Abstract
Background
Immune checkpoint inhibitors (CPIs) have revolutionized cancer therapy
for several tumor indications. However, a substantial fraction of
patients treated with CPIs derive no benefit or have short-lived
responses to CPI therapy. Identifying patients who are most likely to
benefit from CPIs and deciphering resistance mechanisms is therefore
essential for developing adjunct treatments that can abrogate tumor
resistance.
Patients and methods
In this study, we used a machine learning approach that used the
US-based nationwide de-identified Flatiron Health and Foundation
Medicine non-small cell lung carcinoma (NSCLC) clinico-genomic database
to identify genomic markers that predict clinical responses to CPI
therapy. In total, we analyzed data from 4,433 patients with NSCLC.
Results
Analysis of pretreatment genomic data from 1,511 patients with NSCLC
identified. Of the 36 genomic signatures identified, 33 exhibited
strong predictive capacity for CPI response (n=1150) compared with
chemotherapy response (n=361), while three signatures were prognostic.
These 36 genetic signatures had in common a core set of four genes
(BRAF, BRIP1, FGF10, and FLT1). Interestingly, we observed that some
(n=19) of the genes in the signatures (eg, TP53, EZH2, KEAP1 and FGFR2)
had alternative mutations with contrasting clinical outcomes to CPI
therapy. Finally, the genetic signatures revealed multiple biological
pathways involved in CPI response, including MAPK, PDGF, IL-6 and EGFR
signaling.
Conclusions
In summary, we found several genomic markers and pathways that provide
insight into biological mechanisms affecting response to CPI therapy.
The analyses identified novel targets and biomarkers that have the
potential to provide candidates for combination therapies or patient
enrichment strategies, which could increase response rates to CPI
therapy in patients with NSCLC.
Keywords: Lung Cancer, Biomarker, Genome, Immune Checkpoint Inhibitor
__________________________________________________________________
WHAT IS ALREADY KNOWN ON THIS TOPIC
* Checkpoint inhibitors (CPIs) have transformed cancer treatment,
particularly for non-small cell lung cancer, by achieving prolonged
responses in many patients. However, a significant portion of
patients either do not respond or develop resistance to CPI
therapy.
WHAT THIS STUDY ADDS
* Among the 36 genomic signatures identified, 33 demonstrated strong
predictive capacity for CPI response relative to chemotherapy
response, while three signatures (hotspot granular signatures 3, 4,
and 9) did not meet the predictive threshold. The study revealed
core genes such as BRAF, BRIP1, FGF10, and FLT1 that are crucial
for CPI outcomes. The findings also highlight that different
mutations within the same gene can lead to contrasting responses to
CPI therapy. Moreover, the study uncovers multiple biological
pathways, including MAPK, PDGF, IL-6, and EGFR signaling,
associated with CPI response, providing new insights into the
mechanisms of resistance and potential targets for combination
therapies.
HOW THIS STUDY MIGHT AFFECT RESEARCH, PRACTICE OR POLICY
* The identification of specific genetic signatures and pathways
associated with CPI response can guide the development of new
predictive biomarkers and targeted therapies, potentially improving
patient stratification and treatment outcomes. These insights could
lead to personalized treatment strategies and the development of
combination therapies to overcome CPI resistance, ultimately
enhancing clinical practice and informing policy decisions on
cancer treatment protocols.
Introduction
Lung cancer is the leading cause of cancer-related mortality worldwide,
and approximately 85% of these tumors are of non-small cell lung cancer
(NSCLC) histology.[41]^1 2 The discovery and subsequent regulatory
approval of checkpoint inhibitors (CPI) has revolutionized treatment of
diverse cancer types including lung cancer, by achieving prolonged
responses in many patients.[42]3,[43]7 Despite the unprecedented,
prolonged response rates to CPIs, a substantial fraction of patients
become resistant to CPI therapy.[44]^8
There is, therefore, an urgent and unmet need to advance our
understanding of CPI resistance mechanisms to promote the development
of predictive biomarkers and identify potential targets for novel
therapies. Extensive efforts are underway to identify predictive
biomarkers using various omics, histopathologic, clinical, and
computational approaches.[45]9,[46]12 Presently, the only approved
biomarkers for CPI therapy are programmed death ligand-1 (PD-L1),
microsatellite instability and tumor mutational burden (TMB).[47]^13
However, these approved biomarkers do not provide the much-needed
mechanistic insight behind CPI response. To gain insight into CPI
resistance mechanisms in NSCLC and discover predictive genomic
biomarkers, we used an interpretable machine learning (ML) framework
that uses a genetic algorithm (GA) for feature selection. The patient
data was obtained from the nationwide (US-based) de-identified Flatiron
Health and Foundation Medicine NSCLC clinico-genomic database (FH-FMI
CGDB). Using our GA framework, we discovered tumor intrinsic genetic
signatures that can predict CPI response. We discovered that different
mutations within the same gene may lead to opposing clinical responses.
Finally, we used several network-based algorithms to discover pathways
associated with CPI response. Our findings provide insights into
potential mechanisms underlying CPI response in NSCLC, which are
essential for developing new therapeutics that target CPI resistance
and discovering biomarkers for the prediction of CPI response.
Results
Patient cohorts and characteristics
Among 7,360 patients with NSCLC in the FH-FMI CGDB, we found 1,150
advanced stage patients who had received monotherapy with
anti-programmed cell death protein 1 (PD-1) or anti-PD-L1 antibodies
(referred to hereafter as CPI) and had either response or resistance to
CPI ([48]figure 1A,B). The de-identified longitudinal patient data
originated from approximately 280 US cancer clinics (~800 sites of
care). These patients had their pretreatment tumor specimens sequenced
by the Foundation Medicine panels.[49]^14 We selected patients treated
with CPIs as monotherapy to eliminate the confounding effect of
combination therapy on CPI response.
Figure 1. Definition, selection and characteristics of patient outcome
groups. (A) Schematic representation of definition for response and
resistance patient groups. The long blue horizontal arrows represent the
patient’s journey over time. The study duration (270 days), in which
treatment outcomes were investigated, is marked by a green dotted open box.
Green vertical arrows represent the first time a patient was treated with a
CPI, and the greyed-out area represents buffer time (14 days) during which
treatment outcomes were not considered because they were likely influenced by
earlier treatments. The green circle represents real-world response from CPI
therapy, while the red X represents disease progression. Patients in the
response group exhibited durable clinical benefit without evidence of disease
progression during the 270-day study period. In contrast, patients with
innate resistance—defined as those who did not benefit from CPI and
experienced disease progression within the same time frame—were included in
the analyses. Patients with acquired resistance, characterized by an initial
response followed by disease progression within the 270-day study period,
were excluded. (B) Schematic representation of the number of selected
patients in the CPI treatment cohort, based on the exclusion criteria
depicted in the filter box in the scheme (more details in methods). (C)
Clinical characteristics of the patient cohorts divided into chemotherapy
(depicted as chemo) and checkpoint inhibitors (depicted as CPI). CPI,
checkpoint inhibitors; NSCLC, non-small cell lung cancer; TMB, tumor
mutational burden.
[50]Figure 1
[51]Open in a new tab
The patients were categorized into the two response groups using
clinical outcome data[52]^15 16 available in CGDB. Almost all patients
with at least one line of therapy had real-world progression
(rwP)[53]^15 data, and about 84% had real-world response (rwR)[54]^16
data. The first response group consisted of 574 patients who had
durable rwR and did not show rwP during the entire study duration of
270 days were considered to have response (referred to as “response”
patients).[55]^12 15 The second group had 576 patients who did not have
rwR and showed rwP within the study and were considered to be
CPI-resistant (referred to as “resistance” patients) ([56]figure 1A,B).
Comparison of the clinical characteristics of response and resistance
patients revealed no significant difference in gender, race, or disease
stage ([57]figure 1C), but did show significant differences in age,
TMB, smoking status, and histology. These differences are consistent
with previous reports which show the frequency of response is greater
in older patients,[58]^17 patients with higher TMB,[59]^18 different
histology, as well as patients with a history of smoking.[60]^12 19 The
majority of these patients received CPI as their first-line therapy
(89% 1 L, 10% 2 L).
Mutational landscape of patients with NSCLC
We characterized the mutational landscape of our patient group
([61]figure 2A). The five most frequent short variants (single
nucleotide variants and short insertions/deletions) were in TP53, KRAS,
KMT2D, KEAP1 and STK11 ([62]figure 2B). The five most frequent
amplifications were of SOX2, SDHA, RICTOR, KLHL6 and PIK3CA ([63]figure
2C), while the top five most frequent deletions were of CDKN2A, CDKN2B,
PTEN, STK11 and TEK ([64]figure 2D). The five most frequent
rearrangements were in CDKN2A, FGFR1, LRP1B, EGFR and TP53 ([65]figure
2E). A comparison of the top 12 altered genes between response and
resistance patients did not reveal any obvious differences in mutation
frequencies between the two cohorts, with the exceptions of STK11
mutations, which occurred at higher frequency in resistance patients
(14% vs 22%) ([66]online supplemental figure S1C). Cumulatively, the
mutational landscape analysis confirmed high-frequency mutations known
in NSCLC but did not reveal gross differences between response and
resistance patients.
Figure 2. Mutational landscape of the CPI cohort. (A) Oncoplot of the CPI
cohort (n=1,150), depicting the top 20 altered genes and some clinical
characteristics. Columns represent patients, rows represent genes. The top
bar plot shows mutation count and type (missense, splice site, frame shift,
etc) per patient. The middle section indicates mutations in the specified
gene in patients. The bottom heatmap shows clinical characteristics (outcome,
histology, smoking status, and gender) with a color scheme depicted at the
bottom of the figure. The right-side bar graph summarizes the total count and
proportion of mutation types for each gene. (B) Bar graph of top short
variants in the CPI cohort (n=1,150) with color stacking representing
mutation types within each gene (same color scheme as in A). (C) Bar graph
showing prevalence (%) of the top 10 amplified genes. (D) Bar graph showing
prevalence (%) of the top 10 deleted genes. (E) Bar graph showing prevalence
(%) of the top 10 rearrangements. CPI, checkpoint inhibitor.
[67]Figure 2
[68]Open in a new tab
Mutations affecting response to CPIs
The conventional approach to correlating mutations with an observable
characteristic involves testing for statistical significance of the
occurrence of each mutation in its corresponding group. This type of
analysis is useful but does not provide sufficient insight into the
complex biology of response to CPI. Moreover, different mutations
within the same gene can result in distinct biological
outcomes,[69]20,[70]23 which can also be influenced by tissue type,
tumor microenvironment, and/or xenobiotics.[71]^24 With the above
considerations in mind, we categorized the mutational data using three
different approaches (Methods). The first approach was the traditional
classification which notates if the gene is mutated or wild-type (WT),
and is referred to hereafter as binary classification/input. The second
approach divides gene mutations into high-frequency mutations (HFM),
and low-frequency mutations (LFM) (eg, TP53 HFM or TP53 LFM), and is
referred to after this as HFM/LFM classification/input. The third
approach further separates HFM mutations and adds the location of the
amino acid change (eg, TP53 HFM 178, TP53 HFM 248), and is referred to
after this as hotspot granular classification/input. Classifying the
mutations into HFM/LFM and hotspots allowed us to further delineate the
underlying biological mechanism as HFM tend to be gain of function
mutations and LFM tend to be loss of function mutations.[72]^25 We used
the Fisher’s exact test within each set (ie, binary, HFM/LFM and
hotspot granular) to check for mutation enrichment in response or
resistance patients. The analysis revealed that in the binary
classification set, BRAF, BRIP1 and ASXL1 mutations were associated
with response (false discovery rate [FDR]=0.053), while STK11 and
CDKN2B mutations were associated with resistance (FDR=0.053 and 0.056,
respectively; [73]figure 3A, [74]online supplemental table S1A).
Consistent with the BRAF effect on CPI response in NSCLC, we obtained
similar data when analyzing an independent melanoma cohort (data not
shown). In the HFM/LFM classification, we found that BRAF_HFM and
BRIP1_LFM were associated with response (FDR=0.046 and 0.068,
respectively; [75]figure 3B, [76]online supplemental table S1B), and
CDKN2B_LFM (FDR=0.1) was associated with resistance. In the
hotspot-granular set, we found no statistically significant results
after multiple testing adjustments ([77]figure 3C, [78]online
supplemental table S1C). Since mutations tend to co-occur with other
genomic alterations, we examined if there were any significant pairs or
triples of co-occurring mutations associated with CPI response. In
binary classification, we found that STK11 and KRAS co-occurrence was
the only significant pair (FDR=0.023) associated with resistance, and
co-occurring mutations in KEL and TP53 were only marginally associated
with response (FDR=0.096; [79]figure 3A, [80]online supplemental table
S2A). No significant co-occurring mutations in HFM/LFM or hotspot
classifications were observed, but the top result of co-occurring
mutations in KRAS_HFM and TERT_HFM (FDR=0.22) and KRAS_HFM_12 and
PIK3C2G_HFM (FDR=0.25) were associated with response ([81]figure 3B,C,
[82]online supplemental table S2B-C). Additionally, we found no
significant triplet mutations, but top results included co-occurring
mutations in KRAS-NF1-TP53 (FDR=0.3) and ARID1A_LFM-CDKN2A_LFM-TP53_LFM
(FDR=0.33) ([83]figure 3A,B, [84]online supplemental table S3A-B).
Figure 3. Mutation association with outcome in the 1,150-patient cohort.
(A–C) Forest plot of the top-ranked genes of binary (A), HFM/LFM (B) and
hotspot (C) mutations by Fisher’s exact test comparing mutation rates between
patients in response and resistance groups. Each plot shows the top 15 single
gene and top 2 double and triple gene mutations co-occurrences separated by
dashed horizontal lines. Each plot shows the gene name (Gene) and the number
of patients who responded (N_resp) or were resistant (N_resist) having that
single or combination mutation, with the x-axis representing log-scaled OR.
OR>1 indicates association of the mutation with response to CPI, and <1
indicates association with resistance to CPI. CPI, checkpoint inhibitors;
HFM, high frequency mutations; LFM, low frequency mutations.
[85]Figure 3
[86]Open in a new tab
BRAF, BRIP1, STK11, ASXL1, CDKN1B, and co-occurring STK11-KRAS mutations
affect overall survival in patients treated with CPIs
While we found a significant association ([87]figure 3) between several
mutations and response to CPI, we wanted to investigate if mutations in
the genes listed above, affect overall survival (OS) in the 1,150
patient cohort. Additionally, we wanted to explore if the mutations
could be predictive for CPI-specific response or prognostic
(independent of treatment). We therefore compared OS of patients
treated with CPI monotherapy with 361 patients treated with
chemotherapy (patient characteristics are in [88]online supplemental
figure S1A, complete analysis results are in Table S4). OS analysis
revealed significantly greater OS among CPI-treated patients with
mutations in BRAF and a trend for BRIP1 (p=0.09), and significantly
lower OS with mutations in CDKN2B and STK11 (p=0.02) ([89]figure 4A).
No significant change in survival was observed in patients with ASXL1
mutations (p=0.5) ([90]figure 4A). A significantly lower OS among
CPI-treated patients was also observed for patients with co-occurring
mutations in KRAS and STK11 (p=0.0013) ([91]figure 4B). No significant
change in survival was observed in chemo-treated patients with these
gene mutations, indicating that the survival benefit is CPI-specific
([92]figure 4A,B). Of note, we did observe a sharp decrease in survival
among chemo-treated patients with STK11 or STK11 and KRAS co-occurring
mutations ([93]figure 4A,B), but since the survival lines crossed, it
was not statistically significant, though it might be clinically
relevant. Although we did not find statistically significant triplet
mutations in the response data analysis ([94]figure 3A), the top
triplet mutations in KRAS, NF1 and TP53 showed a significant
(p=0.00041) increase in OS ([95]figure 4C). The statistically
non-significant result in the response data analysis might be due to
the small sample sizes (n=20 vs 2).
Figure 4. Overall survival of the 1,150-CPI patient cohort and 361-chemo
patient cohort with mutations found to be significantly associated with CPI
outcome. (A–C) Kaplan-Meier survival curves of overall survival in patients
treated with CPI (left plots) or chemo (right plots) with wild-type (wt; blue
lines) or mutated (mut; red lines) gene in top single (A), double (B) or
triple (C) gene mutations demonstrating a trend for statistical significance
in [96]figure 3. The bottom table in each Kaplan-Meier plot shows the number
of patients at risk at each time point (in month). P values indicated inside
each graph were from the log-rank test comparing patient groups within each
plot. CPI, checkpoint inhibitors.
[97]Figure 4
[98]Open in a new tab
We further investigated the correlation of the OS to the mutations in
the genes described above, with an extended cohort of patients. This
extended cohort of 4,433 patients was selected from the 7,360 patients
with NSCLC in the CGDB the same way as the 1,150-patient cohort
([99]figure 1B). The only difference was that there was no outcome
definition criteria filter for the larger cohort (ie, regardless of
drug response data availability) and regardless of their death status.
These included 2,699 patients treated with CPI monotherapy and 1,734
chemo-treated patients. Examining the effect of the mutations analyzed
in [100]figure 4, on survival in the extended cohort confirmed that
CPI-treated patients with mutations in BRAF, BRIP1 and ASXL1 showed
significant increases in OS when compared with WT patients (p=0.011,
p=0.038, p=0.04, respectively) ([101]online supplemental figure S2A).
Notably, there was no significant difference in OS in chemo-treated
patients with these mutations versus those without ([102]online
supplemental figure S2A). CPI-treated patients with STK11 or CDKN2B
mutations, and co-occurring mutations in STK11 and KRAS, showed a
significant reduction in OS (p=0.0047, p=0.00013 and p=0.0015,
respectively; [103]online supplemental figure S2A,B). We did, however,
observe that chemo-treated patients with these mutations also showed
significant decreases in OS (p<0.0001, p=0.0068 and p=0.0018,
respectively), suggesting these mutations may not have a CPI-specific
effect but are prognostic ([104]online supplemental figure S2A,B).
Although the top triplet mutations in KRAS, NF1 and TP53 showed a
significant substantial increase in OS (p=0.00041) in the 1,150-patient
cohort, the increase was only marginally significant (p=0.056) in the
larger CPI-treated cohort ([105]figure 4C and [106]online supplemental
figure S2C). The same trend was observed for co-occurring mutations in
KEL and TP53 ([107]figure 3 and data not shown). Cumulatively, the
results of [108]figures3 [109]4 are in concordance, and the effect of
the mutations on OR of response and resistance presented in [110]figure
3 and OS presented in [111]figure 4 followed the same pattern and
directionality.
Machine learning workflow identifies genetic markers of CPI outcomes
We used a GA to evolve virtual individuals, each represented by a set
of mutated genes, over multiple generations. The predictive power of
each virtual individual for CPI response was assessed using Naive Bayes
models, with cross-validation (CV) accuracy scores serving as the
evaluation metric. The fittest individuals were selected iteratively
for subsequent generations. From the top 5% of these individuals, based
on their accuracy scores, we identified 12 distinct clusters using
k-means clustering. Within each cluster, the set of mutated genes that
appeared in more than 50% of the members was identified as the genetic
signature for that cluster ([112]online supplemental figure S3,
[113]figure 5A). We identified several mutations affecting outcomes to
CPI including co-occurring mutations, which suggests that these genes
in tandem or as a single gene may mediate outcomes to CPI. This
suggested that decoding the biological mechanism(s) behind CPI outcomes
requires broadening the analysis beyond one or two genes in isolation.
Such a broad analysis aims at identifying a genetic signature(s) (i.e.,
a collection of tumor mutations) that can be used to predict response
to CPI. TMB is an established and important biomarker that correlates
with response to CPI therapy,[114]^26 which we recently validated for
predicting response in NSCLC.[115]^12 We therefore used TMB as a
benchmark for our subsequent results. The TMB model trained on our
NSCLC cohort showed a receiver operating characteristic (ROC) with an
area under the curve (AUC) score of 0.6 ([116]figure 5B–E). Using an ML
analysis workflow (described in Methods), we examined 977 out of the
1,150 patients as the initial training set. The results yielded 36
genetic signatures consisting of 12 signatures for each input: binary,
HFM/LFM and hotspot (Table S5). The average CV ROC AUC scores of these
signatures was 0.7±0.02 (Table S6). We then used an independent test
(validation) set of 173 patients to examine the association of the 36
genetic signatures to CPI response. The average resultant ROC AUC
scores were 0.6±0.02 compared with 0.6 with TMB ([117]figure 5B–D,
Table S6), which underscores the predictive power and generalizability
of these genetic signatures. The average AUC scores for predicting
response in 361 chemo-treated patients using the 36 genetic signatures
was 0.5±0.02, which is consistent with random chance, confirming that
the genetic signatures were CPI response-specific. These results are
consistent with previously published AUC values for TMB[118]^27 28 and
provide evidence that these genetic signatures perform as well as the
established TMB biomarker in prediction of CPI response. However, these
results provide an additional, important advantage since the predictive
mutations can be explored for mechanistic understanding of outcomes to
CPI, which is not possible with TMB.
Figure 5. Machine learning algorithm identifies 36 predictive genetic
signatures with a core of shared genes. (A) Schematic depiction of the
machine learning algorithm. (B–E) Plots showing the area under the ROC curve
(AUC) from the independent validation set (n=173 for CPI and n=361 for
chemo-treated patients) using the genetic signatures from different input
types. (B) Binary, (C) HFM/LFM and (D) hotspot granular and (E) AUC scores
for the three inputs with all unique gene mutation features (82 for binary,
130 for HFM/LFM, and 123 for hotspot granular) from the 12 models combined
into one model for each input type. For each graph, the blue dashed
horizontal line represents the score for the TMB model, and the red dashed
line represents the 50% AUC (depicted as random chance). (F) The top Venn
diagram represents the gene overlap between the 36 individual genetic
signatures across binary, HFM/LFM and hotspot granular inputs, showing that
four genes are included in every single genetic signature of the 36
signatures. The bottom Venn diagram represents overlap across the unique
genes in the 12 signatures of the three inputs (82 binary, 130 HFM/LFM, and
123 hotspot). Detailed information about the overlap is shown in [119]online
supplemental figure S4. CPI, checkpoint inhibitors; HFM, high frequency
mutations; LFM, low frequency mutations; ROC, receiver operating
characteristic; TMB, tumor mutational burden.
[120]Figure 5
[121]Open in a new tab
Characterization of 36 genetic signatures revealed a core set of genes most
frequently associated with CPI response
Since each of the three ML inputs generated 12 separate predictive
genetic signatures (Table S5), we first examined if each genetic
signature within the inputs is diverse. Indeed, the heatmap analysis in
[122]online supplemental figure S4 shows that the 36 signatures are
diverse with some overlaps between them ([123]online supplemental
figure S4). Next, we assessed the frequency of each gene mutation
feature across the 36 genetic signatures that might indicate their
utility for CPI outcomes. We found that within each of the binary,
HFM/LFM and hotspot inputs, there was an overlap of 13, 17, and 20
genes, respectively, within the 12 genetic signatures ([124]online
supplemental figure S4 and Table S7A). These genes were present in all
12 genetic signatures within each input, suggesting that these are
necessary in generating predictive genetic signatures and play an
important role in CPI outcomes. Among these overlaps, four genes
(BRIP1, FLT1, FGF10 and BRAF) were shared ([125]figure 5F top,
[126]online supplemental figure S4 and Table S7B), representing a core
set of mutations most important for predicting CPI outcomes. Next, we
identified 82, 130, and 123 unique gene mutation features contained in
each of the binary, HFM/LFM and hotspot inputs, respectively (Table
S7C). Using these gene mutation features, we built three models, one
for each input type that have ROC AUC scores of 0.61–0.66 ([127]figure
5E and Table S7D). Among the unique gene mutation features, 58 genes
were overlapping, further distinguishing a core set of mutations
central to predicting CPI outcomes ([128]figure 5F, Table S7E).
Overall, this ML approach revealed a significant number of genes, many
of which are previously unreported, which may play a role in outcomes
to CPI therapy.
Relative contributions of individual gene mutations to overall CPI response
To interpret the biological impact of these mutations, we used the
linear Naive Bayesian model. It enabled us to: (1) quantify the
relative contribution of each gene mutation feature to CPI outcomes,
and (2) associate each mutation with response or resistance.
Accordingly, we were able to assign a numerical rank to each gene
mutation feature by their importance to CPI outcomes ([129]figure 6A,
Table S8A). In binary genetic signatures, we found that CTNNA1 was the
top contributor to response and RARA was the top contributor to
resistance. In HFM/LFM genetic signatures, we found that ALOX12B LFM
mutations were the top contributor to response and MYC LFM mutations
were the top contributor to resistance. In hotspot genetic signatures,
we found that ALOX12B LFM was the top contributor to response and EGFR
mutation at amino acid 746 was the top contributor to resistance.
CTNNA1 and RARA and other mutations that were prominent in the binary
signature were also highly ranked in HFM/LFM and hotspot indicating
consistency between inputs of the impact of these genes on response to
CPI.
Figure 6. ML identifies genetic markers associated with outcomes to CPI. (A)
Waterfall plot of the top 10 genes of the 36 ML-derived genetic signatures
ranked by feature importance using the three different inputs (left panel for
binary, middle for HFM/LFM and right for hotspot). Positive values indicate
association with response (green bars), and negative values with resistance
(red bars) to CPI. (B) Plot representing feature importance in genes in which
HFM and LFM mutations within the same gene have divergent contribution to CPI
outcome, with red associated with resistance and green with response. CPI,
checkpoint inhibitors; HFM, high frequency mutations; LFM, low frequency
mutations; ML, machine learning.
[130]Figure 6
[131]Open in a new tab
Remarkably, we observed a set of genes that had opposite effects on CPI
response, depending on the type of mutation ([132]figure 6B, Table
S8B). For example, in HFM/LFM input, we found that LFM mutations in
FGFR2 were associated with response, while HFM mutations were
associated with resistance ([133]figure 6B). Furthermore, in hotspot
inputs, we found that certain TP53 HFM mutations were associated with
response (TP53_HFM_331), while other TP53 HFM mutations were associated
with resistance (TP53_HFM_152) (Table S8A). These findings suggest that
different mutations within the same gene can lead to opposite outcomes
in CPI-treated patients.
Topology/network-based pathway enrichment analysis reveals pathways
associated with CPI response
While it is important to identify individual mutations (or
combinations) that affect CPI outcomes, there is a need to understand
the biological processes affected by these unique gene mutations and
how they affect CPI response. To explore the biological processes
influenced by the genetic signatures, we investigated whether these
signatures fall into meaningful biological processes. We applied a
network/topology-based pathway analysis that takes the upstream and
downstream gene–gene or protein–protein interactions into account. We
used six different algorithms to perform topology-assisted pathway
analysis (please see Methods), using the Computational Biology Methods
for Drug Discovery (CBDD) R package[134]^22 (please see Methods) which
facilitated the discovery of additional pathways that would otherwise
not be detected with an enrichment-only approach. With binary input,
the top pathways across six algorithms were associated with PDGF,
oxidative stress, prolactin, thrombopoietin, AGTR1, IL-3 and YAP/TAZ
and IL-6 signaling ([135]figure 7A, Table S9A). For HFM/LFM input, the
uppermost pathways were associated with androgen, IL-6, PDGF,
epigenetics, oxidative stress, ErbB2 and EGFR signaling ([136]figure
7B, Table S9B). For hotspot input, we found the uppermost pathways to
be associated with PDGF, androgen, IL-6, AGTR1, HGF, EGFR and IL-2
signaling ([137]figure 7C, Table S9C). The enrichment of PDGF and IL-6
as well as other pathways was shared across the binary, HFM/LFM and
hotspots inputs, indicating consistency between these inputs to enable
identification of pathways relevant for response to CPI ([138]figure
7D, Table S9D). Since there was an overlap of 58 genes between the
three ML inputs ([139]figure 5F), we examined the pathways that were
enriched in this overlap. This approach revealed pathways associated
with IFN alpha/beta, PDGF, MAPK, oxidative stress, ALK, ErbB2, IL-6,
leptin, prolactin and IL-2 signaling ([140]figure 7E, Table S9E). The
overall highest-ranking pathways among all inputs ([141]figure 7A–C)
were associated with PDGF, oxidative stress, androgen, MAPK, prolactin,
AGTR1, IL-6, EGFR and YAP/TAZ signaling ([142]figure 7E, Table S9E). In
summary, our pathway analysis reveals several tumor intrinsic pathways
like oxidative stress and PDGF involved in outcomes to CPI as well as
several previously reported immune response-related pathways like
interleukin (IL)-6 and IL-2 to be involved in outcomes to CPI.
Figure 7. Pathway analysis of predictive genetic signatures associated with
CPI outcomes. This figure illustrates the immune response and other
biological pathways associated with CPI outcomes, identified through pathway
analysis of predictive genetic signatures. The top 15 pathways were derived
using the Computational Biology Methods for Drug Discovery pathway analysis,
which used six different network-based algorithms. Pathways are ranked by the
lowest p values (derived from the hypergeometric test) across the algorithms
and are sorted according to the composite score (see Methods for details).
(A–C) The top pathways identified from the (A) binary, (B) HFM/LFM, and (C)
hotspot inputs. (D) The top pathways from A to C (cross input pathways). (E)
Pathways enriched using the 58 overlapping genes among the genetic signatures
from the three inputs, as shown in the bottom Venn diagram of [143]figure 5F
(overlap pathways). Cross input pathways (D) refer to the pathways that are
consistently identified across all three input types (binary, HFM/LFM, and
hotspot), indicating their broad relevance. Overlap pathways (E) refer to the
pathways identified from the subset of genes that are common across all three
genetic signature inputs, highlighting specific pathways that are crucial due
to the shared gene set. CPI, checkpoint inhibitors; HFM, high frequency
mutations; LFM, low frequency mutations.
[144]Figure 7
[145]Open in a new tab
Higher IL-6 serum levels are associated with progressive disease in NSCLC
Since IL-6 signaling appeared in the results of analyses in multiple
pathways, we investigated its importance in a cohort of patients with
NSCLC treated with atezolizumab. We found that serum levels of IL-6
were elevated in both stable disease (SD) and progressive disease (PD)
patients when compared with partial response (PR) patients using either
RECIST (Response Evaluation Criteria in Solid Tumors) ([146]figure 8A)
or irRC (immune-related response criteria) (data not shown).
Furthermore, OS analysis of patients with NSCLC treated with
atezolizumab revealed a significantly longer survival in patients with
lower IL-6 serum levels, which is consistent with previous and more
recently published reports[147]29,[148]31 ([149]figure 8B). The
involvement of IL-6 in resistance to atezolizumab validates our ML
algorithm, which allows greater delineation of the biological pathways
and mechanism(s) behind resistance or response to CPI.
Figure 8. IL-6 validation in the atezolizumab study shows high serum levels
correlate with disease progression. (A) Boxplot depicting baseline (cycle
1 day one pre-dose) serum IL-6 levels in patients whose response to
atezolizumab was assessed by the RECIST criteria (CR, PR, SD or PD). Unpaired
two-sided t-test p values were shown above the boxes. (B) Kaplan-Meier curves
of overall survival of patients treated with atezolizumab (trial PCD4989G)
comparing patients with high (blue) and low (red) IL-6 serum levels. The
number of patients at risk is given below the Kaplan-Meier plot. P value
indicated inside the graph was from the log-rank test comparing the two
patient groups. CR, complete response; IL, interleukin; PD, progressive
disease; PR, partial response; RECIST, Response Evaluation Criteria in Solid
Tumors; SD, stable disease.
[150]Figure 8
[151]Open in a new tab
Discussion
In this study, we report a comprehensive analysis of tumor genomic
determinants that affect outcomes of patients with NSCLC to CPI.
We classified patients by outcomes over an extended period, which
enabled a different perspective compared with objective response rate,
with the extra length of time in our analyses better representing
real-world outcomes of CPI therapy. We focused on innate resistance
rather than acquired resistance. By targeting innate resistance, we
aimed to identify genomic mechanisms intrinsic to tumors that drive
resistance to CPI therapy. This distinction is crucial for
understanding pre-existing actionable resistance pathways. Finally, the
exclusion of early death events in our analyses increases the focus on
drug response and excludes patients who had extremely advanced disease
and morbidities, allowing us to focus on predictive rather than
prognostic effects.[152]^12 This approach enables more focused data
analysis for biomarkers of response. The biomarkers identified from
this analysis included those (eg, STK11 and STK11-KRAS), which have
been previously reported to be associated with response to CPI.[153]^32
33
Using a traditional statistical test comparing response and resistance,
we found that mutations in single genes (BRAF, BRIP1, ASXL1, and
CDKN2B) as well as co-occurring mutations (TP53-KEL and TP53-KRAS-NF1)
correlate with CPI response, including OS. CDKN2B deletion correlated
with decreased OS in CPI-treated patients, which is in keeping with a
previous report in advanced urothelial carcinoma.[154]^34 35 However,
CDKN2B deletion, in our studies with the larger cohort of patients,
correlated with a decrease in OS in chemo-treated patients as well,
suggesting this to be a prognostic rather than predictive marker for
CPI outcomes. We did not observe any significant OS differences between
patients with mutations in TP53 alone but found a significant
difference if TP53 mutations co-occurred with KRAS and NF1. This data
from the co-occurring mutations suggests that specific combinations of
mutations may affect outcomes, and provides insight beyond conventional
single-gene analyses.
We classified the mutations as HFM and LFM, where HFM and LFM tend to
have gain of function and loss of function characteristics,
respectively.[155]^25 We found that not all HFM and LFM or hotspots
mutations within the same gene lead to similar clinical outcomes with
CPI, including TP53[156]^36 and KEAP1.[157]^32 33 36 KEAP1 was
previously reported to be associated with resistance to CPI
therapy.[158]^32 33 However, our analysis showed that not all mutations
in KEAP1 lead to resistance, and while KEAP1 HFM 320 was associated
with resistance, LFM were associated with response. These divergent
therapy outcomes to different mutations within the same gene were also
found in other genes. The number of genes indicates that this
observation is not anecdotal, and that binary classification (mutated
vs WT) of genes is insufficient for exploring the role of mutations in
outcomes to CPI, as two different mutations in the same gene may lead
to opposite clinical consequences. The interpretation of how different
mutations, in the same gene, lead to opposite outcomes requires
experimental investigation as it can be due to different active
mechanisms or non-causal factors. These divergent outcomes from
different mutations in the same gene also suggest that it is necessary
to assess each individual mutation and its effect on CPI outcome, but
this is presently not feasible due to the high number of these variants
that results in limitations in statistical power.
These signatures, as well as a shared signature identified from the
common gene set, could inform a patient enrichment strategy for
patients with NSCLC for treatment with CPI. The signature genes were
analyzed with network algorithms to identify the biological pathways
influencing CPI response. This identified several pathways
significantly associated with CPI outcomes. Notably, some of these
pathways were previously reported to be involved with CPI outcomes like
IFN-alpha/beta,[159]^37 38 EGFR[160]^39 40 and IL-6,[161]^29 41 42
further credence to our analysis. The association of these pathways
with CPI outcomes may have therapeutic implications, as there are
already several Food and Drug Administration-approved drugs that can
target these pathways being tested in combination with CPIs to increase
response rate.[162]^43 Additionally, using data available from a phase
I clinical study, we confirmed IL-6 involvement in CPI resistance and
showed that higher baseline serum levels of IL-6 in CPI-treated
patients were associated with disease progression and poor OS. This is
in line with the reports on higher IL-6 serum levels leading to worse
outcomes in metastatic urothelial bladder cancer and in metastatic
renal cell carcinoma treated with anti-PD-L1,[163]^29 44 45 implying
that IL-6 signaling might be an important target in several cancers and
is not limited to NSCLC. Furthermore, our pathway analysis showed that
WNT/Beta-catenin, PTEN and LKB1/STK11-related pathways may affect CPI
outcomes, consistent with previous reports,[164]^32 44 46 47 further
sustaining our observations. Additionally, the BRAF/MAPK pathway has
been previously reported in preclinical and clinical settings to be of
central importance in inhibiting T cell-dependent antitumor activity,
and inhibiting BRAF/MEK concurrently with CPI has been demonstrated to
have synergistic effects in bringing about tumor
regression.[165]48,[166]50
In addition to these previously reported pathways, we identified
several resistance pathways that were not previously highlighted to
affect CPI outcomes, with the most significant pathways among these
being related to PDGF, ESR1, YAP/TAZ, EGFR, IGF, and AGTR1 signaling.
Our pathway analyses can potentially facilitate biomarker selection.
There are only a limited number of biomarkers presently available for
CPI (eg, PD-L1, TMB and mismatch repair deficiency) all sharing the
characteristic of being related to TMB/immunogenicity.[167]^2
IdentificationThis of novel biomarkers can improve patient selection
and guide better outcomes. This includes potential novel biomarkers in
pathways such as PDGF, Leptin, IGF, Estrogen, and HGF that can be
measured in either the tumor samples or potentially using non-invasive
serum samples.
Our analyses provide a tumor-genome intrinsic view, as we used the
baseline (pre-CPI treatment) tumor genetic profile to predict outcomes
of CPI therapy. The pathway analyses inform on the processes that are
altered in the tumor microenvironment, but an extended analysis of the
processes can be obtained from RNA sequencing analysis that measures
the messenger RNA expression in heterogeneous (immune, fibroblasts,
endothelial and tumor) cell populations. There are several other
aspects to consider when interpreting the data from the current
analysis. This includes the limited data set of 286 genes, variations
in tissue source (primary tumor vs metastatic site) and lines of
therapy as well as the possibility that these mutations could have been
selected at least in part due to their increased immunogenicity as
targets and not because of their functional biological
contribution.[168]^51 This latter possibility, however, is rather low,
because it would require that such mutations create a de novo epitope
presented and recognized by the immune system in all affected patients.
Furthermore, the genetic data used in pathway analysis cannot reveal
whether the pathway is inhibited or activated, which requires further
experimental studies or previously reported differentiation in a
relevant context to interpret the direction of the impact.[169]^42
Notwithstanding these caveats, it is encouraging that with these
limited sets of genes, the models have an efficiency similar to TMB and
could account for up to 65% of clinical outcomes. As more whole
exome/genome sequenced patients become available, it might be possible
to further refine our predictive model and deepen the biological
understanding of CPI outcomes with this approach.
Of note, the adjusted FDR values from multiple Fisher’s exact tests may
have underestimated the significance of some mutations or co-occurring
mutations, as they assume independence. It is accepted, however, that
mutations are not independent of each other,[170]^52 and as we
demonstrated above, some combinations of mutations can affect outcomes.
Patients can be stratified based on mutations identified in the
signatures, and these signatures could potentially be combined with TMB
and PD-L1 to create a model for patient stratification. While testing
these approaches is beyond the scope of this paper, four alternative
strategies with increased complexity can be used to facilitate the
evaluation and use of the identified signatures in clinical trial
design. First, patients can be stratified based on the four genes
(BRIP1, FLT1, FGF10, and BRAF) identified in the signatures
([171]online supplemental figure S4). Second, patients can be
stratified based on one or a combination of the pathways identified,
such as IL-6, or a combination of pathways ([172]figure 7). Third, a
predictive model based on the 36 predictive signatures ([173]figure 5)
can be used to stratify the patients. Fourth, our earlier data
demonstrated that combining PD-L1 and TMB[174]^12 can increase the
likelihood of response. Accordingly, the identified mutations,
pathways, and predictive signatures could potentially be combined with
TMB and PD-L1 to create a more advanced predictive model for patient
stratification.
There are presently no approved therapies to circumvent or reverse
resistance to CPI, which is in part due to the lack of understanding
the mechanism behind this resistance. In this report, we expand our
understanding of CPI outcomes by using a GA framework, which resulted
in the identification of multiple predictive genes, mutations and
biological pathways with several models that can predict a patient’s
outcome to anti PD-(L)1 therapy using pretreatment data. The
identification of mutations, pathways and potential biomarkers also
provide testable hypotheses for the mechanism behind CPI resistance.
Our identification of these novel targets can be used to explore
predictive biomarkers, treatment selection, and patient enrichment
strategies, all possibly leading to better outcomes in CPI-treated
patients.
Methods
Research design and patient cohorts
We used integrated FH-FMI CGDB which consists of longitudinal clinical
data derived from electronic health records curated via
technology-enabled abstraction and were linked to genomic data from the
same patient who underwent FMI comprehensive genomic profiling tests by
de-identified, deterministic matching.[175]^53 The study included 7,360
patients diagnosed with NSCLC from January 2003 to May 2021 who
underwent FoundationOne (F1) or FoundationOne CDx (F1CDx) tests (ie,
only patients with T7 or DX1 baitset data) and had treatment data. We
used rWR data[176]^15 (CR, PR, SD, PD or not evaluable (NE) on a
specified date) and rwP data (progressed or not progressed at a
specified date)[177]^16 for defining the clinical outcome groups. To
study the clinical benefit of CPI therapy and to have a more balanced
number of patients in each of these outcome group, CR, PR and SD from
the rwR data were considered as having tumor response. Progressive
disease and rwP, death or a change to a non-CPI treatment line within
the study duration was considered to be disease progression. A patient
was considered to have a “durable clinical benefit”, hereafter referred
to as “response”, if there was tumor response and no disease
progression starting 14 days after initiation of CPI to the end of the
study duration (270 days). A patient was considered to have “innate
resistance”, hereafter simply referred to as “resistance” if there was
disease progression without any tumor response during the study
duration. Acquired resistant patients, defined as having response
followed by disease progression within the 270-day study duration, were
not included. Using these definitions, 574 patients who had rwP were
assigned to the response group and 576 patients with disease
progression, as defined above, were assigned to the resistance group.
Genetic variations in these two outcome groups were then investigated
by ML methods to identify key variables and pathways that might explain
the drug response. No explicit cohort balancing of the two outcome
groups was performed. However, baseline covariates (age, race, sex,
histology, smoking history, initial disease stage) were examined and
found to have no correlation with the outcomes after FDR correction.
For determining the study duration, sensitivity analysis using periods
ranging from 120 to 365 days, in~2–3 months increments, was performed.
The study duration of 270 days resulted in an optimal balance in
patient number in each outcome group and is clinically relevant.
Disease progression within the first 14 days after CPI treatment was
not counted, since it may not reflect the effect of the current
treatment (recommendation by Flatiron Health).
To better distinguish CPI-specific predictive effects of biomarkers
from prognostic effects (independent of treatment type), we also
analyzed standard-of-care chemotherapy cohorts. We also removed
patients with early deaths (patients who died in the first 18 weeks
after treatment start) to better delineate the two effects. This is
because it has been reported that the CPI and chemotherapy survival
curves did not differentiate well until after about 18 weeks.[178]^5 12
To maximize the number of patients with NSCLC with FMI (Foundation
Medicine) targeted DNA sequencing data, we selected patients with data
from the FoundationOne (F1; T7 baitset) or FoundationOne CDx (F1CDx;
DX1 baitset) panels for solid tumors. We then focused on the 288 genes
profiled on both panels.
Treatment data
CPI analysis included monotherapies nivolumab (~31%), pembrolizumab
(~62%), atezolizumab (~5%), durvalumab (~2%). Chemotherapy patients
included patients with cytotoxic monotherapy or combinations with other
cytotoxics or targeted therapies who did not have “immunotherapy” in
the patient’s history in the database. For patients who had multiple
lines of CPI or chemotherapy, their first CPI or chemotherapy records
were used for analysis.
Tumor mutation burden
TMB (number of mutations per Mb) calculated from targeted DNA
sequencing using F1 or F1CDx panels[179]^53 on tumor biopsies from all
analyzed patients as provided by FMI. TMB data from the most recent
specimens collected before the start of treatment was used. Research
Use Only calculations based on FMI’s research algorithm used at the
time of collection were analyzed.[180]^54 The TMB-only model was
trained using logistic regression on the same train/validation patient
split used for the mutation signature models.
IL-6 analysis
Plasma IL-6 protein levels measured by Luminex multiplex assay were
from the atezolizumab phase I trial ([181]NCT01375842).[182]^55 Samples
from cycle 1 day one predose time point from patients with NSCLC were
analyzed. The survival curves were performed using the Kaplan-Meier
method.
Mutation data preparation
No explicit functional filters were applied to mutations during the ML
analysis, allowing the inclusion of all mutation types (eg, short
variants, copy number variations, and rearrangements) to
comprehensively identify genomic signatures associated with CPI
resistance or response. To ensure data quality, technical filters were
implemented. These included restricting the analysis to mutations
identified using validated bait sets (T7 or DX1) from Foundation
Medicine and excluding synonymous mutations to focus on variants with
potential functional implications. This approach enabled robust
identification of clinically relevant genomic features while minimizing
the inclusion of technical or biologically irrelevant artifacts. After
associating a CPI-resistance label to each patient, the mutations for
each CPI patient were filtered to remove synonymous mutations and then
used as three different sets of inputs in ML with different
aggregations of mutations: (1) binary input (all mutations in a gene
are aggregated into one feature per gene, ie, whether a gene is mutated
or not), (2) HFM or LFM (high/low frequency mutations; see the next
paragraph for definition) inputs (all HFM in a gene are aggregated into
one feature and LFM into another; 1–2 features per gene), and (3)
hotspot granular input (the HFM are further separated into individual
mutations). In the 1,150 patients (March 2015 to May 2021) used for
analysis in this study, there were 288 binary inputs, 460 HFM/LFM
inputs, and 528 hotspot granular inputs. The input data set was
randomly split into training and validation subsets, stratified by
CPI-resistance label, leaving 977 training patients and 173 validation
patients.
HFM/LFM input categorization was performed as follows: (1) we
calculated the total number of amino acids mutated per gene as X (2) we
calculated the frequency of mutations in each gene (ie, how many
patients had any mutation in that gene) as Y. (3) From 1 to 2 we
calculated the average amino acid mutation rate:
[MATH: average aa mutation rate
= % patients with any mutation in the g
mi>ene (Y<
/mi>)tota
mi>l number of <
mi>aa muta<
/mi>ted in<
mtext> the g
ene (X) :MATH]
HFM was assigned to any mutation that had 2× the average mutations per
that specific amino acid and had more than/equal to nine mutations in
that gene. LFM was assigned to any mutation that had lower than 2× the
average mutations per that specific amino acid and/or had less than
nine mutations. Amplifications were considered as HFM and deletions as
LFM; the rationale behind this was that LFM tend to be loss of function
and high-frequency mutations tend to be gain of function. Hotspot
granular classification was the same as in HFM/LFM, but the amino acid
mutation location to any HFM was added (eg, TP53_178, meant HFM in TP53
in amino acid 178). Any HFM that lacked the information about amino
acid location was considered an amplification mutation. To assign HFM,
LFM and hotspot mutations 4,433 patients with NSCLC from the CGDB were
used.
Pathway analysis using CBDD
The predictive genes in the genetic signatures from the ML output were
used to identify extended biological network member genes using six
different network-based algorithms (CBDD R package,[183]^56 which used
the Metabase network and pathway data (Thomson Reuters)). The
algorithms used were Network Propagation,[184]^57
Interconnectivity,[185]^58 Overconnectivity[186]^59 Hidden
nodes[187]^60 GeneMania,[188]^61 and Causal Reasoning.[189]^62 63 The
top 100 genes resulting from each algorithm were then used to run a
pathway enrichment analysis on the Metabase pathways. Each pathway then
got an enrichment p value from each of the six network algorithms.
A composite score was then calculated for each pathway using the six
enrichment p values. A common way of using p values for scoring is the
“−log[10]P” transformation, in which higher value indicates more
significance. However, the relationship between statistical and
biological significance is not linear. For example, a p value of 1e−50
(−log[10]p=50) is not 10 times more significant than 1e−5
(−log[10]p=5). They both mean a high level of significance. To more
closely capture the biological and statistical significance reflected
by p values, we performed a transformation on −log[10]P (Equation 1)
such that the final score better reflected the level of significance,
which increased and then plateaued as −log[10]P increases (Table S10
and see the interactive transformation curve on [190]this page).
[MATH: score=10x12+x x=−log
10P :MATH]
(1)
We then scaled the scores from each algorithm and calculated the
composite score per pathway using the harmonic sum of scores from the
different algorithms (Equation 2), where k is the rank of the algorithm
score per pathway.
[MATH: ∑k=1
mn>∞1k
mi> :MATH]
(2)
Genetic algorithm feature selection
Our ML approach applied Naive Bayes models with a Bernoulli prior as
the model architecture, embedded in a GA for feature selection to
reveal predictive genetic signatures.[191]^64 65 We used GA because of
the large set of available features (binary:288, HFM/LFM:460,
hotspot:528) relative to the number of patients (n=977 in the training
set), as it can efficiently find the most predictive feature
combinations (out of binary:2^288, HFM/LFM:2^460, hotspot:2^528) while
including multifeature interactions.[192]^64 65 Naive Bayes models were
chosen for the simplicity of their internal state, resistance to
overtraining, and interpretability.[193]^66 Random Forest and other
ensemble-based methods were attempted but found to require heavy
hyperparameter tuning to avoid overtraining during the GA search. The
Bernoulli prior is appropriate for binary input data and includes a
penalty term for the feature absent of mutation, differentiating it
from a multinomial prior. The details are outlined in the subsequent
paragraphs, accompanied by a schematic diagram in [194]online
supplemental figure S3. Python scripts ([195]online supplemental file
3) for implementation and performance evaluation are also provided.
In this study, we defined a GA individual as a subset of the available
input features of the data set, represented as a binary mask (mutated
or not) over all features. The fitness of each individual is calculated
based on the predictive power of a model which uses only the features
contained in the subset and is trained to predict the response to CPI
category, “response” or “resistance”.
For each GA individual, the fitness was the CV accuracy score of a
Naive Bayes model on the training set. The accuracy score was
class-balanced to avoid favoring “response” over “resistance,” or vice
versa. The CV uses stratification to keep the same fraction of
“response” and “resistance” patients in each fold. The number of folds
used was 5, a compromise to keep the number of patients in each fold
high while keeping the number of folds high enough to be confident in
the result. The training data was shuffled for each individual before
CV to avoid overfitting on CV folds during GA optimization.
In each generation of the GA, individuals were selected for mutation or
crossover using fitness proportionate selection,[196]^67 which samples
individuals probabilistically based on their fitness in order to
maintain diversity in the GA breeding pool. The breeding pool of each
generation is supplemented by a set of the highest fitness individuals
from all previous generations. Mutation occurs by randomly removing or
adding features to the subset while conserving the average number of
features. The average number of features is conserved during mutation
by partitioning the probability of mutation between adding features and
removing features in order to retain (on an average) the same number of
features removed and added. Without this partition, mutation would tend
to increase the size of models with less than 50% of the total features
used and decrease the size of models with more than 50% of the features
used, regardless of the fitness of the result. Crossover occurs by
randomly selecting each feature flag of the binary mask from two
previous individuals and does not include further correction: crossover
on average will produce offspring with the number of features midway
between the parents.
The GA procedure was run with the following parameters set. The GA was
run for 200 generations, each with a population of 1,000 individuals.
Larger populations and larger numbers of generations were not found to
produce different results, as the GA was able to find an optimum within
this time. The first generation was generated randomly such that on
average, 10% of the features were included in each individual. This
fraction was chosen to correspond roughly to the number of features at
the end of the GA. During mutation, on average, 1% of features were
removed or added to an individual. The top 200 individuals over all
generations were added to the breeding pool for each generation for a
total breeding pool size of 1,200 in each generation after the first.
To generate each successive generation, 600 individuals were formed by
mutating an individual from the breeding pool while the remaining 400
of each generation were formed by crossover of two previous
individuals, a relative fraction chosen to slightly favor mutation in
order to increase diversity in the population.
The GA was run 10 times for each mutational input categorization
(binary per-gene, gain or loss of function, hotspot). Since each run of
the GA tended to find separate local optima, these 10 runs along with
the clustering technique described below were used to identify multiple
local optima that were too distant for a single GA run to identify.
After 10 runs of 200 generations with a population size of 1,000 there
were 2,000,000 GA individuals which are available. The top 5% (100,000)
of all individuals, based on their CV score, were selected and then
clustered according to the similarity of the features they contain. The
clustering was done using a simple k-means clustering with 12 clusters
to account for the expected 10 separate local optima (one from each run
of the GA) plus some leeway for outliers.
In each cluster, the set of features that appeared in more than 50% of
cluster members was considered the characteristic set of features for
that cluster. A final model was then trained on each of the 12
characteristic sets and evaluated on the validation set.
Feature importance calculation
In order to visualize the importance of individual mutations on the
prediction outcomes, the internal state of the 36 Bernoulli Naive Bayes
models were converted into their equivalent linear (logistic
regression) coefficients^[197]68 . This conversion for each gene
mutation feature is outlined below.
[MATH: coefresp=lnPmut|r
esp1<
mo>-Pmut|r
esp
mfenced> :MATH]
[MATH: coefresis
t=lnPmut|r
esist1-Pmut|r
esist :MATH]
[MATH:
coef=co<
mi>efresp-coef(
resist)
:MATH]
Where “P(mut│resp)” is the probability of mutation for the response
group, “P(mut│resist)” is the probability of mutation for the
resistance group. “Coef(resp)” is the linear coefficient for the
feature in the response group, while “coef(resist)” is the linear
coefficient for the feature in the resistance group. “coef” is the
final linear coefficient of the feature. “coef” > 0 indicates
association of the feature with response while “coef” < 0 indicates
association with resistance. The sum of coefficients of all the
features determines the prediction of response or resistance for a
patient.
When
[MATH: ∑mut
coef>
thresho<
mi>ld :MATH]
, response is predicted over resistance.
Where the threshold is the intercept of the model, which is a trained
parameter of the model in addition to the coefficients.
Mutation co-occurrence analysis
To assess potential relationships between co-occurrence of mutation
pairs/triples and CPI response, Fisher’s exact tests were performed,
unless there was zero count(s) in the 2×2 contingency table, then the
Mantel-Haenszel method with continuity correction of 0.5 was used.
Multiple testing adjustment of p values used the false discovery rate
method. The contingency table structure for the number of patients with
mutation pairs/triples is as below:
Response Resistance
All gene mutation features mutated
Not all mutated (any or none mutated)
[198]Open in a new tab
Each row/column sum of the above table had to be at least 5 to remove
very rare mutational combinations.
supplementary material
online supplemental file 1
[199]jitc-13-3-s001.docx^ (562.5KB, docx)
DOI: 10.1136/jitc-2024-009092
online supplemental file 2
[200]jitc-13-3-s002.pdf^ (180.8KB, pdf)
DOI: 10.1136/jitc-2024-009092
online supplemental file 3
[201]jitc-13-3-s003.zip^ (11.9KB, zip)
DOI: 10.1136/jitc-2024-009092
online supplemental table 1
[202]jitc-13-3-s004.xlsx^ (1.6MB, xlsx)
DOI: 10.1136/jitc-2024-009092
Acknowledgements