Purpose: Targeted therapy (TT) provides highly effective cancer treatment for appropriately selected individuals. A major challenge of TT is to select patients who would benefit most.

Experimental Design: The study uses cancer material from 25 patients primarily diagnosed with non–small cell lung cancer (NSCLC). Patient-derived xenografts (PDXs) are treated with cetuximab and erlotinib. Treatment response is measured by tumor shrinkage comparing tumor volume at day 25 (V25) with tumor volume at baseline (V0). Shrinkage below 40% is considered as treatment response: V25/V0 < 0.4 (<40%). Furthermore, RNA-seq data from each tumor sample are used to predict tumor response to either treatment using an in silico molecular signaling map (MSM) approach.

Results: PDX response was 40% (10/25; 95% CI [21.13%, 61.34%]) under cetuximab and 20% (5/25; 95% CI [6.83%, 40.70%]) under erlotinib. MSM predicted response was 48% (12/25; 95% CI [27.8%, 68.7%]) under cetuximab and 40% (10/25; 95% CI [21.13%, 61.34%]) under erlotinib. Agreement between PDX and MSM response prediction is substantial under cetuximab and erlotinib: 84% (21/25, P = 0.001) and 80% (20/25, P = 0.003). A total of 5 from the 25 patients have been treated with cetuximab showing a clinical response identical to both predictions.

Conclusions: For NSCLC patients, this proof-of-concept study shows a considerable agreement in response prediction from MSM and PDX approaches, but MSM saves time and laboratory resources. Our result indicates the potential of MSM-based approach for clinical decision making when selecting cancer TTs. Clin Cancer Res; 22(9); 2167–76. ©2015 AACR.

This article is featured in Highlights of This Issue, p. 2097

Translational Relevance

Targeted therapy can provide highly effective individual treatment alternatives compared with DNA-directed chemotherapy of low therapeutic indices. If the preclinical evaluation of targeted therapies does not follow simple criteria, it heavily relies on the use of animal tumor models: patient-derived xenograft (PDX). Unfortunately, PDX present major disadvantages: long waiting time, failure of tumor transplantation, and extensive laboratory effort. Therefore, alternative clinical decision making is needed. This study compares PDX-based treatment response of targeted therapies (Cetuximab, Erlotinib) with in silico prediction in samples of lung cancer patients. The comparison provides evidence that the in silico response prediction is in high agreement with the corresponding biology based PDX prediction. The in silico prediction allows fast and cheap individual targeted response prediction in parallel for several substances.

Non–small cell lung cancer (NSCLC) is the most common cause of cancer-related death in developed as well as in developing countries causing more than a million deaths each year on a global scale (1, 2). In recent years, molecularly targeted therapy (TT) improved treatment of NSCLC. Targets focus on inhibition or suppression of highly deregulated mitogenic signaling pathways in cancer cells, including EGFR, MAPK, MET, and MYC pathways, and others (3–5). Tyrosine kinase inhibitors (TKI) such as erlotinib, gefitinib, and cetuximab have been approved for clinical TT of NSCLC by the FDA, the European Medicines Agency (EMA), and the China Food and Drug Administration (CFDA). When applied appropriately, TT often demonstrates clinical efficacy associated with improved overall survival and the abatement of symptoms (6–8).

Despite these promising results, one major challenge is to develop effective strategies to select patients with considerable benefits from a specific TT. Studies have focused on potential driver mutations and their association with TT outcome. For instance, mutations in the EGFR gene might be a powerful biomarker for high sensitivity to targeted TKIs (9–11). Although it has been reported that mutated KRAS might contribute to the resistance of TKIs treatment in NSCLC, the prevalence of this mutations is low, about 5% to 10% in Asian and <20% in other countries (12). The fusion between the echinoderm microtubule-associated protein-like 4 (EML4) and the ALK gene, has been reported to be associated with positive clinical outcome of crizotinib treatment (13, 14). Despite these novel discoveries, the prognosis of NSCLC has remained poor, and the 5-year survival is below 20% in most countries (15).

Recently, several studies provide evidence that patient-derived xenograft (PDX) show biologic consistency with the tumor of origin and are phenotypically stable at the histologic and genomic level across multiple generation (16, 17). Moreover, drug-sensitivity pattern of PDXs can recapitulate response in the patients from which they derived (18). Unfortunately, the timeframe to develop these so-called Avatar models (PDXs) is usually long compared with the clinical decision timeframe for a patient. Furthermore, because of the technical limitations, normally in 30% of patients, the generation of a PDX will fail at the first time. This failure rate increases the expense of the PDX approach significantly. There is an ongoing discussion whether PDX is appropriate for clinical decision making (19–21).

Can the response prediction by cheap in silico strategies compete with that of the PDX? Therefore, we compare response prediction of a computational approach with that of the PDX in a small cohort of NSCLC patients. Our in silico strategy combines formal knowledge of cancer-related cellular processes (represented by a molecular signaling map, MSM) with individual patient data in a Flux-comparative-analysis (FCA) and focuses on the change of signal flux within the underlying molecular cellular system by facing external perturbation, including drug treatment. The change of signal flux in a simulated cellular system might reveal the treatment response of the corresponding individuals (22). Therefore, we reasoned that the FCA applied to our MSM might be able to mimic treatment response of the PDX. We use the next-generation RNA seq as specific information of an individual tumor sample to initialize our algorithm (23). The MSM contains a global cellular signaling network and quantifies different hallmarks of tumorigenesis. It also allows modeling the intervention of a TT and calculation of change of the signal flux between a treatment state and a control state, which is the basic strategy of FCA for enabling the individual response prediction to the TT. So far, the MSM has been applied to investigate the therapeutic responses of AML and colon cancer patients under TT. This is the first study where the algorithm is adapted to NSCLC.

Study design

This study consists of two arms (Fig. 1). The first arm involves the PDX, in which individual tumor samples are implanted. Two xenografts are produced for each patient, one to be treated with erlotinib, the other to be treated with cetuximab. The response measurement consists in the comparison of tumor volume at day 25 (V25) with tumor volume at baseline (V0). A tumor responds to a treatment if it's volume shrinkage below 40%: V25/V0 < 0.4 (<40%).

Figure 1.

The concept of comparison between PDX and MSM-based computational approach. Both approaches investigate the individual responses of erlotinib and cetuximab. The PDX requires 25 days to achieve response results, whereas the simulation of the MSM needs approximately 1 hour.

Figure 1.

The concept of comparison between PDX and MSM-based computational approach. Both approaches investigate the individual responses of erlotinib and cetuximab. The PDX requires 25 days to achieve response results, whereas the simulation of the MSM needs approximately 1 hour.

Close modal

The second arm involves NGS-based RNA-seq and computational treatment simulation. Gene-expression profiles (GEP) are generated from tumor samples of each lung cancer patient, which is the input data to the MSM. Response is the relative change of a score quantifying cancer proliferation in the FCA (22). To this end, the proliferation score is calculated from the MSM without implementing the targeted treatment (control state, PROcontrol) as well as from the MSM with implementing the targeted treatment (treatment state, PROtreat). The quantified response prediction is S = log2(PROtreat/PROcontrol). A negative value predicts an individual response to TT, zero or a positive value predicts a TT failure. Goal of the study is to compare responses as observed in the PDX with response as predicted by the MSM-based approach.

Patient samples and transcriptomic data

Surgical biopsies of 25 patients were obtained from primary diagnosed, early-stage large cell lung cancer (NSCLC) patients between January 2010 and March 2013 from the Yu Huang Hospital (Yu Huang, Zhe Jiang, PR China). Written informed consent was obtained from each patient. The PDX model was created to decide whether cetuximab or erlotinib would be the appropriate TT. This study was approved by the medical ethics committee of the Yu Huang hospital.

All tumor samples were put into medium under the controlled sterile condition immediately after surgical resection. For each sample, high-throughput dataset was generated by using an Illumina HiSeq (Illumina). The tool TopHat (version 2.0.6) was applied to generate reads from the raw RNAseq data with h19 genome. The quality control assessment was conducted by using the tool RSeQC (version 2.6.1). Gene annotations were obtained from the Illumina and gene counts were generated by applying the tool HTSeq (version 0.6.1). The reads per kilobase per million mapped reads (RPKM) was applied to measure the mRNA abundance. To reduce different possible potential bias factors (such as GC content, gene size, and others) of this type of mRNA measurement, the conditional quantile normalization with the cqn Bioconductor package (24) was performed.

Patient-derived xenograft studies and treatment

Nine- to 10-week-old SCID and nude (nu/nu) mice (Vital River) were used for receiving surgical tumor samples. The gender of the mice was chosen according to the gender of donors. All animals were housed in pathogen-free facilities with abundant food and water under the guidance approved by the Institutional Animal Care and Use Committee (IACUC). PDXs were established by transplanting fresh tumor tissues on mice as described in the study (18). After stabilization of tumor-implanted mice, 25 mice were treated with cetuximab, which was administrated every 3 day for five total injections at the dose levels range from 1 mg per injection to 0.03 mg per injection. Another 25 mice were administrated with erlotinib every 3 day for five total injections at the dose levels ranging from 15 mg per injection to 5 mg per injection. Tumor volume was recorded twice per week and calculated with L × W2 × π/6. Antitumor activity was defined on the basis of RECIST criteria (25). Tumor shrinkage (TS) ≤ 40% on the day 25 after start of treatment was considered as positive response: V25/V0 < 0.4.

The human molecular signaling map

In the previous study (23), we describe the construction of a large-scale MSM, which consists of 58 cancer-relevant signaling pathways, including different feedback-loops and miRNA regulations. To be able to reflect different aspects of human tumorigenesis, the MSM also incorporates 10 cancer hallmarks (26, 27). The basic modeling pattern in the MSM is that each gene participates in one or more transcription reactions to generate one or more mRNAs; each mRNA participates in one translation reaction to generate a corresponding protein. Each gene, mRNA and protein in the MSM are labeled with their corresponding unique ID, including Ensemble-ID for gene and mRNA; uniProt-ID for protein. Individual patient gene-expression data are used to initialize the MSM model: Each gene in the model is set to the values from gene-expression data and miRNA expression data as given by the associated IDs.

Modeling of erlotinib and cetuximab treatment

Cetuximab (Erbitux) is a chimeric human-murine monoclonal IgG1 antibody, which is designed to block EGFR-mediated signaling pathways through binding to the extracellular domain of the EGFR. Erlotinib (Tarceva) is a quinazolinamine, which binds in a reversible manner to the adenosine triphosphate (ATP) binding site of receptors, including EGFR, ERBB, and others. Both drugs have been approved by the FDA and are frequently applied drugs for TT for treating lung cancer (28, 29). From a molecular perspective, the drug effect of cetuximab is a single function inhibition at the extracellular domain 3 of EGFR, which is 10-fold greater affinity (0.087 nmol/L) than natural ligand, whereas the drug effect of erlotinib consists of inhibitions of multiple tyrosine kinases with different binding affinities that includes EGFR (0.67 nmol/L), ERBB4 (230 nmol/L), LYN (530 nmol/L), and SRC (700 nmol/L) (30). The mass action law is applied to model the inhibition effect of both drugs in the MSM:

where [Target:Drug] = [Target] × [Drug]/BA; [x]: concentration of component x; BA: binding affinity (Fig. 2A).

Figure 2.

A, modeling of the molecular inhibition effect of cetuximab and erlotinib due to high binding specificity of cetuximab on EGFR's domain, cetuximab is implemented as a strong single inhibition, whereas erlotinib's molecular inhibition effect consists of four inhibitions targeting EGFR, ERBB4, LYN, and SRC. Lig: Ligand; Cet: Cetuximab; Erl: Erlotinib; B, Petri net's Conversion of MSM. The compositions of MSM could be divided into two parts: bio-objects (B) and reactions (R), which enables its easy conversion to Petri net.

Figure 2.

A, modeling of the molecular inhibition effect of cetuximab and erlotinib due to high binding specificity of cetuximab on EGFR's domain, cetuximab is implemented as a strong single inhibition, whereas erlotinib's molecular inhibition effect consists of four inhibitions targeting EGFR, ERBB4, LYN, and SRC. Lig: Ligand; Cet: Cetuximab; Erl: Erlotinib; B, Petri net's Conversion of MSM. The compositions of MSM could be divided into two parts: bio-objects (B) and reactions (R), which enables its easy conversion to Petri net.

Close modal

Brief outline of procedure of FCA for drug treatment

FCA is applied to investigate whether a drug could cause a significant flux change in a model system between two different states: control (C) and treatment (T). The MSM was reported in the previous study (23) and is the model system for the FCA. All MSM components are either reactions (including phosphorylation, transcription, and others) or bio-objects (including gene, protein, and others). Thus, the MSM is converted to a B-tree (31) with reaction-nodes (R) and bio-object-nodes (B; Fig. 2B). Each child node is a data-array, whose elements are reactions (R) and bio-objects (B) as defined in the MSM. After the B-tree conversion, the gene components in the MSM can be set to the values of transcriptome data as given by the Ensemble-ID. After initialization, the B tree is converted into a Petri Net (P) with the definition: P = (R, B), where the R contains all transitions (reactions) and the B contains all places (bio-objects; Fig. 2B). The simulation of P is performed by iterating transitions in R for firing evaluation X, until the P reaches a steady. The simulation for each treatment takes 50 minutes within a computer of duo CPU, each with 2.66 GHz, 4Gb memory and 400 MT/s.

Statistical analyses

Descriptive statistics are used to present differences between groups. Scores are compared by using boxplots. Differences are assessed by the non-parametric Mann-Whitney test. The discriminative power of scores between two groups is analyzed by ROC curves and the AUC summary measure (32). The correlation between two scores is studied by Spearman correlation.

Clinical characteristics of lung cancer patients

Twenty-five surgical biopsies histopathologically diagnosed as early stage of NSCLC from Yu Huang hospital from January 2010 and November 2013 were collected for this study together with the patient's demographic and clinical information, including age, gender, stage, histology, and others. The mean age (SD) of patients was 62.08 ± 11.87. No patient received any anticancer treatment before diagnosis. The main smoking history of patients was non-smoking (20/25, 80%). The majority of NSCLC cases were in the stage I (19/25, 76%; See Table 1).

Table 1.

Clinical Information of recruited 25 NSCLC patients

Patient_IDAgeSexStageHistologySmokingPrior treatmentDiagnosed
14957 71 LCC Yes No 2011 Nov 17 
14963 43 LCC No No 2012 Apr 06 
14965 36 LCC No No 2012 Jul 02 
14953 67 LCC No No 2011 Sep 12 
14954 45 LCC No No 2011 Jan 14 
14966 61 LCC Yes No 2010 Jul 19 
14968 74 LCC No No 2010 Sep 21 
14971 77 LCC No No 2013 Nov 19 
14976 57 LCC Yes No 2013 Oct 05 
14975 56 LCC No No 2013 Dec 01 
14956 73 LCC No No 2010 Oct 23 
14960 71 LCC No No 2011 Jan 27 
14955 62 LCC No No 2010 Mar 02 
14961 71 LCC Yes No 2013 Aug 02 
14977 76 LCC No No 2012.May.19 
14962 50 LCC No No 2012 Jul 16 
14964 64 LCC No No 2012 May 28 
14967 43 LCC No No 2013 May 02 
14969 77 LCC No No 2013 Oct 27 
14970 73 LCC No No 2010 Oct 25 
14972 58 LCC Yes No 2010 Jan 15 
14973 73 LCC No No 2011 Jul 24 
14974 57 LCC No No 2011 Feb 26 
14958 58 LCC No No 2012 Mar 16 
14959 59 LCC No No 2012 May 17 
Patient_IDAgeSexStageHistologySmokingPrior treatmentDiagnosed
14957 71 LCC Yes No 2011 Nov 17 
14963 43 LCC No No 2012 Apr 06 
14965 36 LCC No No 2012 Jul 02 
14953 67 LCC No No 2011 Sep 12 
14954 45 LCC No No 2011 Jan 14 
14966 61 LCC Yes No 2010 Jul 19 
14968 74 LCC No No 2010 Sep 21 
14971 77 LCC No No 2013 Nov 19 
14976 57 LCC Yes No 2013 Oct 05 
14975 56 LCC No No 2013 Dec 01 
14956 73 LCC No No 2010 Oct 23 
14960 71 LCC No No 2011 Jan 27 
14955 62 LCC No No 2010 Mar 02 
14961 71 LCC Yes No 2013 Aug 02 
14977 76 LCC No No 2012.May.19 
14962 50 LCC No No 2012 Jul 16 
14964 64 LCC No No 2012 May 28 
14967 43 LCC No No 2013 May 02 
14969 77 LCC No No 2013 Oct 27 
14970 73 LCC No No 2010 Oct 25 
14972 58 LCC Yes No 2010 Jan 15 
14973 73 LCC No No 2011 Jul 24 
14974 57 LCC No No 2011 Feb 26 
14958 58 LCC No No 2012 Mar 16 
14959 59 LCC No No 2012 May 17 

Cetuximab and erlotinib in silico treatment prediction

Because of high efficacy of TKIs in NSCLC (33, 34), cetuximab and erlotinib were chosen to investigate prediction of treatment response. The individual (MSM and PDX based) predicted responses scores under both treatments are listed in the Table 2 (two left columns). For the MSM-based prediction, negative values indicate a positive response.

Table 2.

Response data from PDX and MSM-based prediction

MSM-based treatment predictionPDX study tumor/control (%)
Patient_IDCetuximabErlotinibCetuximabErlotinib
14957 1.01 0.86 73 79 
14963 −0.06 −0.12 51 25 
14955 −0.43 1.35 33 43 
14965 0.79 0.35 24 77 
14960 0.47 0.14 42 93 
14961 −0.82 −0.08 30 53 
14953 −2.4 −2.9 14 23 
14954 −0.7 −1.1 18 30 
14977 −0.77 −0.89 47 78 
14962 0.09 0.82 87 92 
14964 −0.23 −0.12 75 
14967 0.37 0.85 68 98 
14969 0.15 0.61 53 79 
14970 1.06 0.63 48 66 
14966 0.67 0.48 72 67 
14968 0.77 0.85 44 57 
14971 −0.82 −0.46 61 63 
14976 1.05 0.75 88 52 
14975 1.28 1.12 76 67 
14956 0.65 0.44 85 96 
14958 −1.15 −0.72 32 
14959 −0.84 −0.36 27 
14972 −0.22 0.75 57 
14973 −0.61 −0.78 23 68 
14974 0.79 1.13 63 68 
MSM-based treatment predictionPDX study tumor/control (%)
Patient_IDCetuximabErlotinibCetuximabErlotinib
14957 1.01 0.86 73 79 
14963 −0.06 −0.12 51 25 
14955 −0.43 1.35 33 43 
14965 0.79 0.35 24 77 
14960 0.47 0.14 42 93 
14961 −0.82 −0.08 30 53 
14953 −2.4 −2.9 14 23 
14954 −0.7 −1.1 18 30 
14977 −0.77 −0.89 47 78 
14962 0.09 0.82 87 92 
14964 −0.23 −0.12 75 
14967 0.37 0.85 68 98 
14969 0.15 0.61 53 79 
14970 1.06 0.63 48 66 
14966 0.67 0.48 72 67 
14968 0.77 0.85 44 57 
14971 −0.82 −0.46 61 63 
14976 1.05 0.75 88 52 
14975 1.28 1.12 76 67 
14956 0.65 0.44 85 96 
14958 −1.15 −0.72 32 
14959 −0.84 −0.36 27 
14972 −0.22 0.75 57 
14973 −0.61 −0.78 23 68 
14974 0.79 1.13 63 68 

Under cetuximab the minimal score (maximal score) was −2.400 (1.280) with mean (median, SD) of 0.004 (0.090 and 0.890). Under erlotinib the minimal (maximal) score was −2.900 (1.350) with mean (median, SD) of 0.144 (0.440 and 0.94). The paired results in both MSM models are presented in Fig. 3A. The reaction under cetuximab cannot be statistically distinguished from the reaction under erlotinib (Wilcoxon test P value, 0.3746), which is demonstrated by a tight scattering of the combined measurements around the diagonal (line of equality). This figure also shows the predicted response rate under cetuximab of 48% (12/25; 95% CI [27.8%, 68.7%]) and under erlotinib of 40% (10/25; 95% CI [21.13%, 61.34%]).

Figure 3.

A, paired results of predicted responses of cetuximab and erlotinib through the MSM-based computational approach predicted response rate under cetuximab (erlotinib) is 48% (40%); B, paired results of PDX responses of cetuximab and erlotinib response rate under cetuximab (erlotinib) is 40% (20%); C, correlation of PDX responses and MSM-based responses under cetuximab overall agreement is 84%, correlation (Spearman: −0.66, P < 0.001; D, correlation of PDX response and MSM-based response under erlotinib overall agreement is 80%, correlation (Spearman: −0.35, P = 0.1115); E and F, ROC analysis of similarity between PDX responses and MSM-based responses under cetuximab and erlotinib;

Figure 3.

A, paired results of predicted responses of cetuximab and erlotinib through the MSM-based computational approach predicted response rate under cetuximab (erlotinib) is 48% (40%); B, paired results of PDX responses of cetuximab and erlotinib response rate under cetuximab (erlotinib) is 40% (20%); C, correlation of PDX responses and MSM-based responses under cetuximab overall agreement is 84%, correlation (Spearman: −0.66, P < 0.001; D, correlation of PDX response and MSM-based response under erlotinib overall agreement is 80%, correlation (Spearman: −0.35, P = 0.1115); E and F, ROC analysis of similarity between PDX responses and MSM-based responses under cetuximab and erlotinib;

Close modal

The FCA results show that within the responders of cetuximab treatment, signal flux of key components such as FLT1 (>30%), CTNNB1 (>31%), JAK2/3 (>43%), MYD88 (>42%), MYC (>45%), and PDGF (>30%) from diverse signaling pathways, including EGFR, VEGF, WNT, JAK-STAT, TLR, MAPK, and PI3K–AKT, PDGF signaling pathways are clearly downregulated, which indicates the downregulation of proliferation, growth, and angiogenesis in the responders (Supplementary Table S1). Interestingly, many components from the cell-cycle pathway are also downregulated. This reduces the replicative potential of cancers (27). Furthermore, signal flux of several components, including TSC1/2 (>23%), RPTOR (69%), AMPK (50%) from the AMPK pathway, are highly downregulated, which leads to restrict the high energy demand for cancer development. It is noteworthy that signal flux of several important transcription factors, including CREB (>24%), ELK4 (30%), HIF2 (47%), JNKs (33%), SRF (30%), are also downregulated (Supplementary Table S1).

For the responders of erlotinib treatment, the FCA results show that signal flux from many key components in pathways, including EGFR, PI3K/AKT, MAPK, WNT, VEGF, cell cycle, PDGF, and other signaling pathways, are downregulated (Supplementary Table S2). The downregulation of EGFR, PI3K/AKT, MAPK, JAK–STAT, cell cycle, AMPK signaling pathways is a common phenomenon for the predicted responders of both TKI treatments, which retains proliferation and growth of cancer cells. This conclusion is partially in an agreement with several independent studies that discovered crucial signaling pathways for tumorigenesis: The prosurvival arm of pathways comprising PI3K–AKT–mTOR and the proliferative arm consisting of the RAS–RAF–MEK–ERK pathways (35).

Cetuximab and erlotinib treatment on PDXs and comparison with predicted responses

For each of these 25 patient two avatar models of his/her cancer are prepared and used for treatment response testing. The biologic response on the treatment was measured by a change score for the tumor volume (TS, tumor shrinkage: V25/V0): The percentage of volume between start and day 25 of treatment. Shrinkage below 40% was considered as response (Table 2, two right columns). Under cetuximab maximal (minimal) tumor shrinkage was 1% (88%) with mean (median, SD) of 44.68% (47.00%, 27.8%). Under erlotinib maximal (minimal) tumor shrinkage was 23% (98%) with mean (median, SD) of 62.64% (67.00%, 22.6%) (Table 2, two right columns). The paired results in the avatar model are presented in Fig. 3B. Tumor shrinkage under cetuximab is significantly more extend as under erlotinib (Wilcoxon test P value: 0.001562), which is demonstrated by a large number of combined measurement below the diagonal (line of equality). This figure shows the response rate under cetuximab of 40% (10/25; 95% CI [21.13%, 61.34%]) and a response rate under erlotinib of 20% (5/25; 95% CI [6.83%, 40.70%]).

Furthermore, we systematically compared the PDX responses with that in silico response. Figure 3C and D show the correspondence between the TS measurement and the in silico response score under cetuximab and erlotinib, respectively. The correlation (Spearman) between both measurements under cetuximab is as expected negative (rho = −0.66, P = 0.0003431). Higher tumor shrinkage implies lower MSM prediction scores. The correlation (Spearman) between both measurements under erlotinib is as well negative but not significantly different from 0 (rho = −0.35, P = 0.1115). Under cetuximab, both approaches result in positive response predictions for 9 patients and for 12 patients in simultaneous negative response prediction. Divergent results exist for 4 patients. The overall agreement between both responses under cetuximab is 84% (21/25; 95% CI [63.92%, 95.46%]). Under erlotinib, both approaches result in positive response predictions for 5 patients and for 15 patients in simultaneous negative response prediction. Divergent results exist for 5 patients. The overall agreement between both response under erlotinib is 80% (20/25; 95% CI [59.30%, 93.17%]). Under both treatments the overall agreement is significantly different from random agreement. Moreover, to investigate how well the MSM score predicts the biologic responses from PDX, the ROC analysis (36) has been performed, which shows that under cetuximab the AUC is 0.86 (95% CI [0.65, 0.96]); under erlotinib the AUC is 0.915 (95% CI [0.78, 0.99]) (Fig. 3E and F).

Clinical outcome

Among the 25 recruited lung patients, 11 left the hospital shortly after diagnosis and could not be followed. Five of the remaining 14 patients chose TT (cetuximab) as part of the adjuvant therapy according to the PDX results (Table 3). The responses of all 5 are in agreement with the individual in silico prediction result.

Table 3.

Clinical outcome of recruited NSCLC patients

Patient_IDAgeSexStageSmokingTreatmentResponse
14957 71 Yes Targeted therapy SD 
14963 43 No Surgery CR 
14965 36 No Surgery CR 
14953 67 No Surgery CR 
14954 45 No NA NA 
14966 61 Yes Chemotherapy PR 
14968 74 No NA NA 
14971 77 No NA NA 
14976 57 Yes Targeted therapy SD 
14975 56 No NA NA 
14956 73 No Targeted therapy SD 
14960 71 No NA NA 
14955 62 No Surgery CR 
14961 71 Yes Targeted therapy CR 
14977 76 No NA NA 
14962 50 No NA NA 
14964 64 No NA NA 
14967 43 No NA NA 
14969 77 No Surgery CR 
14970 73 No Surgery CR 
14972 58 Yes NA NA 
14973 73 No Surgery CR 
14974 57 No Surgery CR 
14958 58 No Targeted therapy CR 
14959 59 No NA NA 
Patient_IDAgeSexStageSmokingTreatmentResponse
14957 71 Yes Targeted therapy SD 
14963 43 No Surgery CR 
14965 36 No Surgery CR 
14953 67 No Surgery CR 
14954 45 No NA NA 
14966 61 Yes Chemotherapy PR 
14968 74 No NA NA 
14971 77 No NA NA 
14976 57 Yes Targeted therapy SD 
14975 56 No NA NA 
14956 73 No Targeted therapy SD 
14960 71 No NA NA 
14955 62 No Surgery CR 
14961 71 Yes Targeted therapy CR 
14977 76 No NA NA 
14962 50 No NA NA 
14964 64 No NA NA 
14967 43 No NA NA 
14969 77 No Surgery CR 
14970 73 No Surgery CR 
14972 58 Yes NA NA 
14973 73 No Surgery CR 
14974 57 No Surgery CR 
14958 58 No Targeted therapy CR 
14959 59 No NA NA 

NOTE: Chemotherapy, carboplatin taxol; targeted therapy, cetuximab.

In the past several years, molecularly TT has been demonstrated its high potential for lung cancer treatment as monotherapy as well as combination therapy. How to identify patients who can derive most benefit from a specific TT is currently the major challenge, which is also one of the obstacles to make TT a standard routine in clinical practice.

To our knowledge, this study is the first study that compares PDX and a computational strategy with regard to the treatment response of TT for NSCLC patients. The use of PDX is one strategy under investigation to identify potential responders. Although the PDX is expensive, has a high labor-intensity and time-consumption, many recent studies have demonstrated its potential utility in the clinical application for different cancer types (37–39). Nevertheless, one major disadvantage is that the average timeframe for generating response data from PDX would not be less than 15 days, in this study, 25 days. Furthermore, recent studies provided compelling evidence about the strong impact of social distress on the tumor growth in PDXs (40, 41). Stress-related signaling cascades such as adenylyl cyclase/cAMP/PKA signaling (42), MAPK–ERK signaling (40), and Hedgehog signaling (41) may confound the treatment effects of interest. There is the potential bias of producing false response signals: The TT may be effective, but social distress may impair this effect.

As alternative, we propose to use a computational approach based on individual molecular patient data, an MSM and the FCA. The MSM-based response prediction suggests that TKI treatments (cetuximab and erlotinib) imply therapeutic effects, if the signal flux within several mitogenic signaling pathways, including EGFR, PI3K–AKT, and MAPK is downregulated. Activities of different mitogenic pathways are critical for the uncontrolled proliferation and invasion of lung cancer cells (3–5). This is seen in the individual simulation and may elicit the cancer-relevant deregulation at molecular level as well as individual treatment response prediction. The proposed computational approach has been explored in several studies (22, 23). The algorithm can be used to analyze data of an individual patient with respect to several targeted therapies. The computing effort using a standard computer is approximately an hour. By comparison, predicting response for several treatments using PDX is restricted by the amount of tumor tissue and is not affordable in most labs.

In this phase I prognostic study (proof of concept), the PDX- and MSM-based response prediction were compared in 25 NSCLC patients and for two targeted therapies (cetuximab, erlotinib). For both substances, the results demonstrated a potentially strong agreement between PDX- and MSM-based prediction [overall response agreement: 84% (cetuximab) and 80% (erlotinib)]. In terms of a, ROC analysis, it became obvious that MSM response scores may be good surrogates for a complicated PDX prediction [AUC: 0.86 (cetuximab) and 0.915 (erlotinib)]. It could not be clarified in this study how much the disagreement between MSM- and PDX-based response prediction (Fig. 3C and D) may be caused by social stress promoted growth of NSCLC in the xenografts. For both treatments, the MSM-based response prediction is always better compared with the PDX-based (cetuximab: MSM 48% vs. PDX 40%; erlotinib: MSM 40% vs. PDX 20%). The striking difference between MSM and PDX prediction for erlotinib may also be caused by modeling effects. So far, we did not check how the infrastructure of the MSM or the algorithms used for the FCA may be biased with respect to different backgrounds regarding cancer biology and implementation of treatment effects, because all mice were housed under the same standard facility condition we would not expect a noticeable stress effect. However, the stress-related signaling cascades such as adenylyl cyclase/cAMP/PKA signaling (42), MAPK/ERK signaling (40), and Hedgehog signaling (41) are well implemented in the MSM model. To settle this question, we started a project in which patient, PDX, and model-based response can be compared and quality differences between MSM-based and PDX-based response prediction can be quantified. These results will be available within next 2 years. In summary, these results of our study emphasize the potential of the computational molecular modeling for TT and an effective alternative for expensive, labor-intensive and time-consuming PDX studies.

This study is based on a homogeneous well-specified sample of patients. Regarding the functional principle of the computational approach, it is possible to extend the treatment simulation for predicting the therapeutic effect of multiple drugs, which might function as an important part for a drug discovery process. The limitations of our study are: the small sample size of 25 patients does not allow reaching substantial conclusions. It was possible to compare both strategies, but we could not validate the prediction with final clinical result of every patient. Only 5 patients were treated with TT of cetuximab. Moreover, the current limitation of the MSM-based computational approach is that MSM is a large-scale signaling network map, and can therefore only predict the responses from treatment of signaling agents such as cetuximab, imatinib, lapatinib, temsirolimus, and others. Although the FCA, which is based on Petri net, is not so much dependent on kinetic parameters as the standard ordinary differential equation system (ODE) does, the more detailed kinetic parameters are applied, the more accurate could be the result of FCA. Therefore, information from kinetic databases such as Brenda (43) will be used to improve the kinetic parameters of MSM in future studies.

In conclusion, we see the MSM-based computational approach as a potentially effective prediction strategy to determine individual TT for NSCLC patients. Our proof-of-concept study (phase I prognostic study) demonstrates the potential substantial agreement between a PDX- and an MSM-based response prediction.

U.R. Mansmann reports receiving speakers bureau honoraria from Baxter Austria and Pfizer Germany. No potential conflicts of interest were disclosed by the other authors.

Conception and design: J. Li, C. Ye, U.R. Mansmann

Development of methodology: J. Li, U.R. Mansmann

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): C. Ye

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): J. Li, U.R. Mansmann

Writing, review, and/or revision of the manuscript: J. Li, U.R. Mansmann

Study supervision: J. Li, U.R. Mansmann

This work is funded by the German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ).

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.
Molina
JR
,
Yang
P
,
Cassivi
SD
,
Schild
SE
,
Adjei
AA
. 
Non–small cell lung cancer: epidemiology, risk factors, treatment, and survivorship
.
Mayo Clin Proc
2008
;
83
:
584
94
.
2.
Jemal
A
,
Simard
EP
,
Dorell
C
,
Noone
AM
,
Markowitz
LE
,
Kohler
B
, et al
Annual Report to the Nation on the Status of Cancer, 1975–2009, featuring the burden and trends in human papillomavirus(HPV)-associated cancers and HPV vaccination coverage levels
.
J Natl Cancer Inst
2013
;
105
:
175
201
.
3.
Kwak
L
,
Bang
YJ
,
Camidge
DR
,
Shaw
AT
,
Solomon
B
,
Maki
RG
, et al
Anaplastic lymphoma kinase inhibition in non–small cell lung cancer
.
N Engl J Med
2010
;
363
:
1693
703
.
4.
Bergethon
K
,
Shaw
AT
,
Ou
SH
,
Katayama
R
,
Lovly
CM
,
McDonald
NT
, et al
ROS1 rearrangements define a unique molecular class of lung cancers
.
J Clin Oncol
2012
;
30
:
863
70
.
5.
Drilon
A
,
Wang
L
,
Hasanovic
A
,
Suehara
Y
,
Lipson
D
,
Stephens
P
, et al
Response to cabozantinib in patients with RET fusion-positive lung adenocarcinomas
.
Cancer Discov
2013
;
3
:
630
5
.
6.
Lynch
TJ
,
Bell
DW
,
Sordella
R
,
Gurubhagavatula
S
,
Okimoto
RA
,
Brannigan
BW
, et al
Activating mutations in the epidermal growth factor receptor underlying responsiveness of non–small cell lung cancer to gefitinib
.
N Engl J Med
2004
;
350
:
2129
39
.
7.
Pao
M
,
Miller
V
,
Zakowski
M
,
Doherty
J
,
Politi
K
,
Sarkaria
I
, et al
EGF receptor gene mutations are common in lung cancers from “never smokers” and are associated with sensitivity of tumors to gefitinib and erlotinib
.
Proc Natl Acad Sci U S A
2004
;
101
:
13306
11
.
8.
Paez
JG
,
Jänne
PA
,
Lee
JC
,
Tracy
S
,
Greulich
H
,
Gabriel
S
, et al
EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy
.
Science
2004
;
304
:
1497
500
.
9.
Maemondo
M
,
Inoue
A
,
Kobayashi
K
,
Sugawara
S
,
Oizumi
S
,
Isobe
H
, et al
Gefitinib or chemotherapy for non–small cell lung cancer with mutated EGFR
.
N Engl J Med
2010
;
362
:
2380
8
.
10.
Sequist
LV
,
Yang
JC
,
Yamamoto
N
,
O'Byrne
K
,
Hirsh
V
,
Mok
T
, et al
Phase III study of afatinib or cisplatin plus pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations
.
J Clin Oncol
2013
;
31
:
3327
34
.
11.
Wu
YL
,
Zhou
C
,
Hu
CP
,
Feng
JF
,
Lu
S
,
Huang
Y
, et al
Afatinib versus cisplatin plus gemcitabine for first-line treatment of Asian patients with advanced non-small-cell lung cancer harbouring EGFR mutations (LUX-Lung 6): an open-label, randomised phase 3 trial
.
Lancet Oncol
2014
;
15
:
213
22
.
12.
Dearden
S
,
Stevens
J
,
Wu
YL
,
Blowers
D
. 
Mutation incidence and coincidence in non small-cell lung cancer: meta-analyses by ethnicity and histology (mutMap)
.
Ann Oncol
2013
;
24
:
2371
6
.
13.
Soda
M
,
Choi
YL
,
Enomoto
M
,
Takada
S
,
Yamashita
Y
,
Ishikawa
S
, et al
Identification of the transforming EML4–ALK fusion gene in non–small cell lung cancer
.
Nature
2007
;
448
:
561
6
.
14.
Antoniu
SA
. 
Crizotinib for EML4-ALK positive lung adenocarcinoma: a hope for the advanced disease? Evaluation of Kwak EL, Bang YJ, Camidge DR, Shaw AT, Solomon B, Maki RG, et al. Anaplastic lymphoma kinase inhibition in non–small cell lung cancer
.
EN Engl J Med.
2010
;
363
:
1693
703
.
Expert Opin Ther Targets 2011;15:351–3
.
15.
Ryu
JS
,
Memon
A
,
Lee
SK
. 
ERCC1 and personalized medicine in lung cancer
.
Ann Transl Med
2014
;
2
:
32
.
16.
Hidalgo
M
,
Amant
F
,
Biankin
AV
,
Budinská
E
,
Byrne
AT
,
Caldas
C
, et al
Patient-derived xenograft models: an emerging platform for translational cancer research
.
Cancer Discov
2014
;
4
:
998
1013
.
17.
Fichtner
I
,
Rolff
J
,
Soong
R
,
Hoffmann
J
,
Hammer
S
,
Sommer
A
, et al
Establishment of patient-derived non–small cell lung cancer xenografts as models for the identification of predictive biomarkers
.
Clin Cancer Res
2008
;
14
:
6456
68
.
18.
Zhang
XC
,
Zhang
J
,
Li
M
,
Huang
XS
,
Yang
XN
,
Zhong
WZ
, et al
Establishment of patient-derived non-small cell lung cancer xenograft models with genetic aberrations within EGFR, KRAS, and FGFR1: useful tools for preclinical studies of targeted therapies
.
J Transl Med
2013
;
11
:
168
.
19.
Bertotti
A
,
Migliardi
G
,
Galimi
F
,
Sassi
F
,
Torti
D
,
Isella
C
, et al
A molecularly annotated platform of patient-derived xenografts (“xenopatients”) identifies HER2 as an effective therapeutic target in cetuximab-resistant colorectal cancer
.
Cancer Discov
2011
;
1
:
508
23
.
20.
Hidalgo
M
,
Bruckheimer
E
,
Rajeshkumar
NV
,
Garrido-Laguna
I
,
De Oliveira
E
,
Rubio-Viqueira
B
, et al
A pilot clinical study of treatment guided by personalized tumorgrafts in patients with advanced cancer
.
Mol Cancer Ther
2011
;
10
:
1311
6
.
21.
Hammer
S
,
Sommer
A
,
Fichtner
I
,
Becker
M
,
Rolff
J
,
Merk
J
, et al
Comparative profiling of the novel epothilone, sagopilone, in xenografts derived from primary non–small cell lung cancer
.
Clin Cancer Res
2010
;
16
:
1452
65
.
22.
Li
J
,
Mansmann
U
. 
Modeling of Non-Steroidal Anti-Inflammatory Drug effect within signaling pathways and miRNA-regulation pathways
.
PLoS ONE
2013
;
8
:
e72477
.
23.
Li
J
,
Mansmann
U
. 
A human molecular signaling map and its application
.
Cell Signal
2014
;
26
:
2834
42
.
24.
Hansen
KD
,
Irizarry
RA
,
Wu
Z
. 
Removing technical variability in RNA-seq data using conditional quantile normalization
.
Biostatistics
2012
;
13
:
204
16
.
25.
Therasse
P
,
Arbuck
SG
,
Eisenhauer
EA
,
Wanders
J
,
Kaplan
RS
,
Rubinstein
L
, et al
New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada
.
J Natl Cancer Inst
2000
;
92
:
205
16
.
26.
Hanahan
D
,
Weinberg
R
. 
The hallmarks of cancer
.
Cell
2000
;
100
:
57
70
.
27.
Hanahan
D
,
Weinberg
R
. 
Hallmarks of cancer: the next generation
.
Cell
2011
;
144
:
646
74
.
28.
Rossi
A
. 
Cetuximab and non-small-cell lung cancer: end of story?
Lancet
2013
;
14
:
1251
3
.
29.
Gridellia
C
,
Bareschinob
MA
,
Schettinob
C
,
Rossi
A
,
Maione
P
,
Ciardiello
F
. 
Erlotinib in non–small cell lung cancer treatment: current status and future development
.
Oncologist
2007
;
12
:
840
9
.
30.
Karaman
MW
,
Herrgard
S
,
Treiber
DK
,
Gallant
P
,
Atteridge
CE
,
Campbell
BT
, et al
A quantitative analysis of kinase inhibitor selectivity
.
Nat Biotechnol
2008
;
26
:
127
32
.
31.
Bayer
R
Binary B-trees for virtual memory [abstract]
. In:
Proceedings of the 1971 ACM_SIGFIDET Workshop on data description, access and control; 1971 Nov 11–12; San Diego; 1971
.
32.
Pepe
MS
,
Feng
Z
,
Janes
H
,
Bossuyt
PM
,
Potter
JD
. 
Pivotal evaluation of the accuracy of a biomarker used for classification or prediction: standards for study design
.
J Natl Cancer Inst
2008
;
100
:
1432
8
.
33.
Herbst
RS
,
Kelly
K
,
Chansky
K
,
Mack
PC
,
Franklin
WA
,
Hirsch
FR
, et al
Phase II selection design trial of concurrent chemotherapy and cetuximab versus chemotherapy followed by cetuximab in advanced-stage non–small cell lung cancer: Southwest Oncology Group study S0342
.
J Clin Oncol
2010
;
28
:
4747
54
.
34.
Lynch
TJ
,
Patel
T
,
Dreisbach
L
,
McCleod
M
,
Heim
WJ
,
Hermann
RC
, et al
Cetuximab and first-line taxane/carboplatin chemotherapy in advanced non–small cell lung cancer: results of the randomized multicenter phase III trial BMS099
.
J Clin Oncol
2010
;
28
:
911
7
.
35.
Shaw
RJ
,
Cantley
LC
. 
Ras, PI(3)K and mTOR signalling controls tumour cell growth
.
Nature
2006
;
441
:
424
30
.
36.
Pepe
MS
. 
The Statistical Evaluation of medical tests for classification and prediction
(
Oxford Statistical Science Series
). 1st ed.
USA
:
Oxford University Press
; 
2004
.
37.
Gandara
DR
,
Lara
PN
 Jr
,
Mack
PC
. 
Patient-derived xenografts for investigation of acquired resistance in oncogene-driven cancers: building a better Mousetrap
.
J Clin Oncol
2015
;
33
:
2839
40
.
38.
Whittle
JR
,
Lewis
MT
,
Lindeman
GJ
,
Visvader
JE
. 
Patient-derived xenograft models of breast cancer and their predictive power
.
Breast Cancer Res
2015
;
17
:
17
.
39.
Tentler
JJ
,
Tan
AC
,
Weekes
CD
,
Jimeno
A
,
Leong
S
,
Pitts
TM
, et al
Patient-derived tumour xenografts as models for oncology drug development
.
Nat Rev Clin Oncol
2012
;
9
:
338
50
.
40.
Al-Wadei
HA
,
Plummer
HK
 III
,
Ullah
MF
,
Unger
B
,
Brody
JR
,
Schuller
HM
. 
Social stress promotes and γ-aminobutyric acid inhibits tumor growth in mouse models of non–small cell lung cancer
.
Cancer Prev Res
2012
;
5
:
189
96
.
41.
Banerjee
J
,
Papu John
AM
,
Schuller
HM
. 
Regulation of non–small cell lung cancer stem cell like cells by neurotransmitters and opioid peptides
.
Int J Cancer
2015
;
137
:
2815
24
.
42.
Couve
A
,
Thomas
P
,
Calver
AR
,
Hirst
WD
,
Pangalos
MN
,
Walsh
FS
, et al
Cyclic AMP-dependent protein kinase phosphorylation facilitates GABA(B) receptor-effector coupling
.
Nat Neurosci
2002
;
5
:
415
24
.
43.
Scheer
M
,
Grote
A
,
Chang
A
,
Schomburg
I
,
Munaretto
C
,
Rother
M
, et al
BRENDA, the enzyme information system in 2011
.
Nucleic Acids Res
2011
;
39
:
670
6
.