulations matching the observed substrate cleavage (SC > 25% vs. SC ≤ 25%) in the absence of noise. We investigated the RI for up to 30% variation (upper limit for intercellular heterogeneity observed in refs. 21, 22) and defined patient-specific predictions as robust when RI ≥ 90% (gray line in Fig. 3B). When increasing the SD from 10% to 30%, we observed a marginal decrease (87% to 71%) in the number of patients consistently classified as low- versus high-risk. We previously found the relative standard variation in intratumor protein amounts in a set of patients with colorectal cancer to be approximately 11% (14). Thus, in this study, we selected 10% as the coefficient of variation when assessing robustness in downstream analyses. Visualizing the distribution of substrate cleavage simulations revealed that despite adding variation, apoptosis competency remained stable in regions of very low or high substrate cleavage for most patients (Fig. 3C). Next, we sought to examine the clinical implications of the robustness analyses. We categorized patients into 3 subpopulations (“bootstrap robustness group”): (i) robust low-risk if at least 90% of the simulations classified the patient as low-risk, (ii) robust high-risk if at least 90% of the simulations classified the patients as high-risk, and (iii) non-robust if otherwise. We observed statistically significant differences in DFS (P = 0.01) and OS (P = 0.0004) curves when comparing the robust low- versus high-risk groups (Fig. 3D and E). Only 16 patients (13%) were not classified as low- or high-risk for at least 90% of the simulations (Fig. 3F). These results suggest that while a degree of uncertainty may be present in the protein measurements (in terms of both technical measurement error and true biological variability), APOPTO-CELL still retained its prognostic value. This robustness is a distinctive advantage over classical threshold-based stratification by protein biomarkers, for which continuous readouts and cutoff values are strongly influenced by measurement noise.

Figure 3.

APOPTO-CELL predictions are robust against perturbations in protein concentrations. A, Schematic representation of the workflow to test the robustness of the APOPTO-CELL signature by bootstrapping analysis. For each patient, perturbations (± a given SD) were applied to all protein concentrations and 1,000 simulations were run by randomly sampling with replacement from these parameterizations. B, Robustness of the apoptosis competency predictions as a function of the percentage of the SD in the APOPTO-CELL proteins. Patient predictions were defined as robust if the robustness index RI (percentage of the simulations matching the observed substrate cleavage values) ≥ 90% (gray line). C, Heatmap showing the distribution of the substrate cleavage values reached at 300 minutes for each of 1,000 simulations run with ±10% SD for each of the 120 patients of the discovery cohort (black, robust SC; shades of yellow, non-robust SC). Hierarchical clustering (on the left) is based on correlations (computed with average as linkage method) between rows of patients to highlight groups of patients who behaved similarly. D and E, Kaplan–Meier estimates and log-rank tests comparing DFS (D) and OS curves (E) by the bootstrap robustness group suggested that the association between APOPTO-CELL and prognosis is retained despite ±10% noise in protein concentrations. F, Breakdown of robust versus non-robust predictions obtained by applying ±10% SD for patients categorized as low- versus high-risk.

Figure 3.

APOPTO-CELL predictions are robust against perturbations in protein concentrations. A, Schematic representation of the workflow to test the robustness of the APOPTO-CELL signature by bootstrapping analysis. For each patient, perturbations (± a given SD) were applied to all protein concentrations and 1,000 simulations were run by randomly sampling with replacement from these parameterizations. B, Robustness of the apoptosis competency predictions as a function of the percentage of the SD in the APOPTO-CELL proteins. Patient predictions were defined as robust if the robustness index RI (percentage of the simulations matching the observed substrate cleavage values) ≥ 90% (gray line). C, Heatmap showing the distribution of the substrate cleavage values reached at 300 minutes for each of 1,000 simulations run with ±10% SD for each of the 120 patients of the discovery cohort (black, robust SC; shades of yellow, non-robust SC). Hierarchical clustering (on the left) is based on correlations (computed with average as linkage method) between rows of patients to highlight groups of patients who behaved similarly. D and E, Kaplan–Meier estimates and log-rank tests comparing DFS (D) and OS curves (E) by the bootstrap robustness group suggested that the association between APOPTO-CELL and prognosis is retained despite ±10% noise in protein concentrations. F, Breakdown of robust versus non-robust predictions obtained by applying ±10% SD for patients categorized as low- versus high-risk.

Close modal

Combining the APOPTO-CELL signature with Procaspase-3 expression (APOPTO-CELL-PC3) resulted in enhanced stratification in stage III patients of the discovery cohort

Apart from their defined roles in the mitochondrial apoptosis pathway, Procaspase-9, XIAP, SMAC, and Procaspase-3 may play roles in MOMP- and apoptosome-independent cell death pathways. They may also regulate other cellular processes such as proliferation, autophagy, immune response, and differentiation (23–29). As these processes could affect patient response to therapy, we examined whether these proteins were independently associated with clinical outcome. Only Procaspase-3 (PC3) showed potential as an independent biomarker (Supplementary Fig. S1). We therefore included Procaspase-3 levels as an additional prognostic factor in our analysis. This approach compensates for apoptosome-independent Procaspase-3 activation. To explore whether PC3 could further improve prognostic accuracy, we combined PC3 quantifications with APOPTO-CELL model outputs (APOPTO-CELL-PC3; Fig. 4A). We categorized patients into 3 groups: (i) high-risk, patients with low PC3 expression (≤ median) and apoptosis resistance (SC ≤ 25%); (ii) low-risk, patients with high PC3 expression (> median) and apoptosis competency (SC > 25%), and (iii) medium-risk, all other patients. We observed statistically significant differences between OS and DFS curves when comparing these groups (log-rank P < 0.01; Fig. 4B and C), which was driven by differences between the high- and low-risk groups (pairwise log-rank P < 0.01). Patients categorized as high-risk by the APOPTO-CELL-PC3 signature had significantly increased risks of relapse (HR, 3.90; 95% CI, 1.59–9.57; P = 0.008) and death (HR, 9.30; 95% CI, 2.06–41.98; P = 0.002) compared with patients categorized as low-risk. Harrell's concordance index suggested that the APOPTO-CELL-PC3 signature had superior prognostic discrimination compared with the APOPTO-CELL signature alone for both OS (0.73 vs. 0.67) and DFS (0.64 vs. 0.59).

Figure 4.

An enriched APOPTO-CELL signature (APOPTO-CELL-PC3) is a prognostic biomarker for stage III colorectal cancer that is independent of established clinicopathologic predictors. A, Workflow leading to the derivation of the enriched APOPTO-CELL signature on the basis of the combination of the APOPTO-CELL model output with the Procaspase-3 (PC3) expression to define patient subgroups. B and C, Kaplan–Meier plots comparing patient subgroups identified by the APOPTO-CELL-PC3 signature shown in (A) for DFS and OS. The enriched signature identifies 3 risk groups, with highly significant separation of high- and low-risk patients. P values were determined by log-rank tests. D, Estimated HRs, 95% CIs, and P values from likelihood ratio tests from univariate (white-shaded) and multivariate (gray-shaded) Cox proportional hazards models examining associations between clinical factors and the APOPTO-CELL-PC3 signature with the risks of recurrence (left) and death (right). The multivariate analysis (“APOPTO-CELL-PC3 signature adj.”) was adjusted for clinical variables found to be associated with outcome in the Cox univariate analysis (T stage and lymphovascular invasion).

Figure 4.

An enriched APOPTO-CELL signature (APOPTO-CELL-PC3) is a prognostic biomarker for stage III colorectal cancer that is independent of established clinicopathologic predictors. A, Workflow leading to the derivation of the enriched APOPTO-CELL signature on the basis of the combination of the APOPTO-CELL model output with the Procaspase-3 (PC3) expression to define patient subgroups. B and C, Kaplan–Meier plots comparing patient subgroups identified by the APOPTO-CELL-PC3 signature shown in (A) for DFS and OS. The enriched signature identifies 3 risk groups, with highly significant separation of high- and low-risk patients. P values were determined by log-rank tests. D, Estimated HRs, 95% CIs, and P values from likelihood ratio tests from univariate (white-shaded) and multivariate (gray-shaded) Cox proportional hazards models examining associations between clinical factors and the APOPTO-CELL-PC3 signature with the risks of recurrence (left) and death (right). The multivariate analysis (“APOPTO-CELL-PC3 signature adj.”) was adjusted for clinical variables found to be associated with outcome in the Cox univariate analysis (T stage and lymphovascular invasion).

Close modal

The APOPTO-CELL-PC3 signature is an independent prognostic marker for patients with stage III colorectal cancer of the discovery cohort

We used Cox proportional hazards regression models to examine the prognostic value of APOPTO-CELL-PC3 and established clinical risk factors (Fig. 4D). T stage and lymphovascular invasion were significantly associated with the risks of relapse or death in univariate analyses. Patients categorized as high-risk by the APOPTO-CELL or APOPTO-CELL-PC3 signature were significantly overrepresented in stage T4 (χ2P = 0.03 and χ2P = 0.006, respectively). No association was detected between lymphovascular invasion and the APOPTO-CELL (χ2P = 0.58) or APOPTO-CELL-PC3 signatures (χ2P = 0.65). When adjusting for T stage and lymphovascular invasion, patients categorized as high-risk by the APOPTO-CELL-PC3 signature had increased risks of relapse (HR, 3.26; 95% CI, 1.27–8.35; P = 0.04) and death (HR, 11.10; 95% CI, 2.35–52.33; P = 0.001) compared with patients categorized as low-risk.

We explored whether staging, primary tumor location, and lymphovascular invasion further aided in patient stratification (Supplementary Fig. S2). We observed significant differences between OS and DFS curves by lymphovascular invasion (log-rank P = 0.04 for both, Supplementary Fig. S2C, i–ii), although not for staging or tumor location (Supplementary Fig. S2A and S2B, i–ii). When stratifying by stage and location, we observed significant differences between OS and DFS curves (log-rank P < 0.05) by the APOPTO-CELL-PC3 signature among stage III-A/B and proximal tumors (Supplementary Fig. S2A and S2B, iii–iv). There were differences between DFS curves by APOPTO-CELL-PC3 among patients without lymphovascular invasion (Supplementary Fig. S2C, iii) and between OS curves among patients with lymphovascular invasion (Supplementary Fig. S2C, iv).

APOPTO-CELL and APOPTO-CELL-PC3 are independent prognostic markers of recurrence in the expansion cohort

The prognostic relevance of BRAF and MSI status has been comprehensively examined, and testing of MSI status has been introduced in clinical practice (2). We used an expansion cohort (GSE39582 dataset; ref. 16) to explore whether MSI and BRAF status potentially confound associations between our prognostic signatures and recurrence risk (Fig. 1). We analyzed stage III patients with known status for microsatellites and BRAF mutation (n = 157; Supplementary Table ST1).

APOPTO-CELL and APOPTO-CELL-PC3 signatures, estimated from gene expression (Supplementary Methods SM2), were associated with DFS in univariate and multivariate analyses (Supplementary Table ST2). In unadjusted analyses, patients categorized as high- versus low-risk by APOPTO-CELL (SC ≤ 25% vs. SC > 25%) had an increased risk of relapse (HR, 1.94; 95% CI, 1.17–3.24; P = 0.01). Patients categorized as high- and medium-risk by APOPTO-CELL-PC3 had an increased risk of relapse compared with those classified as low-risk (HR, 2.39; 95% CI, 1.20–4.78 and HR, 2.66; 95% CI, 1.32–5.34; P = 0.008). We fit 2 multivariate models (i) stratifying by MSI (dMMR vs. pMMR) status and adjusting for BRAF (wild-type vs. mutant) as well as treatment received (chemotherapy vs. no chemotherapy) and (ii) additionally adjusting for age (continuous linear), sex (male vs. female), tumor location (distal vs. proximal), and KRAS mutational status (wild-type vs. mutant). Relative risk estimates were similar between unadjusted and adjusted models, suggesting that these factors did not likely confound associations between our signatures and DFS. Furthermore, both signatures remained independent prognostic markers for DFS (Supplementary Table ST2).

A machine-learning RF signature for recurrence identifies the APOPTO-CELL-PC3 signature as the most salient predictor

We built a RF signature using the APOPTO-CELL-PC3 signature, the remaining single proteins (dichotomized as > or ≤ median), and the clinical variables analyzed in Fig. 4D (“RF-all predictors”). A reduced RF classifier was constructed using the most influential predictors of relapse (“RF-reduced predictors set”). The reduced classifier predicted disease recurrence without any loss in accuracy compared to the full RF classifier (McNemara P > 0.05). Thus, we performed our downstream analysis with the reduced classifier, which included the APOPTO-CELL-PC3 signature, T stage, sex, N stage, age, and nodal count (Fig. 5B). The AUC (see Materials and Methods) suggested this model satisfactorily predicted relapse (0.73; 95% CI, 0.62–0.74; Fig. 5C).

Figure 5.

A machine-learning RF signature for relapse identifies the APOPTO-CELL-PC3 signature as the most important predictor and sheds light on how distinct features contribute to the risk predictions. A, Workflow for the development of the machine-learning RF signature. B, Identified RF classifier consists of 6 predictors for recurrence, ranked here by importance (see Materials and Methods). C, Evaluation of the performance of the RF classifier by ROC analysis. Error bars represent 95% CIs obtained from 1,000 bootstraps. Inset represents area under the curve with 95% CI. D, Dependence plot highlighting how the predictors (and their interactions) contribute to the relative risk of recurrence predicted by the RF classifier. Size of the dot encodes the magnitude of the relative risk compared with baseline, whereas color indicates the direction of change (red and blue for increase and decrease, respectively). E, Distribution of the probability of recurrence for the patients used to develop the random forest signature highlighting the cutoff thresholds used to define risk groups. F, Kaplan–Meier estimates comparing DFS curves for the risk groups defined by the RF classifier in E. P values were computed by log-rank tests.

Figure 5.

A machine-learning RF signature for relapse identifies the APOPTO-CELL-PC3 signature as the most important predictor and sheds light on how distinct features contribute to the risk predictions. A, Workflow for the development of the machine-learning RF signature. B, Identified RF classifier consists of 6 predictors for recurrence, ranked here by importance (see Materials and Methods). C, Evaluation of the performance of the RF classifier by ROC analysis. Error bars represent 95% CIs obtained from 1,000 bootstraps. Inset represents area under the curve with 95% CI. D, Dependence plot highlighting how the predictors (and their interactions) contribute to the relative risk of recurrence predicted by the RF classifier. Size of the dot encodes the magnitude of the relative risk compared with baseline, whereas color indicates the direction of change (red and blue for increase and decrease, respectively). E, Distribution of the probability of recurrence for the patients used to develop the random forest signature highlighting the cutoff thresholds used to define risk groups. F, Kaplan–Meier estimates comparing DFS curves for the risk groups defined by the RF classifier in E. P values were computed by log-rank tests.

Close modal

RF classifiers are considered “black box” machine-learning algorithms that have limited interpretability. To investigate how the recurrence probability predicted by the RF classifier depended on each feature and their interactions (Supplementary Fig. S3), we generated a synthetic cohort where each patient was characterized by a unique combination of input features (see Materials and Methods). The RF classifier predicted a reduced likelihood of recurrence for patients categorized as low-risk by the APOPTO-CELL-PC3 signature, aged > 40, with a nodal count between 10 and 50, males, and those with less advanced T and N stage (Supplementary Fig. S3A–S3F).

We examined how the RF features interact to deploy the final recurrence predictions. For each combination of predictors, we visualized recurrence risk relative to the overall average recurrence risk (the reference baseline; Fig. 5D). The relative risks associated with age and nodal count were aggregated into 3 levels by averaging over all risks in the corresponding group. Supplementary Figure S3G presents this visualization without aggregation of these predictors. Overall, patients categorized as low-risk by the APOPTO-CELL-PC3 signature had a reduced recurrence risk. This effect was lost in advanced cancers (T4 and N2), which exhibited an overall higher probability of recurrence. Females also had a higher risk of relapse than males.

We categorized patients into 3 risk groups according to the probability of recurrence predicted by the reduced RF classifier: < 20%, 20%–50%, and > 50%. There were significant differences between DFS curves by these groups (log-rank P < 0.001; Fig. 5F). Patients with > 50% risk had an approximately 5-fold increased recurrence risk (HR, 5.13; 95% CI, 2.12–12.41; P = 0.002) compared with patients with < 20% risk. Results were similar after adjusting for T stage and lymphovascular invasion. Harrell's concordance index suggested that the RF signature was better at discriminating recurrence than the APOPTO-CELL-PC3 signature (0.67 vs. 0.64).

Validation of the prognostic signatures in an independent external cohort

We evaluated the prognostic value of our signatures using an independent publically available cohort: TCGA COAD (Fig. 1). Supplementary Table ST1 presents clinicopathologic and demographic characteristics of patients who met our inclusion criteria (Supplementary Methods SM1).

The 3 signatures identified in the discovery cohort were tested on stage III patients in the validation cohort. In this dataset, protein expression of SMAC and XIAP were available. For the amounts of Procaspase-3 and Procaspase-9, transcript abundance was used as surrogate for protein expression (Supplementary Methods SM2). Despite the limited sample size and follow-up time, we nevertheless observed statistically significant differences between DFS curves when comparing risk groups for the APOPTO-CELL, APOPTO-CELL-PC3, and RF signatures (log-rank P < 0.05; Fig. 6A–C).

Figure 6.

Validation of the prognostic value of the signatures identified in the discovery cohort and their relationship to the colorectal cancer CMS in an independent external cohort. A–C, Kaplan–Meier plots for DFS stratification of patients with stage III colon cancer on the basis of the APOPTO-CELL signature (A), the APOPTO-CELL-PC3 signature (B), and the RF signature (C). D–I, Kaplan–Meier estimates comparing DFS curves for patients with stage I to IV colon cancer on the basis of the APOPTO-CELL (D, G), the APOPTO-CELL-PC3 (E, H), and the RF (F, I) signatures, stratified by CMS1-3 versus CMS4.

Figure 6.

Validation of the prognostic value of the signatures identified in the discovery cohort and their relationship to the colorectal cancer CMS in an independent external cohort. A–C, Kaplan–Meier plots for DFS stratification of patients with stage III colon cancer on the basis of the APOPTO-CELL signature (A), the APOPTO-CELL-PC3 signature (B), and the RF signature (C). D–I, Kaplan–Meier estimates comparing DFS curves for patients with stage I to IV colon cancer on the basis of the APOPTO-CELL (D, G), the APOPTO-CELL-PC3 (E, H), and the RF (F, I) signatures, stratified by CMS1-3 versus CMS4.

Close modal

Evaluation of the prognostic value of the signatures in the context of the colorectal cancer consensus molecular subtypes

A recent study identified 4 distinct consensus molecular subtypes (CMS) for colorectal cancer (30), with CMS4 (“mesenchymal”) versus CMS1–3 subtypes being associated with poorer outcome. Among our validation cohort, there were no differences between DFS curves by CMS4 versus 1–3 subtypes among stage III (log-rank P = 0.90) or stage I–IV patients (log-rank P = 0.54). Nevertheless, substrate cleavage retained its prognostic value among CMS1–3 (log-rank P = 0.02), although not among CMS4 (log-rank P = 0.11; Fig. 6D). Analogously, the APOPTO-CELL-PC3 and RF signatures had prognostic value among CMS1–3, but not CMS4 (Fig. 6E and F).

We evaluated APOPTO-CELL, a mathematical model that calculates apoptosis competency of patient tumors, as a signature of stage III colorectal cancer patient outcome to 5-FU–based chemotherapy. Moreover, we present a novel systems medicine workflow to optimize model predictions. This study is the first large-scale demonstration that determining apoptosis competency by systems modeling adds independent prognostic value to current clinicopathologic markers.

Previous studies demonstrated that systems biological approaches toward apoptosis signaling may have great potential as novel avenues toward discovery of biomarkers (31) and therapeutic viable targets (e.g., SMAC-mimetics; ref. 32) resulting in an educated stratification of patients for clinical trials (33). Further development and exploitation of such new strategies is warranted, as despite extensive efforts to identify prognostic and predictive biomarkers for colorectal cancer TNM staging remains the most reliable prognostic marker that still drives treatment decisions. Other clinicopathologic tumor characteristics, such as tumor location, differentiation, and lymphovascular invasion, have been associated with prognosis and response to treatment (2). MSI and wild-type status for KRAS, BRAF, PIK3CA, and TP53 have been associated with better outcome (2, 34, 35). Conversely, patients with wild-type APC showed adverse survival (35). Recent studies have highlighted the intertwined relationship of MSI and somatic mutations status (35–39). MSI/BRAFwild-type and MSS/BRAFmutant patients exhibited the most favorable and unfavorable outcome, respectively (36–39). Remarkably, MSI/BRAFmutant showed superior prognosis compared with MSS/BRAFwild-type, suggesting that microsatellite status prevails over BRAF mutation in steering patients survival in these subgroups (36–39). Mutations in TP53, KRAS, and APC have been more frequently observed in microsatellite-stable (MSS) patients with the most adverse outcome being observed in the MSS triple-mutant subpopulation (35). Remarkable advances in “-omics” technologies have brought about a shift in prognostic marker discovery, moving from conventional clinicopathologic risk factors toward their integration with molecular characteristics. Multigene-based classification algorithms have been shown to aid in stratifying patient outcome, but these classifiers so far provide only small improvements in stratification compared with the use of clinical markers alone (40). Reviewing and re-assessing promising multigene and multiprotein panels using systems biological approaches may allow us to further improve the prognostic value of such signatures.

We demonstrated that patient-specific predictions by the APOPTO-CELL signature were highly robust against noise in measurements and parameterization. This robustness emanates from the multiprotein interplay during the apoptosis execution phase, which includes several positive feedback loops. If sufficiently stimulated, these bring about an irreversible cell death decision with a pronounced binary/switch-like character (9, 10, 18). As our discovery cohort was composed of patients recruited at 3 different centers in three European countries, this high robustness may be fundamental for successful model-based patient stratification. Our approach of apoptosis modeling was successful in the expansion and validation cohort, which made use of FF materials and transcript abundance in colorectal cancer tumor samples collected at various centers across the France and United States, respectively.

We introduced the APOPTO-CELL-PC3 signature which capitalizes on both the prognostic information embedded in the apoptosome-dependent cell death pathways modeled by APOPTO-CELL and Procaspase-3 paracrine signaling to further improve prognostic accuracy. Procaspase-3 exerts important roles outside of MOMP-dependent apoptosis execution including mediating tissue regeneration and immune responses (23–29). Upon Caspase-3 activation, apoptotic cells release growth signals such as prostaglandin E2 that ultimately induce tissue regeneration and wound healing in the neighbor cells via the “Phoenix Rising” pathway (25). Furthermore, antitumor immune responses trigger apoptosis cascades through Procaspase-8 activation or granzyme B activation, that can drive Procaspase-3 activation independent of the mitochondrial pathway (23–25, 29).

There is extensive genomic, epigenomic, and molecular interpatient heterogeneity in colorectal cancer (41). A key step toward personalized precision oncology is to develop panels of combinatorial biomarkers, as opposed to single biomarkers, that can better capture disease complexity and provide individualized predictions on cancer progression and treatment response. To develop optimal stratification tools, it is paramount to integrate clinical patient-specific characteristics with molecular phenotypes or, as in our case, patient-specific systems-level characteristics. By applying RF machine learning, we investigated how the APOPTO-CELL-PC3 signature could be combined with conventional clinical variables currently used for colorectal cancer prognosis. The RF approach identified APOPTO-CELL-PC3 as the most impactful predictor, and a reduced RF classifier allowed us to optimally stratify patients into risk groups from a limited number of variables. Reducing the number of clinicopathologic variables and molecular markers required for optimal and individualized prognostication minimizes the economic burden of otherwise possibly excessively complex assays and data acquisition. We generated a valuable visualization of how the predictors contribute to shifting recurrence risk, which could serve as a nomogram to aid medical practitioners in planning disease management.

Extensive RNA profiling work (16, 42–45) recently resulted in the definition of 4 major subtypes for colorectal cancer (CMS1–4; ref. 30). Compared with patients with CMS1–3, CMS4 patients had higher risks of relapse and mortality. Analysis of the interplay between apoptosis competency and CMS revealed a potential differential role of apoptosis signaling in CMS1–3 compared with CMS4, with apoptosis competency associated with good prognosis in CMS1–3. The salient features of the CMS4 subtype (“mesenchymal”) include CIN/MSI alterations, NOTCH3 overexpression, TGFβ, and VEGF activation (30). TGFβ and VEGF signaling has been reported to suppress Caspase-3-dependent apoptosis execution in various cell types (46, 47). Conversely, caspases have been shown to be enriched in the CMS1 (“immune”) subtype.

Previous proof-of-concept studies on apoptosis modeling in colorectal cancer have been limited to the use of FF materials for the generation of tissue protein extracts (14). Our study shows that formalin-fixed, paraffin-embedded (FFPE)-derived protein extracts are suitable for calculating apoptosis competency, representing a key advancement toward aligning with routine surgical and pathologic workflows. It also supports independent retrospective validation in larger biobanked collections of stage III colorectal cancer cohorts. While RPPA-based protein quantification pipelines may not be amenable for integration into standard clinical workflows, the development of calibrated protein quantification assays, such as multiplex ELISAs or novel quantitative digital histopathology techniques, may address current limitations in routine quantitative clinical proteomics (48).

Recent research has pinpointed the pivotal role played by the tumor microenvironment in regulating response to chemotherapeutics (49). Factoring in relative abundance of key apoptotic proteins in both the tumor and the surrounding tissue (stroma, immune cells, etc.) will further fine-tune risk scores. Emerging technologies, such as multiplexed fluorescence microscopy (MxIF; ref. 50) now allow simultaneous co-staining of multiple proteins and reliable quantitative measurements within different compartments which will be instrumental in future studies to tackle these challenges.

P.G. Johnston is a consultant/advisory board member for Chugai. P. Laurent-Puig is a consultant/advisory board member for Amgen, Boerhinger-Ingelheim, Integragen, Merck Serono, Pfizer, Roche, and Sanofi. No potential conflicts of interest were disclosed by the other authors.

Conception and design: M. Salvucci, M. Lawler, P.G. Johnston, D.A. McNamara, P. Laurent-Puig, M. Rehm, J.H.M. Prehn

Development of methodology: M. Salvucci, M.L. Würstle, P. Laurent-Puig, M. Rehm, J.H.M. Prehn

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): M. Salvucci, M.L. Würstle, C. Morgan, M. Cremona, O. Bacon, R. O'Bryne, L. Flanagan, S. Dasgupta, N. Rice, C. Pilati, S. Toomey, R. Wilson, S. Camilleri-Broët, M. Salto-Tellez, D.A. McNamara, E.W. Kay, P. Laurent-Puig, D.B. Longley

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): M. Salvucci, M. Cremona, A.J. Resler, N. Rice, E. Zink, L.M. Schöller, P.G. Johnston, E.W. Kay, P. Laurent-Puig, B.T. Hennessy, M. Rehm, J.H.M. Prehn

Writing, review, and/or revision of the manuscript: M. Salvucci, M.L. Würstle, A.U. Lindner, A.J. Resler, M. Lawler, P.G. Johnston, R. Wilson, M. Salto-Tellez, D.A. McNamara, E.W. Kay, P. Laurent-Puig, S. Van Schaeybroeck, B.T. Hennessy, D.B. Longley, M. Rehm, J.H.M. Prehn

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): M. Salvucci, M.L. Würstle, S. Curry, O. Bacon, Á.C. Murphy, L. Flanagan, S. Dasgupta, N. Rice, D.A. McNamara, S. Van Schaeybroeck

Study supervision: M. Rehm, J.H.M. Prehn

The authors acknowledge support for their work by the European Union Framework Programme 7 (FP7 APO-DECIDE) under contract no. 306021. J.H.M. Prehn is supported by a Science Foundation Ireland Investigator Award (13/IA/1881) and by the Irish Cancer Society BreastPredict Collaborative Research Centre (CCRC13GAL). J.H.M. Prehn, M. Rehm, and D.B. Longley are supported by a Science Foundation Ireland/Department of Enterprise and Learning Partnership Award (14/IA/2582). E. Zink was supported by a Health Research Board Translational Research Supplementary Award (TRA/2007/26). L.M. Schöller was supported by the European Community Action Scheme for the Mobility of University Students (ERASMUS).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Jemal
A
,
Bray
F
,
Center
MM
,
Ferlay
J
,
Ward
E
,
Forman
D
. 
Global cancer statistics
.
CA Cancer J Clin
2011
;
61
:
69
90
.
2.
Dienstmann
R
,
Salazar
R
,
Tabernero
J
. 
Personalizing colon cancer adjuvant therapy: selecting optimal treatments for individual patients
.
J Clin Oncol
2015
;
33
:
1787
96
.
3.
Longley
DB
,
Harkin
DP
,
Johnston
PG
. 
5-fluorouracil: mechanisms of action and clinical strategies
.
Nat Rev Cancer
2003
;
3
:
330
8
.
4.
Ewald
B
,
Sampath
D
,
Plunkett
W
. 
Nucleoside analogs: molecular mechanisms signaling cell death
.
Oncogene
2008
;
27
:
6522
37
.
5.
Hanahan
D
,
Weinberg
RA
. 
Hallmarks of cancer: the next generation
.
Cell
2011
;
144
:
646
74
.
6.
Longley
DB
,
Johnston
PG
. 
Molecular mechanisms of drug resistance
.
J Pathol
2005
;
205
:
275
92
.
7.
Taylor
RC
,
Cullen
SP
,
Martin
SJ
. 
Apoptosis: controlled demolition at the cellular level
.
Nat Rev Mol Cell Biol
2008
;
9
:
231
41
.
8.
Huber
HJ
,
Duessmann
H
,
Wenus
J
,
Kilbride
SM
,
Prehn
JHM
. 
Mathematical modelling of the mitochondrial apoptosis pathway
.
Biochim Biophys Acta
2011
;
1813
:
608
15
.
9.
Spencer
SL
,
Sorger
PK
. 
Measuring and modeling apoptosis in single cells
.
Cell
2011
;
144
:
926
39
.
10.
Rehm
M
,
Huber
HJ
,
Dussmann
H
,
Prehn
JHM
. 
Systems analysis of effector caspase activation and its control by X-linked inhibitor of apoptosis protein
.
EMBO J
2006
;
25
:
4338
49
.
11.
Huber
HJ
,
Rehm
M
,
Plchut
M
,
Düssmann
H
,
Prehn
JHM
. 
APOPTO-CELL–a simulation tool and interactive database for analyzing cellular susceptibility to apoptosis
.
Bioinformatics
2007
;
23
:
648
50
.
12.
Schmid
J
,
Dussmann
H
,
Boukes
GJ
,
Flanagan
L
,
Lindner
AU
,
O'Connor
CL
, et al
Systems analysis of cancer cell heterogeneity in caspase-dependent apoptosis subsequent to mitochondrial outer membrane permeabilization
.
J Biol Chem
2012
;
287
:
41546
59
.
13.
Murphy
ÁC
,
Weyhenmeyer
B
,
Schmid
J
,
Kilbride
SM
,
Rehm
M
,
Huber
HJ
, et al
Activation of executioner caspases is a predictor of progression-free survival in glioblastoma patients: a systems medicine approach
.
Cell Death Dis
2013
;
4
:
e629
.
14.
Hector
S
,
Rehm
M
,
Schmid
J
,
Kehoe
J
,
McCawley
N
,
Dicker
P
, et al
Clinical application of a systems model of apoptosis execution for the prediction of colorectal cancer therapy responses and personalisation of therapy
.
Gut
2012
;
61
:
725
33
.
15.
Flanagan
L
,
Lindner
AU
,
de Chaumont
C
,
Kehoe
J
,
Fay
J
,
Bacon
O
, et al
BCL2 protein signalling determines acute responses to neoadjuvant chemoradiotherapy in rectal cancer
.
J Mol Med
2014
;
93
:
315
26
.
16.
Marisa
L
,
de Reyniès
A
,
Duval
A
,
Selves
J
,
Gaub
MP
,
Vescovo
L
, et al
Gene expression classification of colon cancer into molecular subtypes: characterization, validation, and prognostic value
.
PLoS Med
2013
;
10
:
e1001453
.
17.
Breiman
L
. 
Random Forests
.
Mach Learn
2001
;
45
:
5
32
.
18.
Rehm
M
,
Dussmann
H
,
Janicke
RU
,
Tavare
JM
,
Kogel
D
,
Prehn
JHM
. 
Single-cell fluorescence resonance energy transfer analysis demonstrates that caspase activation during apoptosis is a rapid process. Role of caspase-3
.
J Biol Chem
2002
;
277
:
24506
14
.
19.
Flusberg
DA
,
Sorger
PK
. 
Surviving apoptosis: life–death signaling in single cells
.
Trends Cell Biol
2015
;
25
:
446
58
.
20.
Kolata
G
. 
The Art of Learning from Experience: Statistician Bradley Efron tells what his field is about and how a new method, the bootstrap, exploits the power of large-scale computing
.
Science
1984
;
225
:
156
8
.
21.
Aldridge
BB
,
Gaudet
S
,
Lauffenburger
DA
,
Sorger
PK
. 
Lyapunov exponents and phase diagrams reveal multi-factorial control over TRAIL-induced apoptosis
.
Mol Syst Biol
2011
;
7
:
553
.
22.
Sigal
A
,
Milo
R
,
Cohen
A
,
Geva-Zatorsky
N
,
Klein
Y
,
Liron
Y
, et al
Variability and memory of protein levels in human cells
.
Nature
2006
;
444
:
643
6
.
23.
Huang
Q
,
Li
F
,
Liu
X
,
Li
W
,
Shi
W
,
Liu
F-F
, et al
Caspase 3-mediated stimulation of tumor cell repopulation during cancer radiotherapy
.
Nat Med
2011
;
17
:
860
6
.
24.
Kurtova
AV
,
Xiao
J
,
Mo
Q
,
Pazhanisamy
S
,
Krasnow
R
,
Lerner
SP
, et al
Blocking PGE2-induced tumour repopulation abrogates bladder cancer chemoresistance
.
Nature
2015
;
517
:
209
13
.
25.
Li
F
,
Huang
Q
,
Chen
J
,
Peng
Y
,
Roop
DR
,
Bedford
JS
, et al
Apoptotic cells activate the “phoenix rising” pathway to promote wound healing and tissue regeneration
.
Sci Signal
2010
;
3
:
ra13
.
26.
Lin
F
,
Ghislat
G
,
Luo
S
,
Renna
M
,
Siddiqi
F
,
Rubinsztein
DC
. 
XIAP and cIAP1 amplifications induce Beclin 1-dependent autophagy through NFκB activation
.
Hum Mol Genet
2015
;
24
:
2899
913
.
27.
Lu
M
,
Lin
S-C
,
Huang
Y
,
Kang
YJ
,
Rich
R
,
Lo
Y-C
, et al
XIAP induces NF-kappaB activation via the BIR1/TAB1 interaction and BIR1 dimerization
.
Mol Cell
2007
;
26
:
689
702
.
28.
Murray
TVA
,
McMahon
JM
,
Howley
BA
,
Stanley
A
,
Ritter
T
,
Mohr
A
, et al
A non-apoptotic role for caspase-9 in muscle differentiation
.
J Cell Sci
2008
;
121
:
3786
93
.
29.
Shalini
S
,
Dorstyn
L
,
Dawar
S
,
Kumar
S
. 
Old, new and emerging functions of caspases
.
Cell Death Differ
2014
;
22
:
526
39
.
30.
Guinney
J
,
Dienstmann
R
,
Wang
X
,
de Reyniès
A
,
Schlicker
A
,
Soneson
C
, et al
The consensus molecular subtypes of colorectal cancer
.
Nat Med
2015
;
21
:
1350
6
.
31.
Würstle
ML
,
Zink
E
,
Prehn
JHM
,
Rehm
M
. 
From computational modelling of the intrinsic apoptosis pathway to a systems-based analysis of chemotherapy resistance: achievements, perspectives and challenges in systems medicine
.
Cell Death Dis
2014
;
5
:
e1258
.
32.
McCarthy
N
. 
Therapeutics: Smac it to them!
Nat Rev Cancer
2008
;
8
:
4
5
.
33.
Bielekova
B
,
Vodovotz
Y
,
An
G
,
Hallenbeck
J
. 
How implementation of systems biology into clinical trials accelerates understanding of diseases
.
Front Neurol
2014
;
5
:
102
.
34.
Kandioler
D
,
Mittlböck
M
,
Kappel
S
,
Puhalla
H
,
Herbst
F
,
Langner
C
, et al
TP53 mutational status and prediction of benefit from adjuvant 5-fluorouracil in stage III colon cancer patients
.
EBioMedicine
2015
;
2
:
823
8
.
35.
Schell
MJ
,
Yang
M
,
Teer
JK
,
Lo
FY
,
Madan
A
,
Coppola
D
, et al
A multigene mutation classification of 468 colorectal cancers reveals a prognostic role for APC
.
Nat Commun
2016
;
7
:
11743
.
36.
Lochhead
P
,
Kuchiba
A
,
Imamura
Y
,
Liao
X
,
Yamauchi
M
,
Nishihara
R
, et al
Microsatellite instability and BRAF mutation testing in colorectal cancer prognostication
.
J Natl Cancer Inst
2013
;
105
:
1151
6
.
37.
Hamilton
SR
. 
BRAF mutation and microsatellite instability status in colonic and rectal carcinoma: context really does matter
.
J Natl Cancer Inst
2013
;
105
:
1075
7
.
38.
Toon
CW
,
Chou
A
,
DeSilva
K
,
Chan
J
,
Patterson
J
,
Clarkson
A
, et al
BRAFV600E immunohistochemistry in conjunction with mismatch repair status predicts survival in patients with colorectal cancer
.
Mod Pathol
2014
;
27
:
644
50
.
39.
Seppälä
TT
,
Böhm
JP
,
Friman
M
,
Lahtinen
L
,
Väyrynen
VMJ
,
Liipo
TKE
, et al
Combination of microsatellite instability and BRAF mutation status for subtyping colorectal cancer
.
Br J Cancer
2015
;
112
:
1966
75
.
40.
Di Narzo
AF
,
Tejpar
S
,
Rossi
S
,
Yan
P
,
Popovici
V
,
Wirapati
P
, et al
Test of four colon cancer risk-scores in formalin fixed paraffin embedded microarray gene expression data
.
J Natl Cancer Inst
2014
;
106
:
dju247
dju247
.
41.
Hanahan
D
,
Weinberg
RA
,
Wang
Y
,
Kaiser
CE
,
Frett
B
,
Li
HY
, et al
The hallmarks of cancer
.
Cell
2014
;
100
:
57
70
.
42.
Sadanandam
A
,
Lyssiotis
CA
,
Homicsko
K
,
Collisson
EA
,
Gibb
WJ
,
Wullschleger
S
, et al
A colorectal cancer classification system that associates cellular phenotype and responses to therapy
.
Nat Med
2013
;
19
:
619
25
.
43.
Roepman
P
,
Schlicker
A
,
Tabernero
J
,
Majewski
I
,
Tian
S
,
Moreno
V
, et al
Colorectal cancer intrinsic subtypes predict chemotherapy benefit, deficient mismatch repair and epithelial-to-mesenchymal transition
.
Int J Cancer
2014
;
134
:
552
62
.
44.
Schlicker
A
,
Beran
G
,
Chresta
CM
,
McWalter
G
,
Pritchard
A
,
Weston
S
, et al
Subtypes of primary colorectal tumors correlate with response to targeted treatment in colorectal cell lines
.
BMC Med Genomics
2012
;
5
:
66
.
45.
Budinska
E
,
Popovici
V
,
Tejpar
S
,
D'Ario
G
,
Lapique
N
,
Sikora
KO
, et al
Gene expression patterns unveil a new level of molecular heterogeneity in colorectal cancer
.
J Pathol
2013
;
231
:
63
76
.
46.
Prehn
JH
,
Bindokas
VP
,
Marcuccilli
CJ
,
Krajewski
S
,
Reed
JC
,
Miller
RJ
. 
Regulation of neuronal Bcl2 protein expression and calcium homeostasis by transforming growth factor type beta confers wide-ranging protection on rat hippocampal neurons
.
Proc Natl Acad Sci U S A
1994
;
91
:
12599
603
.
47.
Healy
ME
,
Bergin
R
,
Mahon
BP
,
English
K
. 
Mesenchymal stromal cells protect against caspase 3-mediated apoptosis of CD19(+) peripheral B cells through contact-dependent upregulation of VEGF
.
Stem Cells Dev
2015
;
24
:
2391
402
.
48.
Brennan
DJ
,
O'Connor
DP
,
Rexhepaj
E
,
Ponten
F
,
Gallagher
WM
. 
Antibody-based proteomics: fast-tracking molecular diagnostics in oncology
.
Nat Rev Cancer
2010
;
10
:
605
17
.
49.
Quail
DF
,
Joyce
JA
. 
Microenvironmental regulation of tumor progression and metastasis
.
Nat Med
2013
;
19
:
1423
37
.
50.
Gerdes
MJ
,
Sevinsky
CJ
,
Sood
A
,
Adak
S
,
Bello
MO
,
Bordwell
A
, et al
Highly multiplexed single-cell analysis of formalin-fixed, paraffin-embedded cancer tissue
.
Proc Natl Acad Sci U S A
2013
;
110
:
11982
7
.

Supplementary data