computational health informatics program, boston children ... · associated with improved...

48
1 Neoantigen directed immune escape in lung cancer evolution Rachel Rosenthal 1,2,3 , Elizabeth Larose Cadieux* 4 , Roberto Salgado* 5,6 , Maise Al Bakir* 3 , David A. Moore* 7 , Crispin T. Hiley* 1,3 , Tom Lund* 8 , Miljana Tanić 9 , James L. Reading 8,10 , Kroopa Joshi 8 , Jake Y. Henry 8,10 , Ehsan Ghorani 8,10 , Gareth A. Wilson 1,3 , Nicolai J. Birkbak 1,3 , Mariam Jamal-Hanjani 1 , Selvaraju Veeriah 1 , Zoltan Szallasi 11,12 , Sherene Loi 5 , Matthew D. Hellmann 13,14 , Andrew Feber 15 , Benny Chain 16,17 , Javier Herrero 2 , Sergio Quezada 8,9 , Jonas Demeulemeester 4,18 , Peter Van Loo 4,17 , Stephan Beck 9 , Nicholas McGranahan 1,19# and Charles Swanton 1,3# , on behalf of the TRACERx consortium. *equal contribution #Joint corresponding authors Affiliations 1. Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6BT, United Kingdom 2. Bill Lyons Informatics Centre, University College London Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6BT, United Kingdom 3. Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, 1 Midland Rd, London, NW1 1AT, United Kingdom 4. Cancer Genomics Laboratory. The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK. 5. Department of Pathology, GZA-ZNA, Antwerp, Belgium 6. Division of Research, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia 7. Department of Pathology, UCL Cancer Institute, London, UK 8. Cancer Immunology Unit, University College London Cancer Institute, University College London, London, UK 9. Department of Cancer Biology, UCL Cancer Institute, University College London, London, UK. 10. Research Department of Haematology, University College London Cancer Institute, University College London, London, UK 11. Computational Health Informatics Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA 12. Danish Cancer Society Research Center, Copenhagen, Denmark 13. Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA 14. Weill Cornell School of Medicine, New York, NY, USA 15. Division of Surgery and Interventional Science, University College London, London WC1E 6BT, UK. 16. Division of Infection and Immunity, UCL, London, UK 17. Department of Computer Sciences, UCL, London, UK 18. Department of Human Genetics, University of Leuven, Herestraat 49, B-3000, Leuven, Belgium. 19. Cancer Genome Evolution Research Group, University College London Cancer Institute, University College London, London, UK

Upload: others

Post on 17-Jun-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

1

Neoantigen directed immune escape in lung cancer evolution

Rachel Rosenthal1,2,3, Elizabeth Larose Cadieux*4, Roberto Salgado*5,6, Maise Al Bakir*3, David A. Moore*7, Crispin T. Hiley*1,3, Tom Lund*8, Miljana Tanić9, James L. Reading8,10, Kroopa Joshi8, Jake Y. Henry8,10, Ehsan Ghorani8,10, Gareth A. Wilson1,3, Nicolai J. Birkbak1,3, Mariam Jamal-Hanjani1, Selvaraju Veeriah1, Zoltan Szallasi11,12, Sherene Loi5, Matthew D. Hellmann13,14, Andrew Feber15, Benny Chain16,17, Javier Herrero2, Sergio Quezada8,9, Jonas Demeulemeester4,18, Peter Van Loo4,17, Stephan Beck9, Nicholas McGranahan1,19# and Charles Swanton1,3#, on behalf of the TRACERx consortium.

*equal contribution

#Joint corresponding authors Affiliations 1. Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6BT, United Kingdom 2. Bill Lyons Informatics Centre, University College London Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6BT, United Kingdom 3. Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, 1 Midland Rd, London, NW1 1AT, United Kingdom 4. Cancer Genomics Laboratory. The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK. 5. Department of Pathology, GZA-ZNA, Antwerp, Belgium 6. Division of Research, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia 7. Department of Pathology, UCL Cancer Institute, London, UK 8. Cancer Immunology Unit, University College London Cancer Institute, University College London, London, UK 9. Department of Cancer Biology, UCL Cancer Institute, University College London,

London, UK.

10. Research Department of Haematology, University College London Cancer Institute, University College London, London, UK 11. Computational Health Informatics Program, Boston Children’s Hospital, Harvard

Medical School, Boston, MA, USA

12. Danish Cancer Society Research Center, Copenhagen, Denmark

13. Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY,

USA

14. Weill Cornell School of Medicine, New York, NY, USA

15. Division of Surgery and Interventional Science, University College London, London

WC1E 6BT, UK.

16. Division of Infection and Immunity, UCL, London, UK

17. Department of Computer Sciences, UCL, London, UK

18. Department of Human Genetics, University of Leuven, Herestraat 49, B-3000, Leuven, Belgium. 19. Cancer Genome Evolution Research Group, University College London Cancer

Institute, University College London, London, UK

Page 2: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

2

Abstract

The interplay between an evolving cancer and the dynamic immune-microenvironment

remains unclear. Here, we analyze 258 regions from 88 early-stage untreated non-small cell

lung cancers (NSCLCs) using RNAseq and pathology tumor infiltrating lymphocyte estimates.

The immune-microenvironment was variable both between and within patients’ tumors.

Diverse immune selection pressures were associated with different mechanisms of

neoantigen presentation dysfunction restricted to distinct microenvironments. Sparsely

infiltrated tumors exhibited evidence for historical immunoediting, with a waning of neoantigen-

editing during tumor evolution, or copy number loss of historically clonal neoantigens.

Immune-infiltrated tumor regions exhibited ongoing immunoediting, with either HLA LOH or

depletion of expressed neoantigens. Promoter hypermethylation of genes harboring

neoantigens was identified as an epigenetic mechanism of immunoediting. Our results

suggest the immune-microenvironment exerts a strong selection pressure in early stage,

untreated NSCLCs, producing multiple routes to immune evasion, which are clinically relevant,

forecasting poor disease-free survival in multivariate analysis.

Introduction

Anti-tumor immune responses require the functional presentation of tumor antigens and a

microenvironment replete with competent immune effectors 1,2. However, the extent to which

an active immune system sculpts tumor genome evolution has not been well characterized.

Although associations between immune infiltration and tumor clonal diversity have been

observed in certain contexts 3,4, whether the immune system acts as a dominant selective

force in early stage untreated cancer is unclear. Furthermore, transcriptomic heterogeneity

might confound conclusions drawn from sampling a single tumor sample, leading to inaccurate

interpretations of mechanisms of immune evasion.

To determine immune infiltration in untreated NSCLC, assess how it varies between and within

tumors, and characterize immune evasion mechanisms and their associations with clinical

Page 3: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

3

outcome, we integrated 164 RNAseq samples from 64 tumors and 234 tumor infiltrating

lymphocyte (TIL) pathological estimates from 83 tumors for a combined cohort of 258 tumor

regions from 88 prospectively acquired tumors within the TRACERx 100 cohort 5. We explore

how selection pressures from a diverse tumor microenvironment impact upon neoantigen

presentation, as well as the tumor-specific mechanisms leading to immune escape, and their

clinical impact.

Results

Heterogeneity of immune infiltration

To estimate immune infiltration in the multi-region NSCLC TRACERx RNAseq cohort, we

benchmarked published in silico immune deconvolution tools (Methods). Compared to other

transcriptomic approaches 6-11, the Danaher immune signature optimally estimated immune

infiltrates in NSCLC (Extended Data Fig. 1).

Using this approach, RNAseq-derived infiltrating immune cell populations were estimated for

the 164 tumor regions from 64 TRACERx 100 cohort patients 5, for which there was RNA of

sufficient quality (Extended Data Fig. 2A-B, Table S1).

A wide range of immune-infiltration was observed between and within histologies (Extended

Data Fig. 3), as well as between separate regions from the same tumor. Unsupervised

hierarchical clustering revealed two distinct immune clusters, corresponding to high and low

levels of immune infiltration, for each histology. Individual tumor regions were stratified as

either having high or low immune infiltrate (Figure 1).

Validating our clustering approach, immune-high tumor regions contained greater pathology

estimates of TIL infiltrate compared to immune-low regions (p=3e-05) (Extended Data Fig.

4A). Due to the strong correlation observed with pathology TIL estimates (Extended Data Fig.

1E), we also used pathology estimated TILs to group tumor regions without RNAseq

(Extended Data Fig. 4B-C, Methods). The predicted abundance of myeloid-derived

Page 4: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

4

suppressor cells and tumor associated M2 macrophages 12 negatively correlated with the

immune activating cell subsets (Extended Data Fig. 4D-E), indicating that immunosuppressive

cells may influence the immune microenvironment. A small number (11%) of mostly lung

adenocarcinoma cases had pathology TIL estimates that were not reflected by the assigned

immune cluster potentially reflecting heterogeneity of sampling due to variation from the

mirrored tissue samples used to score TILs and extract RNA.

Overall, while 63 patients had tumors with consistently low (38 tumors, 43%) or high (25

tumors, 31%) immune infiltration, 25 patients had tumors with disparate immune infiltration

between regions (31%) (Extended Data Fig. 4C). Intratumor heterogeneity was also found to

confound genomic and transcriptomic biomarkers for the prediction of response to immune

checkpoint blockade. For example, the classifier “TIDE” 12 was heterogeneous in 17/42 tumors

(Extended Data Fig. 5A) and heterogeneously infiltrated tumors from our analysis tended to

exhibit a heterogeneous TIDE signature (p=0.05) (Extended Data Fig. 5A). Likewise, a

transcriptomic signature predicting innate resistance to PD-1 immune checkpoint blockade

(IPRES) 13 and an IFN-signaling score 14 were also heterogeneous (Extended Data Fig. 5B-

D).

In a recent prospective study, high tumor mutation burden (TMB) (>10 mutations/megabase)

associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB

had at least one tumor region containing a low TMB (Extended Data Fig. 5E). Heterogeneously

infiltrated tumors were also more likely to exhibit heterogeneous TMB (p=7e-04) (Extended

Data Fig. 5F). Among tumors with heterogeneous TMB, the regions with low TMB had

significantly lower tumor purity than regions with high TMB, indicating the importance of

considering tumor stromal content as a confounding factor (paired t-test p=0.04) (Extended

Data Fig. 4F).

Page 5: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

5

Interaction between immune infiltration and tumor evolution

To explore the relationship between tumor genomic features and the immune

microenvironment, a distance measure in both genomic and immune space was calculated

for all pairwise combinations of tumor regions from the same tumor (Methods). We observed

a significant correlation between the two pairwise distance measures (Figure 2A; lung adeno.:

p=3.5e-04, lung squam.: p=2e-03). A similar relationship was observed when the pairwise

immune and copy number alteration distance was compared, reaching statistical significance

among the lung adenocarcinoma cohort (Extended Data Fig. 6A). These results support an

interplay between the immune and cancer genomic landscape.

To further explore this interplay, we considered the relationship between the clonal structure

of each tumor region and its immune infiltrate. RNAseq-estimated CD8+ T-cell infiltration was

compared to the within region subclonal diversity (Shannon entropy; Methods). A significant

negative correlation was observed in lung adenocarcinoma but not squamous cell carcinoma;

regions with high CD8+ T-cell infiltration had lower subclonal diversity (lung adeno.: p=0.035,

rho=-0.22; lung squam.: p=0.91, rho=-0.02) (Extended Data Fig. 6B-C). Lung adenocarcinoma

regions from tumors with consistently low levels of immune infiltration exhibited greater

subclonal diversity compared to those from tumors with high or heterogeneous immune

infiltration (Figure 2B-C; lung adeno.: p=0.01). When pathology estimated TILs (which did not

correlate with tumor purity; Extended Data Fig. 6D) were used to stratify patients, a reduction

in tumor diversity was again observed in regions with high/heterogeneous TIL (Extended Data

Fig. 6E; p=0.02).

Immune editing in response to an active immune microenvironment

If T-cell mediated immune surveillance of neoantigens influences cancer genome evolution,

one would predict to observe evidence for neoantigen depletion in tumors and/or disruption to

antigen presenting machinery 16. Conceivably, neoantigen depletion may occur at the DNA

level through events such as copy number loss, at the RNA level through suppression of

Page 6: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

6

transcripts harboring neoantigens, at the epigenetic level through silencing of the genomic

segments encoding neoantigens, or through post-translational mechanisms. Alternatively,

tumor subclones expressing neoantigens may be preferentially eliminated by the immune

system resulting in purifying selection of subclones harboring them.

To investigate neoantigen depletion, we predicted neoantigens and their clonal status.

Neoantigens were peptides with a predicted binding affinity <500nM or rank percentage score

<2% and strong neoantigens had a predicted binding affinity <50nM or rank percentage score

<0.5% 17 (Methods). We used a published method to quantify the extent of immunoediting in

each tumor sample 16. This method compares the observed to expected number of

neoantigens present in a tumor, such that a score <1 suggests immunoediting has occurred.

While no significant difference in observed/expected neoantigen occurred between lung

adenocarcinomas and lung squamous cell carcinomas (Extended Data Fig. 6F), we noted this

score depends on the number of patient germline heterozygous HLA alleles (p=2.1e-05,

rho=0.43) (Extended Data Fig. 6G) since fewer unique HLA types will decrease the number

of observed neoantigens. To mitigate this, we investigated whether this measure changed

during tumor evolution, from clonal to subclonal events within each tumor. Among low infiltrate

tumors, a decrease in immunoediting (increase in observed/expected neoantigens) was noted

from clonal to subclonal mutations (p=8.8e-03, paired t-test) (Figure 2D), possibly reflecting

an ancestral immune-active microenvironment which has subsequently become cold.

Neoantigen depletion may also occur at the DNA level through copy number loss (Figure 2E)

18. Across this cohort, 43/88 tumors showed evidence for >1 historically clonal neoantigen

being subclonally lost due to subclonal copy number events (Figure 2F; range 0-42% clonal

neoantigens).

To determine if the elimination of historically clonal neoantigens through copy number loss

occurred more frequently than expected by chance, we compared neoantigens with non-

neoantigenic non-synonymous mutations. In tumor regions with low immune infiltration non-

Page 7: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

7

synonymous mutations predicted to be neoantigens, were more likely to occur on genomic

segments subject to subclonal copy number loss as compared to their non-neoantigenic

counterparts (p=1.2e-04) (Figure 2G). In low infiltration tumors, reduced immunoediting of

subclones was observed more frequently in tumors without evidence of neoantigen copy-

number loss, supporting its role in subclonal immunoediting (p=0.88 vs. p=2.2e-04) (Figure

2H).

Repression of neoantigenic transcripts

To investigate alternative neoantigen depletion mechanisms, we determined whether each

neoantigen was identified at the transcript-level. Overall only 33% of clonal neoantigens were

expressed in every tumor region and a significantly lower proportion of ubiquitously expressed

clonal neoantigens among immune high (median: 29%) or heterogeneous (median: 35%)

tumors as compared to immune low (median: 41%) tumors was observed (Figure 3A-B) (p=1e-

02). To further investigate if down-regulation of neoantigenic transcripts reflects selection

pressure, we considered whether neoantigens were preferentially subject to reduction in

expression compared to non-neoantigens, an approach not confounded by the influence of

tumor purity.

Among tumors with intact HLA alleles, significant reduction of expressed neoantigens

compared to non-neoantigenic non-synonymous mutations was observed (Figure 3C;

p=0.01). Moreover, when tumors were divided by immune classification, only immune high

and heterogeneous tumors with intact HLA alleles showed depletion of expressed

neoantigens, suggesting that subclones in immune infiltrated tumors may be selected for, by

virtue of immune evasion through either HLA LOH or through repression of neoantigen

expression (Figure 3C). Diminished neoantigen expression among immune-high tumors

without HLA LOH was more pronounced when the more stringent definition of strongly binding

neoantigens was used (Extended Data Fig. 6H).

Page 8: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

8

We explored two potential mechanisms for neoantigen expression downregulation: negative

selection of clones harboring the expressed neoantigens, and epigenetic downregulation

through promoter hypermethylation. We observed an enrichment of neoantigens in genes that

were lowly expressed in the tumor sample (<= 1TPM) as compared to non-synonymous non-

neoantigens (p=5.5e-10, OR=1.3) (Extended Data Fig. 6I). This enrichment was stronger

when we only considered strong neoantigens (p=6.8e-13, OR=1.4) (Extended Data Fig. 6I).

Neoantigens identified in TRACERx were also less likely to occur in genes that were

consistently expressed across 1019 NSCLC samples from TCGA (Figure 3D) compared to

non-synonymous predicted non-neoantigens. While the generation of neoantigenic mutations

in genes consistently expressed in TCGA was most reduced among tumors with high immune

infiltration (p=2.1e-04, OR=0.77), we also observed this reduction among heterogeneous and

low infiltrated tumors (p=1.8e-03, OR=0.82 & p=4.4e-02, OR=0.88, respectively). This is

consistent with low-immune tumors once being subject to the selective pressures of an active

immune microenvironment (Figure 3D).

To investigate methylation status of neoantigens, we performed multi-region reduced-

representation bisulfite sequencing on 79 out of the 164 samples (28/64 patients) in the

TRACERx RNAseq cohort in addition to the adjacent normal (Figure 3E, Table S2). Among

genes harboring neoantigens, an 11.4-fold increase in promoter hypermethylation was

observed for genes that were not expressed compared to those genes that were expressed

(χ2-test, p=1.6e-04) (Figure 3F). To determine if the observed down-regulation was

neoantigen-specific, promoter hypermethylation was further compared between all

neoantigens and the same genes which did not carry the neoantigen in purity/ploidy-matched

samples. Overall, non-expressed neoantigens were more likely to exhibit promoter

hypermethylation than the same genes without a neoantigen (χ2-test, p=4.5e-02, OR=2.3)

(Figure 3G, Table S3). Among expressed neoantigens, no difference in promoter

hypermethylation state was observed when compared to purity/ploidy-matched samples (χ2-

test, p=6.7e-01, OR=0.48) (Figure 3H, Table S4). These findings suggest that immune

Page 9: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

9

pressures may select for promoter hypermethylation and neoantigen silencing in evolving

subclones.

Pervasive disruption to antigen presentation

Defects in antigen presentation that interrupt tumor antigen recognition 19,20 may provide

another immune evasion mechanism. To understand the importance of these avenues of

immune escape in the treatment-naive setting, we mapped their occurrence, region by region

(Figure 4A-B, Extended Data Fig. 7A; Methods).

Disruption to antigen presentation, through HLA LOH or through mutations affecting MHC

stability, the HLA enhanceosome, and peptide generation were frequently observed in both

lung histologies (56% of lung adenocarcinomas and 78% of lung squamous cell carcinomas).

HLA LOH and alterations affecting other components of the antigen presentation machinery,

including B2M mutations, had a tendency for mutually exclusivity (lung adeno.: p=9.3e-04;

lung squam.: p=1.5e-02), supporting antigen presentation dysfunction as a potent immune

escape mechanism. Moreover, consistent with prior findings 20, highly infiltrated lung

adenocarcinoma tumor regions were prone to exhibit HLA LOH (OR=2.4, p=3e-03).

Loss of HLA-C in particular may result in loss of the killer-cell immunoglobulin-like receptor

(KIR) signal that inhibits elimination through NK cell activity 21. There are two groups of HLA-

C alleles, HLA-C1 and HLA-C2, each with different KIR specificity 22. Thus, tumor cells from

heterozygous patients (HLA-C1 and HLA-C2) would be expected to be targeted for NK cell-

mediated elimination following loss of either HLA-C allele (Extended Data Fig. 7B).

Conversely, patients with homozygous HLA-C alleles may avoid NK cell-mediated elimination.

Consistent with this, NK cell infiltration was increased among heterozygous HLA-C1/C2 tumor

regions with HLA-C LOH (p=6.2e-07) (Extended Data Fig. 7C). Increased NK cell infiltration

was not observed among tumors without HLA-C LOH (p=0.12), suggesting that this change

in the tumor microenvironment results from loss of the HLA-C inhibitory “self” signal.

Page 10: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

10

Immune evasion capacity is prognostic in NSCLC

Finally, we examined whether combining estimates of immune infiltration and tumor immune

evasion potential could provide prognostic power. Tumors were classified as exhibiting low

evasion capacity (homogeneously high immune infiltration or no evidence of immune evasion

[DNA immunoediting score > 1 and no antigen presentation disruption]) or high evasion

capacity (at least one region with low immune infiltration as well as defective antigen

presentation or DNA immunoediting score < 1). Patients whose tumors had a low immune

evasion capacity, had significantly longer disease-free survival times (p=9.0e-04) (Figure 4C).

To explore these results in the context of our prior findings relating to the importance of clonal

neoantigens 23, we also grouped patients into those harboring high or low clonal neoantigen

burden using the previously defined threshold (upper quartile of the cohort) 23. Validating

previous results, high clonal neoantigen burden was associated with improved disease-free

survival among both lung adenocarcinoma and lung squamous cell carcinoma (lung adeno.:

p=2.2e-02; lung squam.: p=2.5e-02) (Extended Data Fig. 8A). The association observed

between clonal neoantigens and disease-free survival was not dependent on the specific

threshold used (Extended Data Fig. 8B) and clonal neoantigen burden remained significant in

a multivariate model with stage, histology, age, gender, pack years, and adjuvant therapy

(p=0.02). Conversely, no significant relationship between subclonal neoantigen burden, nor

total neoantigen burden, and disease-free survival was observed (Extended Data Fig. 8C-E).

However, intriguingly, when we focused on tumors with a low clonal neoantigen load, the

immune evasion capacity of a tumor was still prognostic (p=5.3e-03), indicating that in the

absence of immune evasion, even a low clonal neoantigen burden may be sufficient to elicit

an effective immune response (Figure 4D).

Furthermore, we observed that tumors with either a high clonal neoantigen load or low immune

evasion capacity exhibited significantly improved disease-free survival times (p=4.9e-06)

(Figure 4E). This association remained significant in a multivariate model with stage, histology,

Page 11: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

11

age, gender, pack years, and adjuvant therapy (p<0.001) (Extended Data Fig. 8F). These data

suggest that considering the many facets of the interaction between the tumor and immune

microenvironment is important for predicting clinical outcome.

Discussion

To capture the complex interplay between cancer genomic evolution and anti-tumor immunity

in lung cancer, we integrated genomic, transcriptomic, epigenomic, and pathologic data to

define how tumors are sculpted by the immune microenvironment, what mechanisms of

immune escape influence tumor evolution, and the clinical impact of active tumor-immune

interaction. Our results suggest the immune microenvironment is highly variable between

patients but also markedly different between distinct regions of the same tumor, with nearly a

third of tumors exhibiting diverse immune infiltration.

Our results show evidence of tumor evolution shaped through different immunoediting

mechanisms, either affecting antigen presentation or neoantigenic mutations themselves at

both the DNA and RNA-level.

Consistent with disruption to antigen presentation machinery being subject to strong positive

selection 24, we found HLA LOH tended towards mutually exclusivity with other forms of

antigen presentation disruption, such as mutations affecting MHC stability, the HLA

enhanceosome, or peptide generation. At the DNA level, sparsely infiltrated tumors showed

enrichment for the elimination of clonal neoantigens, indicating the importance of

chromosomal instability driving neoantigen loss.

As a whole, tumors exhibited fewer neoantigens in expressed genes than expected, potentially

reflecting historical purifying selection of neoantigens. High-immune tumors with intact HLA

alleles also displayed transcriptomic neoantigen depletion, suggesting that these tumors may

evade immune predation either through HLA LOH or by suppressing neoantigen expression,

but seldom both. Promoter hypermethylation was identified as a potential mechanism of

Page 12: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

12

transcriptomic neoantigen depletion, leading to the preferential repression of genes harboring

neoantigenic mutations. Promoter hypermethylation affected neoantigen expression level in

~23% of the neoantigens studied, indicating that additional mechanisms of neoantigen

transcription repression require elucidation.

Through the combination of immune microenvironment and tumor immune escape factors we

defined an estimate of each tumor’s immune evasion capacity, which associated with poorer

outcome. As TRACERx is a prospective study of early stage untreated NSCLC, it will be

important to validate these findings in the extended longitudinal cohort as the study matures.

The observation that clonal neoantigens can be subject to copy number loss and transcript

repression, even in untreated early stage disease, may have important implications for

predicting response and resistance to immune checkpoint blockade. Relapse samples

following checkpoint blockade therapy have been shown to eliminate clonal neoantigens,

reshaping the TCR repertoire of those samples 18. Clonal neoantigens occurring in expressed

genes which are required for lung cancer cell fitness may make ideal targets for vaccine or

adoptive cell therapies.

The extent to which neoantigen transcript depletion is dynamic in response to therapy and

tumor dissemination and whether such phenomena may be harnessed to improve

immunotherapy response is unknown. Epigenetic immune evasion supports the potential for

epigenetic modulatory agents, in combination with immunotherapy, to restore or improve

tumor immunogenicity 25. One possibility is that epigenetic repression of a neoantigen in a

lung cancer expressed gene may result at a fitness cost. This may shed light on recent

phenomenon observed in some patients with acquired resistance to checkpoint inhibitor

therapy, who are subsequently re-challenged with the same drug and respond a second time

26.

Taken together, our results suggest early stage, untreated NSCLCs are frequently

characterized by multiple independent mechanisms of immune evasion within individual

Page 13: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

13

tumors, emphasizing the strong selection pressures that the immune system imposes upon

tumor evolution. Our results suggest that the beneficial role of successful immune

surveillance, and the diversity of immune evasion mechanisms should be considered and

harnessed in therapeutic interventions.

Acknowledgments

We thank the members of the TRACERx consortium for participating in this study. C.S is Royal

Society Napier Research Professor. C.S is supported by the Francis Crick Institute

(FC001169), the Medical Research Council (FC001169 ), and the Wellcome Trust (FC001169

); by the UK Medical Research Council (grant reference MR/FC001169 /1); C.S. is funded by

Cancer Research UK (TRACERx and CRUK Cancer Immunotherapy Catalyst Network), the

CRUK Lung Cancer Centre of Excellence, Stand Up 2 Cancer (SU2C), the Rosetrees and

Stoneygate Trusts, NovoNordisk Foundation (ID 16584), the Breast Cancer Research

Foundation (BCRF), the European Research Council Consolidator Grant (FP7-THESEUS-

617844), European Commission ITN (FP7-PloidyNet-607722), Chromavision – this project

has received funding from the European Union’s Horizon 2020 research and innovation

programme under grant agreement No 665233, National Institute for Health Research, the

University College London Hospitals Biomedical Research Centre, and the Cancer Research

UK University College London Experimental Cancer Medicine Centre. N.M is a Sir Henry Dale

Fellow, jointly funded by the Wellcome Trust and the Royal Society (Grant Number

211179/Z/18/Z), and also receives funding from CRUK Lung Cancer Centre of Excellence,

Rosetrees, and the NIHR BRC at University College London Hospitals. P.V.L. is a Winton

Group Leader in recognition of the Winton Charitable Foundation’s support towards the

establishment of The Francis Crick Institute. J.D. is a postdoctoral fellow of the Research

Foundation - Flanders (FWO). S.A.Q is funded by a CRUK Senior Cancer Research

Fellowship (C36463/A22246), a CRUK Biotherapeutic Program Grant (C36463/A20764), and

Rosetrees. The TRACERx study (Clinicaltrials.gov no: NCT01888601) is sponsored by

University College London (UCL/12/0279) and has been approved by an independent

Page 14: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

14

Research Ethics Committee (13/LO/1546). TRACERx is funded by Cancer Research UK

(C11496/A17786) and coordinated through the Cancer Research UK and UCL Cancer Trials

Centre. For the RRBS methylation data, we acknowledge technical support from the CRUK-

UCL Centre-funded Genomics and Genome Engineering Core Facility of the UCL Cancer

Institute and grant support from the NIHR-BRC (BRC275/CN/SB/101330). The results

published here are in part based upon data generated by The Cancer Genome Atlas pilot

project established by the NCI and the National Human Genome Research Institute. The data

were retrieved through database of Genotypes and Phenotypes (dbGaP) authorization

(Accession No. phs000178.v9.p8). Information about TCGA and the investigators and

institutions who constitute the TCGA research network can be found at

http://cancergenome.nih.gov/.

Author Contributions

R.R. created the bioinformatics analysis pipeline and wrote the manuscript. R.S., M.A.B,

D.A.M, C.T.H, and T.L jointly analyzed pathology TIL estimates. J.L.R., J.Y.H., and E.G.

performed flow cytometry experiments for validating immune signatures. K.J. performed

TCRseq experiments for validating immune signatures. S.V. performed sample preparation

and RNA extraction. E.L-C., J.D, A.F, G.A.W, and M.T generated and analyzed RRBS data.

E.L.C and J.D performed DNA methylation analyses and neoantigen methylation analyses,

under supervision of S.B. and P.V.L. N.J.B. gave immune signatures advice, conducted

analyses of multiregion sequencing exome data, and reviewed the manuscript. M.J-H.

designed study protocols and advised the clinical understanding of patients. Z.S., S.L, and

M.D.H. helped direct avenues of bioinformatics and pathology TIL analysis. B.C, J.H., and

S.A.Q. provided data analysis support and supervision. N.M. and C.S. jointly supervised the

study and helped write the manuscript.

Author Information

Page 15: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

15

Reprints and permissions information is available at www.nature.com/reprints. The authors

declare competing financial interests: C.S. receives grant support from Pfizer, AstraZeneca,

BMS, and Ventana. C.S. has consulted for Boehringer Ingelheim, Eli Lily, Servier, Novartis,

Roche-Genentech, GlaxoSmithKline, Pfizer, BMS, Celgene, AstraZeneca, Illumina, and

Sarah Cannon Research Institute. C.S. is a shareholder of Apogen Biotechnologies, Epic

Bioscience, GRAIL, and has stock options and is co-founder of Achilles Therapeutics. S.A.Q.

is a co-founder of Achilles Therapeutics. R.R., N.M., and G.A.W. have stock options and have

consulted for Achilles Therapeutics. Correspondence and material requests should be

addressed to C.S. ([email protected]) and N.M.

([email protected]).

Page 16: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

16

References

1 Galon, J. et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science (New York, N.Y 313, 1960-1964, doi:10.1126/science.1129139 (2006).

2 Charoentong, P. et al. Pan-cancer Immunogenomic Analyses Reveal Genotype-Immunophenotype Relationships and Predictors of Response to Checkpoint Blockade. Cell reports 18, 248-262, doi:10.1016/j.celrep.2016.12.019 (2017).

3 Zhang, A. W. et al. Interfaces of Malignant and Immunologic Clonal Dynamics in Ovarian Cancer. Cell 173, 1755-1769 e1722, doi:10.1016/j.cell.2018.03.073 (2018).

4 Milo, I. et al. The immune system profoundly restricts intratumor genetic heterogeneity. Sci Immunol 3, doi:10.1126/sciimmunol.aat1435 (2018).

5 Jamal-Hanjani, M. et al. Tracking the Evolution of Non-Small-Cell Lung Cancer. The New England journal of medicine 376, 2109-2121, doi:10.1056/NEJMoa1616288 (2017).

6 Davoli, T., Uno, H., Wooten, E. C. & Elledge, S. J. Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy. Science (New York, N.Y 355, doi:10.1126/science.aaf8399 (2017).

7 Racle, J., de Jonge, K., Baumgaertner, P., Speiser, D. E. & Gfeller, D. Simultaneous enumeration of cancer and immune cell types from bulk tumor gene expression data. eLife 6, doi:10.7554/eLife.26476 (2017).

8 Li, B. et al. Comprehensive analyses of tumor immunity: implications for cancer immunotherapy. Genome biology 17, 174, doi:10.1186/s13059-016-1028-7 (2016).

9 Newman, A. M. et al. Robust enumeration of cell subsets from tissue expression profiles. Nat Methods 12, 453-457, doi:10.1038/nmeth.3337 (2015).

10 Hendry, S. et al. Assessing Tumor-Infiltrating Lymphocytes in Solid Tumors: A Practical Review for Pathologists and Proposal for a Standardized Method from the International Immuno-Oncology Biomarkers Working Group: Part 2: TILs in Melanoma, Gastrointestinal Tract Carcinomas, Non-Small Cell Lung Carcinoma and Mesothelioma, Endometrial and Ovarian Carcinomas, Squamous Cell Carcinoma of the Head and Neck, Genitourinary Carcinomas, and Primary Brain Tumors. Adv Anat Pathol 24, 311-335, doi:10.1097/PAP.0000000000000161 (2017).

11 Aran, D., Hu, Z. & Butte, A. J. xCell: digitally portraying the tissue cellular heterogeneity landscape. Genome biology 18, 220, doi:10.1186/s13059-017-1349-1 (2017).

12 Jiang, P. et al. Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response. Nature medicine 24, 1550-1558, doi:10.1038/s41591-018-0136-1 (2018).

13 Hugo, W. et al. Genomic and Transcriptomic Features of Response to Anti-PD-1 Therapy in Metastatic Melanoma. Cell 165, 35-44, doi:10.1016/j.cell.2016.02.065 (2016).

14 Ayers, M. et al. IFN-gamma-related mRNA profile predicts clinical response to PD-1 blockade. The Journal of clinical investigation 127, 2930-2940, doi:10.1172/JCI91190 (2017).

Page 17: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

17

15 Hellmann, M. D. et al. Tumor Mutational Burden and Efficacy of Nivolumab Monotherapy and in Combination with Ipilimumab in Small-Cell Lung Cancer. Cancer cell 33, 853-861 e854, doi:10.1016/j.ccell.2018.04.001 (2018).

16 Rooney, M. S., Shukla, S. A., Wu, C. J., Getz, G. & Hacohen, N. Molecular and genetic properties of tumors associated with local immune cytolytic activity. Cell 160, 48-61, doi:10.1016/j.cell.2014.12.033 (2015).

17 Hoof, I. et al. NetMHCpan, a method for MHC class I binding prediction beyond humans. Immunogenetics 61, 1-13, doi:10.1007/s00251-008-0341-z (2009).

18 Anagnostou, V. et al. Evolution of Neoantigen Landscape during Immune Checkpoint Blockade in Non-Small Cell Lung Cancer. Cancer discovery 7, 264-276, doi:10.1158/2159-8290.CD-16-0828 (2017).

19 Tran, E. et al. T-Cell Transfer Therapy Targeting Mutant KRAS in Cancer. The New England journal of medicine 375, 2255-2262, doi:10.1056/NEJMoa1609279 (2016).

20 McGranahan, N. et al. Allele-Specific HLA Loss and Immune Escape in Lung Cancer Evolution. Cell 171, 1259-1271 e1211, doi:10.1016/j.cell.2017.10.001 (2017).

21 Thielens, A., Vivier, E. & Romagne, F. NK cell MHC class I specific receptors (KIR): from biology to clinical intervention. Curr Opin Immunol 24, 239-245, doi:10.1016/j.coi.2012.01.001 (2012).

22 Fischer, J. C. et al. Relevance of C1 and C2 epitopes for hemopoietic stem cell transplantation: role for sequential acquisition of HLA-C-specific inhibitory killer Ig-like receptor. J Immunol 178, 3918-3923 (2007).

23 McGranahan, N. et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science (New York, N.Y 351, 1463-1469, doi:10.1126/science.aaf1490 (2016).

24 Garrido, F., Ruiz-Cabello, F. & Aptsiauri, N. Rejection versus escape: the tumor MHC dilemma. Cancer Immunol Immunother 66, 259-271, doi:10.1007/s00262-016-1947-x (2017).

25 Dunn, J. & Rao, S. Epigenetics and immunotherapy: The current state of play. Mol Immunol 87, 227-239, doi:10.1016/j.molimm.2017.04.012 (2017).

26 Bernard-Tessier, A. et al. Outcomes of long-term responders to anti-programmed death 1 and anti-programmed death ligand 1 when being rechallenged with the same anti-programmed death 1 and anti-programmed death ligand 1 at progression. Eur J Cancer 101, 160-164, doi:10.1016/j.ejca.2018.06.005 (2018).

27 Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15-21, doi:10.1093/bioinformatics/bts635 (2013).

28 Li, B. & Dewey, C. N. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC bioinformatics 12, 323, doi:10.1186/1471-2105-12-323 (2011).

29 Danaher, P. et al. Gene expression markers of Tumor Infiltrating Leukocytes. J Immunother Cancer 5, 18, doi:10.1186/s40425-017-0215-8 (2017).

30 Denkert, C. et al. Standardized evaluation of tumor-infiltrating lymphocytes in breast cancer: results of the ring studies of the international immuno-oncology biomarker working group. Mod Pathol 29, 1155-1164, doi:10.1038/modpathol.2016.109 (2016).

Page 18: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

18

31 Oakes, T. et al. Quantitative Characterization of the T Cell Receptor Repertoire of Naive and Memory Subsets Using an Integrated Experimental and Computational Pipeline Which Is Robust, Economical, and Versatile. Front Immunol 8, 1267, doi:10.3389/fimmu.2017.01267 (2017).

32 Best, K., Oakes, T., Heather, J. M., Shawe-Taylor, J. & Chain, B. Computational analysis of stochastic heterogeneity in PCR amplification efficiency revealed by single molecule barcoding. Scientific reports 5, 14629, doi:10.1038/srep14629 (2015).

33 Andreatta, M. & Nielsen, M. Gapped sequence alignment using artificial neural networks: application to the MHC class I system. Bioinformatics 32, 511-517, doi:10.1093/bioinformatics/btv639 (2016).

34 Arrieta, V. A. et al. The possibility of cancer immune editing in gliomas. A critical review. Oncoimmunology 7, e1445458, doi:10.1080/2162402X.2018.1445458 (2018).

Page 19: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

19

Main Figure Legends

Figure 1: Heterogeneity of immune infiltration in NSCLC. (A-B) TRACERx regions from

lung adenocarcinoma (A) and lung squamous cell carcinoma (B) are shown, clustered by the

level of estimated immune infiltrate. Each row represents an immune cell population, as

estimated by the Danaher method. Immune populations are: B cells, CD4+ T-cells, CD8+ T-

cells, exhausted CD8+ T-cells, helper T-cells, regulatory T-cells, CD45+ cells, NK cells, NK

CD56- cells, dendritic cells, mast cells, macrophages, neutrophils, cytotoxic cells, total T-cells,

and total TIL score. Each column represents a tumor region. Regions classified as having low

immune infiltration are shown in blue, whereas regions classified as having high immune

infiltration are shown in red. If all regions from a patient’s tumor are classified as low immune,

that patient is indicated in blue. If all regions from a patient’s tumor are classified as high

immune, that patient is indicated in red. Patients with tumors containing heterogeneous

immune infiltration are indicated in orange. Below each heatmap, example pathology images

from heterogeneous tumors are shown to display a region of high immune infiltration and a

region of low immune infiltration from the same tumor.

Figure 2: Immune editing at the DNA level. (A) Pairwise genomic and immune distances

between every two tumor regions from the same patient are compared (lung adeno: p=3.5e-

04, n=217 lung squam: p=0.002, n=186). (B-C) The Shannon diversity index for each tumor

region is shown grouped by immune classification. Lung adenocarcinomas (n=159) (B) and

lung squamous cell carcinomas (n=103) (C) are shown. Minima and maxima indicated by

extreme points of boxplot. Median indicated by thick horizontal line. First and third quartiles

indicated by box edges. A two-sided Wilcoxon rank-sum test is used. (D) The change in the

observed/expected immunoediting score from clonal (C) to subclonal (S) is shown for each

immune classification (high, n=24; hetero., n=25; low, n=33). A two-sided paired t-test is used.

(E) Example of historically clonal neoantigens loss by subclonal copy number event.

Neoantigens present in CRUK0071:R3 on one copy are shown in one panel (black). These

neoantigens are lost in CRUK0071:R6 (red). (F) The number of historically clonal neoantigens

Page 20: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

20

on a region of copy number loss are shown per tumor. Below shows the proportion of clonal

neoantigens lost subclonally through a copy number event. (G) The odds ratio and 95% CI of

copy number neoantigen depletion is shown, calculated with Fisher’s exact test. Values >1

indicate neoantigens are more likely to be in regions of subclonal copy number loss as

compared to non-synonymous mutations that are not neoantigens. Tumor regions are

classified by immune cluster. (H) The change in immunoediting score is shown for immune

low tumors by whether any neoantigens are subclonally lost through copy number events (CN-

loss, n=17; no-CN-loss, n=16). A two-sided paired t-test is used. No corrections were made

for multiple comparisons.

Figure 3: Transcriptional neoantigen depletion. (A) The patient-level number of clonal and

subclonal expressed neoantigens is shown. The fraction of clonal neoantigens that are

ubiquitously detected is plotted below. The immune class is provided as high (red), low (blue),

or heterogeneous (orange). (B) The fraction of clonal neoantigens that are ubiquitously

detected in every region is plotted by immune classification of the tumor (n=63). Minima and

maxima indicated by extreme points of boxplot. Median indicated by thick horizontal line. First

and third quartiles indicated by box edges. A two-sided Wilcoxon rank-sum test is used. (C)

The odds ratio and 95% CI of transcriptional neoantigen depletion is shown, calculated with

Fisher’s exact test. Values <1 indicate that putative neoantigens are less likely to be expressed

as compared to non-synonymous mutations that are not putative neoantigens. Tumors are

plotted by HLA LOH status and immune classification. (D) The odds ratio and 95% CI of a

neoantigen occurring in a gene that is consistently expressed among TCGA NSCLC tumors

is shown, calculated with Fisher’s exact test. (E) CpG-methylation patterns across the LAMB1

promoter in tumor samples CRUK0057:R1 and CRUK0002:R1 and their matched normals.

The locus encodes two non-expressed neoantigens and exhibits hypermethylation in

CRUK0057:R1. The purity/ploidy-matched unmutated control sample CRUK0002:R1 shows

no differential methylation. (F-H) Numbers of (non)-hypermethylated gene promoters for (F)

expressed vs. non-expressed neoantigens, (G) non-expressed neoantigens vs. the same

Page 21: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

21

genes in purity/ploidy-matched controls and (H) non-expressed neoantigens vs. the same

genes in purity/ploidy-matched controls. Odds ratios (OR) and p-values (χ2-test) are shown

for each comparison. No corrections were made for multiple comparisons.

Figure 4: Immune evasion capacity in early-stage non-treated NSCLC. (A-B) The number

of clonal and subclonal neoantigens found in the tumor region, immune cluster, patient

prognosis, immunoediting classification, HLA LOH status, and antigen presentation defects

are plotted for every tumor region for each tumor. Patients are split according to their immune

evasion capacity. (C) Immune evasion capacity is determined by the level of immune

infiltration and presence of immune escape mechanisms. Patients whose tumors have low

immune evasion capacity have prolonged disease-free survival times. (D) A Kaplan Meier

curve is shown for tumors with low clonal neoantigen burden (lowest three quartiles) split by

their immune evasion capacity. (E) A Kaplan Meier curve is shown that combines clonal

neoantigen load (upper quartile) and immune evasion capacity. For all survival curves, the

number of patients in each group for every time point is indicated below the time point and

significance is determined using a two-sided log-rank test.

Page 22: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

22

Methods

Patients and samples

The cohort evaluated within this study comes from the first 100 patients prospectively

analyzed by the lung TRACERx study (https://clinicaltrials.gov/ct2/show/NCT01888601,

approved by an independent Research Ethics Committee, 13/LO/1546) and mirrors the

prospective 100 patient cohort described in 5.

Informed consent for entry into the TRACERx study was mandatory and obtained from every

patient. There were 68 male and 32 female non-small cell lung cancer patients in the

TRACERx study, with a median age of 68. The cohort is predominantly early-stage: Ia(26),

Ib(36), IIa(13), IIb(11), IIIa(13), IIIb(1). Seventy-two had no adjuvant treatment and 28 had

adjuvant therapy. All patients were assigned a study ID that was known to the patient. These

were subsequently converted to linked study Ids such that the patients could not identify

themselves in study publications. All human samples, tissue and blood, were linked to the

study ID and barcoded such that they were anonymized and tracked on a centralized database

overseen by the study sponsor only.

TRACERx 100 RNA-sequencing

RNA was extracted from the TRACERx 100 cohort using a modification of the AllPrep kit

(Qiagen) as described in Jamal-Hanjani et al. 5. RNA integrity was assessed by TapeStation

(Agilent Technologies). Samples that had a RIN score >=5 were sent to the Oxford Genomics

Centre for whole RNA (RiboZero depleted) paired end sequencing. The ribodepleted fraction

was selected from the total RNA provided before conversion to cDNA. Second strand cDNA

synthesis incorporated dUTP. The cDNA was end-repaired, A-tailed and adapter-ligated. Prior

to amplification samples underwent uridine digestion. The prepared libraries were size

selected, multiplexed and QC’ed before paired end sequencing. Reads were 75 base pairs in

Page 23: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

23

length. FASTQ data was quality controlled and aligned to the hg19 genome using STAR 27.

Transcript quantification was performed using RSEM with default parameters 28.

TRACERx 100 RRBS

Reduced representation bisulfite sequencing (RRBS) was obtained for roughly half of the

NSCLC cohort with RNA-Seq data (79/164 tumor regions from 28/64 patients, each with

matched normal). The NuGEN Ovation RRBS Methyl-Seq System, adapted by the

manufacturer for automation on an Agilent Bravo liquid handling robot, was used to generate

sequencing libraries by enzymatically digesting 100 ng of gDNA using MspI, followed by

adaptor ligation and the final repair step. Generated libraries were bisulfite converted using

Qiagen’s EpiTect Fast DNA Bisulfte Kit purchased separately from the kit, PCR amplified for

12 cycles and purified using Agencourt® RNAClean® XP magnetic beads. Purified libraries

were quantified by Qubit dsDNA HS Assay (Invitrogen) and quality controlled using Agilent

Bioanalyzer HighSensitivity DNA Assay (Agilent Technologies). Eight samples were

multiplexed per flow cell and sequenced on an Illumina HiSeq2500 system using HiSeq SBS

Kit v4 in paired-end 100bp runs for CRUK0062 and single end 100bp runs for the others

yielding on average 150M raw sequencing reads per sample. Sequencing results were

checked with FastQC v0.11.2 (Babraham Institute, https://www.babraham.ac.uk/), adapter

sequences were trimmed with Trim Galore! v0.3.7, which is a wrapper around Cutadapt

(doi:10.14806/ej.17.1.200), and NuGEN v1.0 diversity trimming script

(https://github.com/nugentechnologies/NuMetRRBS) and reads aligned to the UCSC hg19

reference assembly using Bismark v0.14.430. Read deduplication was carried out using

NuDup (pre-release version dated March 2015,

https://github.com/nugentechnologies/nudup/), leveraging NuGEN’s molecular tagging

technology producing on average 100M unique reads per sample.

Statistical information

Page 24: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

24

All statistical tests were performed in R. No statistical methods were used to predetermine

sample size. Tests involving correlations were done using “cor.test” with the Spearman’s

method. Tests involving comparisons of distributions were done using “wilcox.test” or “t.test”

using the unpaired option, unless otherwise stated. Hazard ratios and p-values were

calculated with the “survival” package. For all statistical tests, the number of data points

included are plotted or annotated in the corresponding figure.

Selection of immune infiltration approach

Previously defined measures of immune infiltration and activity were used to classify the

immune microenvironment of all tumors (and tumor regions) with RNAseq data available 6-

8,11,29. The genes used in each one of the immune estimation approaches were tested to see

if they fit two criteria: 1) have a negative relationship with tumor purity, as genes defining

immune subtypes are expressed in infiltrating immune cells 8 and 2) not show a positive

correlation with tumor copy number at the gene locus, a positive correlation may indicate that

the gene is expressed by the tumor cell, thereby confounding immune estimates. The

proportion of genes in each immune estimation method that passed these two criteria was

compared. Finally, for each method, the immune estimates themselves were compared

against independent ground truth measures (pathology TIL estimation, flow cytometry

quantification, and TCR abundance). The immune estimation that performed best in the

TRACERx cohort was chosen.

Estimating immune cell populations

RNAseq-based estimations

The Danaher method 29 was used to estimate immune cell populations for every tumor region

with RNAseq data available. The immune cell populations were: CD8+ T-cells (cd8),

exhausted CD8+ T-cells (cd8.exhausted), CD4+ T-cells (cd4), regulatory T-cells (treg), helper

T-cells (th1), dendritic cells (dend), B cells (bcell), mast cells (mast), NK cells (nk), NK

Page 25: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

25

CD56dim cells (nkcd56dim), neutrophils, macrophages, CD45+ cells (cd45), and measures

for total T-cells (tcells), total TILs (total.til), and cytotoxic cells (cyto). Because the original

Danaher paper did not identify any suitable genes for CD4+ T-cell population estimation and

a poor relationship with ground truth measures was observed in the TRACERx cohort using

the Danaher CD4+ T-cell estimates, the Davoli CD4+ T-cell estimates were used instead. The

Davoli estimate was chosen as overall, they matched the Danaher estimates closely and

performed nearly as well for the selection criteria.

The Jiang immune measures were calculated using the TIDE web interface

(http://tide.dfci.harvard.edu/)

Pathology TIL estimation

TILs were estimated from pathology slides using international established guidelines

developed by the International Immuno-Oncology Biomarker Working Group the Salgado

method 10. Briefly, from the pathology slide of a given tumor region, the relative proportion

stromal area to tumor area was determined. TILs were reported for the stromal compartment

(=% stromal TILs). The denominator used to determine the % stromal TILs is the area of

stromal tissue (i.e. area occupied by mononuclear inflammatory cells over total intratumoral

stromal area), not the number of stromal cells (i.e. fraction of total stromal nuclei that represent

mononuclear inflammatory cell nuclei). This method has been demonstrated to be

reproducible among trained pathologists 30. An intra-personal concordance was performed

and this demonstrates high reproducibility. The International Immuno-Oncology Biomarker

Working Group has developed a freely available training tool to train pathologists for optimal

TIL-assessment on hematoxylin eosin slides (www.tilsincancer.org).

Flow measurements

Tissue samples were collected and transported in RPMI-1640 (Sigma, cat# R0883-500ML).

Single cell suspensions were produced by enzymatic digestion using liberase with subsequent

Page 26: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

26

cellular disaggregation using a Miltenyi gentleMACS Octo Dissociator. Lymphocytes were

isolated from single cell suspension by gradient centrifugation on Ficoll Paque Plus (GE

Healthcare, cat# 17-1440-03) and stored in liquid nitrogen. Blood samples were collected in

BD Vacutainer EDTA blood collection tubes (BD cat# 367525), PBMC’s were then isolated by

gradient centrifugation on Ficoll Paque (GE Healthcare, cat# 17-1440-03) and stored in liquid

nitrogen.

FC receptors were blocked with Human Fc Receptor Binding Inhibitor (Thermo) before

staining. Non-viable cells were stained using the eBioscience Fixable Viability Dye eFluor 780

(Thermo). Cells were stained in BD Brilliant stain buffer (BD cat# 563794) with the following

monoclonal antibodies: anti-human CD3 (clone SK7, BD cat# 565511), anti-human CD4

(clone SK3, BD cat# 566003), anti-human CD8 (clone RPA-T8, BD cat# 564804). Data was

acquired on a BD Symphony flow cytometer and analyzed in FlowJo. Cells were gated for

size, single cells, live cells, CD3+CD8+ T cells.

TCR abundance

A previously developed quantitative experimental and computational TCR sequencing

pipeline 31 was used for the high throughput sequencing of α and β TCR chains. TCR

sequencing was performed on whole RNA extracted from multi-region tumor specimens. A

distinct feature of this TCR sequencing protocol is the utilization of a unique molecular

identifier (UMI) that enables correction for PCR and sequencing errors, thereby providing a

quantitative and reproducible method of library preparation 31,32.

Classifying tumor regions as immune high/low

Tumors were split into either lung adenocarcinoma or lung squamous cell carcinoma. The

Danaher estimates for all tumor regions from each histological type were clustered together

using “ward.D2”. The dendrogram was cut into two, and the samples which fell in the portion

with higher levels of immune infiltrate estimation were considered immune high tumor regions.

Page 27: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

27

Conversely, the samples which portion with lower levels of immune infiltrate estimation were

considered immune low tumor regions. If all tumor regions from a given sample were classified

as immune low, that tumor was designated as consistently immune low; if all tumor regions

from a given sample were classified as immune high, that tumor was designated as

consistently immune high. If some tumor regions from the same tumor were immune high and

others were immune low, the tumor overall was classified as heterogeneous.

If a tumor region had no RNAseq available, it could be rescued using the pathology TIL

estimations. A tumor region was classified based on pathology TILs by determining if the

pathology TIL estimate for the tumor region in question was closer to the median of the

pathology TILs from the immune high or immune low tumor regions with RNAseq that had

been clustered. The RNAseq cohort (164 tumor regions from 64 TRACERx patients) was

expanded by rescuing tumor regions without RNAseq data (Extended Data Fig. 2A) with

pathology estimated TILs (234 tumor regions from 83 TRACERx patients) (Extended Data Fig.

4E).

Calculation of IPRES score

The calculation of the IPRES score was done according to Hugo et al. 13.

Distance measures

Immune distance

The immune distance was determined by taking the Euclidean distance of immune infiltrate

estimates between tumor regions.

Genomic distance

The genomic distance was calculated by taking the Euclidean distance of the mutations

present between tumor regions. All mutations present in any region from a tumor were turned

Page 28: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

28

into a binary matrix, where the rows were mutations and columns tumor regions. This matrix

was clustered and the pairwise distance between any two tumor regions was determined.

Calculation of Shannon entropy

For each tumor region, the Shannon entropy was estimated using the command

“entropy.empirical” from the “entropy” R package. This was calculated based on the number

and prevalence of different tumor subclones found in that region, such that a tumor region

containing only one subclone was assigned a value of 0.

The Shannon entropy score, H, followed the formula: H = -Σpi log (pi), where pi is the

probability of the ith clone appearing in the tumor cell population.

Predicted neoantigen binders

Novel 9-11mer peptides that could arise from identified non-silent mutations present in the

sample 5 were determined. The predicted IC50 binding affinities and rank percentage scores,

representing the rank of the predicted affinity compared to a set of 400,000 random natural

peptides, were calculated for all peptides binding to each of the patient’s HLA alleles using

netMHCpan-2.8 17,33 and netMHC-4.0 33. Using established thresholds, predicted binders were

considered those peptides that had a predicted binding affinity <500nM or rank percentage

score <2% by either tool. Strong predicted binders were those peptides that had a predicted

binding affinity <50nM or rank percentage score <0.5%. Of the 28,489 non-synonymous

mutations in this cohort, 24,494 were predicted to encode peptides capable of binding to at

least one of the patient’s HLA class I alleles (binding affinity < 500nM or rank% < 2) and 13,884

were predicted to strongly bind (binding affinity < 50nM or rank% < 0.5) 17.

When RNAseq data was available, a neoantigen was considered to be expressed if at least

five RNAseq reads mapped to the mutation position, and at least three contained the mutated

base.

Page 29: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

29

Neoantigen depletion

Transcriptional

Transcriptional neoantigen depletion was identified by first dividing tumors into immune

classifications and HLA LOH categories (loss/no loss). All non-synonymous mutations were

annotated as expressed in the RNAseq or not using the definitions above. Then a test for

enrichment was performed to determine if non-synonymous mutations that were neoantigens

were less likely to be expressed as compared to the non-synonymous mutations which were

not predicted to be neoantigens.

Copy number

Copy number neoantigen depletion was identified by first dividing tumors into immune

classifications. All non-synonymous mutations were annotated as either in a region of

subclonal copy number loss or not as identified in Jamal-Hanjani et al. 5. Then a test for

enrichment was performed to determine if non-synonymous mutations that were neoantigens

were more likely to be in regions of subclonal copy number loss as compared to the non-

synonymous mutations which were not predicted to be neoantigens.

Methylation

Neoantigens in genes that are consistently expressed across the TCGA NSCLC cohort were

classified in two groups: expressed, where the mutant is detected in at least 30 reads, and

non-expressed, where no mutant transcript is observed. Of the 375 non-expressed and 883

expressed neoantigens with matched RRBS data, 77 and 406 were unique, respectively

(others were duplicates from different regions of the same patient). We down-sampled the

expressed neoantigens list to match as closely as possible the gene expression and the

variant allele frequency distributions observed for the non-expressed neoantigens. We then

assessed differential methylation as follows: bulk and normal per-CpG methylation rates in

promoters (2kb up- and downstream of TSS) modelled as beta distributions, B(α+1,β+1),

Page 30: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

30

where α represents the observed methylated read counts and β the unmethylated read counts,

and we compute 𝑃(𝐵(𝛼, 𝛽)𝑡𝑢𝑚 > 𝐵(𝛼, 𝛽)𝑛𝑜𝑟𝑚) exactly via:

𝑃𝑟 𝑃𝑟 (𝑝𝑡𝑢𝑚 > 𝑝𝑛𝑜𝑟𝑚) = ∑

𝛼𝑡𝑢𝑚−1

𝑖=0

𝐵(𝛼𝑛𝑜𝑟𝑚 + 𝑖, 𝛽𝑛𝑜𝑟𝑚 + 𝛽𝑡𝑢𝑚)

(𝛽𝑡𝑢𝑚 + 𝑖)𝐵(1 + 𝑖, 𝛽𝑡𝑢𝑚)𝐵(𝛼𝑛𝑜𝑟𝑚, 𝛽𝑛𝑜𝑟𝑚)

Hochberg family-wise error rate (FWER) correction is then applied and promoters are flagged

as hypermethylated when ≥3 CpGs are significantly hypermethylated (q<0.05). Promoter

counts are tested in a 2x2 contingency table (methylation status vs expression status or

mutation status) using a χ^2-test.

Identifying tumor regions with HLA LOH

Tumor regions harboring an HLA LOH event were identified using the LOHHLA method,

described in 20.

Immune evasion alterations

Antigen presentation pathway genes were compiled from 34 and affected the HLA

enhanceosome, peptide generation, chaperones, or the MHC complex itself. They included

disruptive events (non-synonymous mutations or copy number loss defined relative to ploidy

5) of the following genes: CIITA, IRF1, PSME1, PSME2, PSME3, ERAP1, ERAP2, HSPA,

HSPC, TAP1, TAP2, TAPBP, CALR, CNX, PDIA3, B2M.

TCGA data

RNA-sequencing data was downloaded from the TCGA data portal. For each LUAD and LUSC

sample, all available ‘Level_3’ gene-level data was obtained. TCGA genes were considered

consistently expressed if they were expressed at >= 1TPM in 95% of the samples for each

histology.

Page 31: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

31

Data Availability

Sequence data used during the study will be deposited at the European Genome-phenome

Archive (EGA), which is hosted by The European Bioinformatics Institute (EBI) and the Centre

for Genomic Regulation (CRG) under the accession code: EGAS00001003458. Further

information about EGA can be found at https://ega-archive.org.

Code Availability

All code used for analyses was written in R version 3.3.1 and is available at:

https://bitbucket.org/snippets/raerose01/EeLrLB

Page 32: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

32

Extended Data Figure Legends

Extended Data Fig. 1: Determination of robust immune infiltration approach. (A-D) The

expression of the genes used in the each of the immune signature definitions is correlated

against tumor purity (A-B) and tumor copy number (C-D). Plotted are random genes (n=1000),

TIMER genes (n=575), EPIC genes (n=98), Danaher genes (n=60), Rooney genes (n=100),

and Davoli genes (n=75). The Spearman’s rho value of the correlation is plotted for the

immune genes comprising each signature definition, colored by the p-value of the association.

The comparisons are performed separately for lung adenocarcinoma and lung squamous cell

carcinoma. The median rho value for the immune signature set is indicated by the red line.

The fraction of genes whose expression value is significantly correlated with purity or tumor

copy number is shown and compared to a set of random genes. For every immune signature

considered, there was significant enrichment of genes whose expression negatively correlated

with tumor purity as compared to the random selection of genes and a significant enrichment

of genes whose expression positively correlated with tumor copy number as compared to the

random selection of genes. (E) Scatterplots show the Spearman correlation between TIL

scores and CD8+ T-cells as measured by the Danaher approach (n=140), between flow CD8+

T-cell estimates and Danaher CD8+ T-cells (n=36), TCRseq abundance and Danaher CD8+

T-cells (n=72), normalized live flow CD8+ T-cell estimates and Danaher CD8+ T-cells (n=39),

and normalized live flow CD8+ T-cell/Treg and Danaher CD8+/Treg estimates (n=38). Blue

dots indicate regions from a lung adenocarcinoma tumor, red dots indicate regions from a lung

squamous cell carcinoma tumor. Spearman rho values, p-values, and 95% CI (shaded area)

are given for all tumor regions (black), lung adenocarcinoma tumor regions (blue), and lung

squamous cell carcinoma tumor regions (red). (F) A scatterplot showing the correlation

between pathology TIL estimates and CD8+ estimates from each of the immune infiltration

methods is shown (n=140). Lung adenocarcinoma tumor regions are shown in blue; lung

squamous cell carcinoma tumor regions are shown in red. Below, the top six correlations

between pathology TIL estimates and an immune cell subset is shown for each method. Blue

Page 33: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

33

boxes indicate positive correlation, whereas red boxes indicate negative correlation. P-values

were FDR corrected. (G) Example of CD8 T-cell quantification in a representative TRACERx

TIL sample. TILs were isolated from tumor regions of surgical resections as previously

described and cryopreserved. Thawed samples were stained with a custom-designed 20-

marker antibody panel to measure T cell activation, dysfunction and differentiation by flow

cytometry.

Extended Data Fig. 2: TRACERx 100 sample selection and patient characteristics. (A)

CONSORT diagram showing the selection of TRACERx 100 patients for RNAseq and/or

pathology TIL analysis. (B) Patient characteristics for the TRACERx 100 cohort are shown.

Patient characteristics can be found in tabular form in Table S1.

Extended Data Fig. 3: Difference in immune infiltration by histology. The distribution of

Danaher estimated CD8+ T-cell infiltrate is displayed for lung adenocarcinomas (adeno.) and

lung squamous cell carcinomas (squam.) (n=145). Minima and maxima indicated by extreme

points of boxplot. Median indicated by thick horizontal line. First and third quartiles indicated

by box edges. A two-sided Wilcoxon rank-sum test is used.

Extended Data Fig. 4: Rescuing regions without RNAseq using pathology TILs. (A) The

difference in pathology TIL estimates is shown by RNAseq-derived immune cluster (n=139).

(B) All regional pathology estimated TILs are plotted for each tumor sample (lung

adenocarcinoma n=121; lung squamous cell carcinoma n=90). If a region also had RNAseq

information available, the immune cluster that region belonged to is also shown as immune

high (red) or immune low (blue). Immune clusters for tumor regions without RNAseq are

annotated as grey. The immune class for the patients is also provided as high (red), low (blue),

heterogeneous (orange), or unknown (grey). For all boxplots, minima and maxima indicated

by extreme points of the plot. Medians are indicated by thick horizontal line. First and third

quartiles are indicated by box edges. A two-sided Wilcoxon rank-sum test is used for

comparisons. (C) The number of patients in each immune classification is plotted as inferred

Page 34: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

34

from using RNAseq data alone or by also incorporating pathology TIL estimates. (D) A

correlation matrix of the Danaher immune cell estimates with the Jiang immunosuppressive

cell subsets is shown (Spearman’s test). Positive correlations are indicated in blue and

negative correlations are indicated in red. Correlations are significant unless marked with a

black X. (E) The Jiang immune infiltration estimates are shown for TAM M2 (tumor associated

macrophage M2) and MDSC (myeloid-derived suppressor cells) cells split by immune cluster

(n=163). (F) The tumor purity is shown for the low tumor mutational burden (TMB) and high

TMB regions of every tumor with heterogeneous TMB (n=12) Two-sided paired t-test is used

for comparison. No corrections were made for multiple comparisons.

Extended Data Fig. 5: Heterogeneity of biomarkers predicting checkpoint blockade

response. (A) The TIDE gene signature score of each tumor region is shown per patient for

patients with >1 region available (n=39). Using threshold defined by (dashed line), patients

are classified as having low TIDE (light blue), high TIDE (dark blue), or heterogeneous TIDE

(orange). (B) The IPRES gene signature score of each tumor region is shown per patient for

patients with >1 region available (n=39). Using threshold defined by Hugo et al. 13 (dashed

line), patients are classified as having low IPRES (light blue), high IPRES (dark blue), or

heterogeneous IPRES (orange). (C) The expanded Ayers IFN signature is shown for each

tumor region per patient for patients with >1 region available (n=38). For (A-C) the immune

classification of the patient is also given. (D) The greatest difference in expanded Ayers IFN

signature between tumor regions from the same tumor is plotted according to whether the

tumor has heterogeneous immune infiltration or not (n=38). A two-sided Wilcoxon rank-sum

test is used for comparison. (E) Tumor mutational burden (TMB) of each tumor region is shown

per patient (n=93). Using a 10 mutations/mB threshold (dashed line), patients are classified

as having low TMB (light blue), high TMB (dark blue), or heterogeneous TMB (orange). For

all boxplots, minima and maxima indicated by extreme points of the plot. Medians are indicated

by thick horizontal line. First and third quartiles are indicated by box edges. (F) A summary of

the tumor histology, immune classification, TMB status, TIDE category, and IPRES category

Page 35: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

35

is shown for each tumor (n=93). There is an enrichment for heterogeneously immune infiltrated

tumors to have heterogeneous TMB status and heterogeneous TIDE scores (Fisher’s exact

test). No corrections were made for multiple comparisons.

Extended Data Fig. 6: Relationship between immune infiltration and tumor region

diversity. (A) The pairwise copy number (cn) and immune distances between every two tumor

regions from the same patient are compared for lung adenocarcinoma (n=91) and lung

squamous cell carcinoma (n=60). (B-C) For each tumor region, the CD8+ T-cell score is

plotted against the Shannon diversity score. Lung adenocarcinomas (n=89) (B) and lung

squamous cell carcinomas (n=50) (C) are shown. (D) The correlation between pathology TIL

estimates and tumor purity is shown for lung adenocarcinoma (n=120) (blue) and lung

squamous cell carcinoma (n=90) (red) regions. No relationship for either histology is observed.

Spearman’s test is used to determine relationship. (E) The Shannon diversity score per lung

adenocarcinoma tumor region (n=137) is plotted by immune classification as determined

solely by pathology TIL estimates. A two-sided Wilcoxon rank-sum test is used for comparison.

(F) A comparison of observed/expected immunoediting score between lung adenocarcinoma

and lung squamous cell carcinoma tumors (n=92) is shown. A two-sided Wilcoxon rank-sum

test is used for comparison. (G) The observed/expected immunoediting score is shown by

number of unique HLAs present in the tumor (patients heterozygous at HLA-A, -B, and -C will

have six unique HLA alleles) (n=90). For all boxplots, minima and maxima indicated by

extreme points of the plot. Medians are indicated by thick horizontal line. First and third

quartiles are indicated by box edges. (H) The odds ratio and 95% CI of transcriptional

neoantigen depletion is shown for strongly binding neoantigens, calculated with Fisher’s exact

test. Values <1 indicate that putative neoantigens are less likely to be expressed as compared

to non-synonymous mutations that are not putative neoantigens. Tumors are broken down by

HLA LOH status and their immune classification. (I) The enrichment for neoantigens and

strongly binding neoantigens to occur in non-expressed genes as compared to non-

Page 36: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

36

synonymous non-neoantigens is shown, calculated with Fisher’s exact test. No corrections

were made for multiple comparisons.

Extended Data Fig. 7: Components of immune evasion mechanisms in NSCLC. (A) Each

of the potential immune evasion mechanisms explored in Figure 4 are shown broken down by

their component genes. Patients are split according to their immune evasion capacity status.

Copy number losses are shown in blue and mutations are shown in green. (B) A schematic of

how LOH of the HLA-C locus in HLA-C1/C2 heterozygous tumors may lead to NK cell-

mediated destruction is shown. (C) The level of Danaher estimated NK cell infiltration / Total

TIL estimate is shown for tumor regions with (n=45) and without (n=90) HLA-C LOH according

to their HLA-C1/C2 heterozygosity status. A two-sided Wilcoxon rank-sum test is used for

comparison.

Extended Data Fig. 8: Relationship between clonal neoantigen burden, immune

infiltration, and patient prognosis. (A, C, E) Kaplan-Meier curves are shown for lung

adenocarcinoma and lung squamous cell carcinoma. The curves are split based on the upper

quartile of clonal neoantigen burden (A), on the upper quartile of subclonal neoantigen burden

(C), and on the upper quartile of total neoantigen burden (E). For all survival curves, the

number of patients in each group for every time point is indicated below the time point and

significance is determined using a log-rank test. (B, D) The hazard ratio is shown for each

threshold value of clonal neoantigen (B) and subclonal neoantigen (D) load, indicating that a

high clonal neoantigen burden remains significantly prognostic across a wide range of

thresholds. Significant associations are indicated in red, whereas non-significant associations

are plotted in black. (F) Both clonal neoantigen load and immune infiltration classification are

incorporated in a multivariate analysis, becoming more significant when the variables are

combined as compared to either metric individually. Other tumor and clinical characteristics

are also controlled for in the multivariate analysis. Hazard ratios of each variable with a 95%

CI are shown on the horizontal axis. Significance is calculated using a Cox proportional

hazards model. All statistical tests were two-sided.

Page 37: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region
Page 38: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

A

lung adeno.lung squam.

Sha

nnon

div

ersi

tyC

opy

num

ber

Sha

nnon

div

ersi

ty

low heterogeneous high low highImmune classification Immune classification

heterogeneous

Lung adenocarcinoma Lung squamous cell carcinomaBp: 0.01 p: 0.18

0.0

0.5

1.0

1.5

2.0

0.0

0.5

1.0

1.5

2.0

C

G

E F

Odd

s ra

tio:

neo

in re

gion

of C

N lo

ss

+neo-neo

p = 0.97 p = 0.13 p = 3.2e−03C S

0.7

0.8

0.9

1.0

1.1

1.2

Obs

erve

d/E

xpec

ted

Imm

unoe

ditin

g

0.7

0.8

0.9

1.0

1.1

1.2

0.7

0.8

0.9

1.0

1.1

1.2

D High Hetero. Low

CN loss no CN loss

Clonal Subclonal

0.7

0.8

0.9

1.0

1.1

1.2

p = 0.88Clonal Subclonal

0.7

0.8

0.9

1.0

1.1

1.2

p = 2.2e−04

H Immune low tumors

All High Low

0

1

2

3

p = 9.4e−04 p = 3.8e−01 p = 3.3e−04

1192/51325 540/22493 515/21337147/8356 80/3721 50/3423

CRUK0071:R3

0

1

2

3

4CRUK0071:R6

Patient

Num

ber n

eo in

regi

on C

N lo

ss

0

0

10

20

30

40

major allele CNminor allele CN CN loss neo

neo present

0.5

Obs

erve

d/E

xpec

ted

Imm

unoe

ditin

g

C S C S

0 10 20 30 400

2

4

6

8

10

Pairwise genomic distance

Pai

rwis

e im

mun

e di

stan

ce p: 3.5e−04Spearman’s rho: 0.35

p: 0.002Spearman’s rho: 0.37

Lung adeno.

Lung squam.

Page 39: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

C

F G H

A

D

BO

R: n

eo d

eple

tion

Clonal Neo / exp.Subclonal Neo / exp.

p=0.49 p=0.01 p=0.39 p=0.01 p=0.54 p=0.03 p=0.84 p=0.96

HLALOH

All High Hetero. Low

0

0.5

1

1.5

lung adeno.lung squam.

0

200

400

600

800N

umbe

r neo

.

Fraction clonal neo. ubiquitously expressed0

1

High Hetero. Low

OR

:exp

ress

ed n

eo

0

0.5

1

p=2.1e−04 p=0.04 p=1.8e−03

expressednot expressed

60

Num

ber n

eo.

40

20

0

OR= 11.4; p=1.6e-04 OR=0.48; p= 6.7e-01OR=2.5; p=3.0e-02

Non-mutatedcontrol

Non-mutatedcontrol

Non-expressedneo.

Non-expressedneo.

Expressedneo.

Expressedneo.

182

59

18

5975

2

75 73

49

68

Chromosome position (kb)

●●●● Normal methylation rate

●●●

●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●

●●●●●●●●●●●●●●●●●●●●●●●●●●●●● ●

●●●●

●● ●●●●●●●●

●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●● ●●

●●

TSS

4kb

Hypermethylatednon-expressed neoantigen

0.80

0.40

0.00Met

hyla

tion

rate

* Neoantigenic mutation

LAMB1

CRUK0057:R1

●●●●

●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●

●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●● ●●

●●

●●●● ●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●

●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●● ●●

CRUK0002:R1

Unmutated LAMB1control methylation profile

E

107575 107600 107625

Tumor methylation rate

Promoter region

Hypermethylated (tumor)Hypomethylated (tumor)

Frac

tion

clon

al n

eo.

ubiq

uito

usly

exp

ress

ed

lung adeno. lung squam.

Hypermeth:

-+ -+ -+ -+

- + - + - +- + - + - +

* *

low highhetero.Immune classification

p: 0.01

0.0

0.2

0.4

0.6

0.8

1.0

Page 40: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

Lung squam.

Lung adeno.

no alteration

alteration

no immuneeditingimmuneediting

increasing DFS (patient not recurred)

increasing DFS (patient recurred)

high immune region

low immune regionclonal neoantigen

subclonal neoantigenNA

Immune

HLALOHAntigen pres.

DFSImmune edit.

Pack years

Immune

HLALOHAntigen pres.

DFSImmune edit.

Pack years

high evasion capacitylow evasion capacityunknown evasion capacity

increasing pack years smoked

0

Neo

Neo

1000

0

500

low immune evasion capacityhigh immune evasion capacity

All tumors, low immune evasion or high clonal neoAll tumors Low clonal neoantigen tumors

C

A

B

D E

0 400 800 12000.0

0.2

0.4

0.6

0.8

1.0

Time (days)

HR = 0.198 (0.092 − 0.427)logrank P = 4.9e−06

40 32 25 17 15 11 5 046 43 42 41 38 26 10 0

0 400 800 12000.0

0.2

0.4

0.6

0.8

1.0

Time (days)

HR = 0.30 (0.12 − 0.74)logrank P = 5e-03

40 32 25 17 15 11 523 21 20 19 17 10 3

Dise

ase

Free

Sur

vival

0 400 800 12000.0

0.2

0.4

0.6

0.8

1.0

Time (days)

HR = 0.27 (0.12 − 0.61)logrank P = 9e-04

49 40 33 25 23 17 7 034 32 31 30 28 19 7 0

Immune evasion capacity Immune evasion capacityImmune evasion capacity

High immune infiltration-- or --

No immune editingNo HLA LOHNo APC processing defect

Low immune evasion

Low/mixedimmune infiltration

Immune escape

High immune evasion

-- and --

{No immune escape

Immune editing /HLA LOH /APC processing defect{

Page 41: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

Lung adenocarcinoma Lung squamous cell carcinomacorrelation between purity and gene expression correlation between purity and gene expression

A B

random EPIC Danaher Rooney DavoliTIMER random EPIC Danaher Rooney DavoliTIMER

−0.5

0.0

0.5

Spe

arm

an’s

rho

Spe

arm

an’s

rho

Spe

arm

an’s

rho

Spe

arm

an’s

rho

0

100 * * * * *

−0.5

0.0

0.5

0

100 * * * * *

p<0.001 p<0.01 p<0.05p<0.05

ns

Figure S1

Lung adenocarcinoma Lung squamous cell carcinomacorrelation between gene CN and expression correlation between gene CN and expression

C D

−0.5

0.0

0.5

random EPIC Danaher Rooney Davoli0

100 * * * * *−0.5

0.0

0.5

random EPIC Danaher Rooney Davoli% g

enes

corr

elat

ed0

100 * * * * *

TIMER TIMER

−0.5 0 0.5

Davoli TIMERDanaher EPIC

rho

lung adeno.lung squam. * *

CD

8+ Cyt

Tcel

lTo

talT

ILC

D45

+D

C

M1/

M2

NK

Pro

/Ant

i Cyt

CD

8+/T

reg

Pro

CD

8+

CD

8+C

D4+

Imm

uneS

core DC

Treg

sN

K

CD

8+ Neu DC

CD

4+B

cell

Mac

Mac

CD

4+ NK

CD

8+E

ndo

CA

F

** ** * ** * * * *

xCell

Lung adenocarcinomaLung squamous cell carcinoma

* * ** * * *

FDR corrected p-value < 0.01FDR corrected p-value < 0.05**

*

% g

enes

corr

elat

ed

% g

enes

corr

elat

ed%

gen

esco

rrel

ated

E

Top six correlations from each method

p: 4.6e-09rho: 0.47

p: 4.8e-04rho: 0.39

p: 0.003rho: 0.44

p: 1.7e−02rho: 0.4

p: 8.4e−03rho: 0.42

p: 8.1e−04rho: 0.53

0 20 40 60 80

1

2

3

4

5

pathology TILs

CD

8+ D

anah

er

30 40 50 60 70 80

0

1

2

3

4

5

flow CD8+

CD

8+ D

anah

er

0 10 20 30 40 50 60

−1.0

−0.5

0.0

0.5

1.0

1.5

flow live cd8

CD

8+ D

anah

er e

nric

hmen

t

0 5 10 15

0

2

4

6

8

10

flow live cd8/treg

cd8/

treg

Dan

aher

Pathology estimated TILs Flow CD8+ T-cells TCRseq abundance Normalized flow CD8 Normalized flow CD8/Treg

CD8+ Danaherp: 4.6e−09 rho: 0.47

CD8+ Davolip: 1.2e−06 rho: 0.4

CD8+ xCellp: 1.3e−05 rho: 0.36

CD8+ TIMERp: 5.5e−02 rho: 0.16

CD8+ EPICp: 3.5e−01 rho: 0.08

CD

8+ E

stim

ate

pathology TILs

F

*

7 8 9 10 11 12 13

−0.5

0.0

0.5

1.0

1.5

total TCR (log)

CD

8+ D

anah

er (l

og)

p: 1.2e−06rho: 0.54

Page 42: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

Figure S2

TRACERx exome cohort (n=100 patients; 327 regions)(2017, Jamal-Hanjani)

Multi-region RNA-sequencing (n=64 patients; 164 regions) Pathology TIL estimates (n=83 patients; 234 regions)

TRACERx immune infiltration cohort (n=88 patients; 277 regions)

Excluded (n=36 patients; 139 regions):- RIN score < 5 (n=36 patients; 139 regions)

Excluded (n=17 patients; 69 regions):- Tumor used for sequencing (n=16 patients; 27 regions)- Too little tissue (n=20 patients; 34 regions)- Necrotic (n=2 patients; 3 regions)- Other (n=4 patients; 5 regions)

A CONSORT diagram

B Sample characteristics

Stage

Pack yearsRecurrence/Death

Number regions

Number regions (RNA)

Immune classification

No Yes13001a 1b 2 3 4 low hetero. high5 6 7 8Tumor Stage Pack-Years Recurrence or Death No. of Regions

2a 2b 3a 3bImmune classification

1

- Region failed exome analysis (n=19 patients, 24 regions)

TRACERx analyzed exome cohort (n=100 patients; 303 regions)

HistologyLung adenocarcinoma Lung squamous cell carcinoma Other

Page 43: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

lung adeno. lung squam.

1

2

3

4

Dan

aher

CD

8+ S

core

p: 1.3e−05

Figure S3

NSCLC by histology

Page 44: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

Figure S4

Lung adenocarcinomaLung adenocarcinoma Lung squamous cell carcinoma

immune high clusterimmune low clusterno RNAseq for region

immune high clusterimmune low clusterno RNAseq for region

A

ED

C

patient with low immune infiltratepatient with high immune infiltrate patient with heterogeneous immune infiltrate

patient without RNAseq

low hetero. high

Num

ber p

atie

nts

0

10

20

30

40

low hetero. high0

10

20

30

40

immune cluster immune cluster

path

olog

y es

timat

ed T

ILs

p: 8e-07 p: 0.05

p: 0.04 p: 0.04

path

olog

y es

timat

ed T

ILs

0

20

40

60

80

100

0

20

40

60

80

100

immuneclassimmuneclass

GF H

B

Immune classification Immune classification

RNAseq only RNAseq plus rescued TIL regions

lowhigh

acinarsolid

lepidiccribriform

papillaryNAother

Homo.immune

Hetero.immune

Lung adenocarcinoma patients

Num

ber p

atie

nts

0

5

10

15

20

25

30

35 p = 0.02

Heterogeneous pathologyHomogeneous pathology

Tum

or R

egion

s

CRUK

0004

CRUK

0005

CRUK

0017

CRUK

0018

CRUK

0029

CRUK

0041

CRUK

0046

CRUK

0061

0

4

8

Immune cluster Hist. subtype

paired t−test p: 0.04

Regions from heterogeneousTMB tumors

cd8

cd4

bcel

lcd

45cy

tode

ndm

ast

nkcd

56di

mnk tc

ells

th1

MDS

CCA

F

TAM

.M2

cd8cd4

bcellcd45

cytodend

mastnkcd56dim

nktcells

th1

MDSCCAF

TAM.M2

Dan

aher

mea

sure

sJi

ang

mea

sure

s

highlow low

low highlow low

low

1

2

3

4

CD

8+ s

core

(Dan

aher

)

0.5

1.0

1.5

2.0

2.5

Treg

sco

re (D

anah

er)

TILimmune.

low high

−0.08

−0.06

−0.04

−0.02

0.00

0.02

0.04

0.06

MD

SC (J

iang

)

low high

−0.04

−0.02

0.00

0.02

TAM

M2

(Jia

ng)

Low TMB High TMB

0.1

0.2

0.3

0.4

0.5

0.6

Tum

or re

gion

pur

ity

rho-1

1

low high

0

20

40

60

80

immune cluster

path

olog

y es

timat

ed T

ILs

p: 1.9e−05

Page 45: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

Figure S5

tumor with high score/TMB tumor with low score/TMB tumor with heterogeneous score/TMB

E

F

A

C D

0.5

1.0

2.0

5.0

10.0

20.0

50.0

100.0

TMB threshold

TMB

(mut

/Mb)

TMB +

−2

−1

0

1

2

3

TIDE thresholdTID

E Sc

ore

TIDEimmune immune

histologyimmune

TMB+TIDE

IPRES

p: 7e-04

Enrichment inhet. immune:*

p: 0.05p: 0.75

* only considering tumors with >1 RNAseq regiontumor with high immune tumor with low immune tumor with heterogeneous immunelung adeno. lung squam.

B

−2

−1

0

1

2

IPRES threshold

IPR

ES S

core

immune high clusterimmune low cluster

IPRES

immune high clusterimmune low cluster

4

5

6

7

8

9

10

IFN

g si

gnat

ure,

exp

ande

d

immuneFALSE TRUE

0.0

0.5

1.0

1.5

2.0

Immune heterogeneous tumor

Gre

ates

t dife

renc

e be

twee

n re

gion

sIF

Ng

sign

atur

e, e

xpan

ded

(Aye

rs)

p: 1.1e−02

NA

Page 46: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

Figure S6

D E

0.0 0.5 1.0 1.5 2.0

1

2

3

4

Tumor region diversity (Shannon) Tumor region diversity (Shannon)

CD

8+ D

anah

er

p: 0.035Spearman’srho: −0.22

0.0 0.5 1.0 1.5

0.5

1.0

1.5

2.0

2.5

3.0

3.5

CD

8+ D

anah

er

p: 0.907Spearman’srho: −0.02

Lung adenocarcinoma Lung squamous cell carcinoma

I

A

low heterogeneous high

p: 0.02

Immune classification (pathology TILs)region with low immune infiltrateregion with high immune infiltrate

Lung adenocarcinoma HG

p: 0.65rho: -0.04

p: 0.39rho: 0.09

Lung adeno.

Lung squam.

0.0

0.5

1.0

1.5

2.0

Tum

or re

gion

div

ersi

ty (S

hann

on)

KJ

F

Expressedgene

0.0

0.2

0.4

0.6

0.8

1.0

p: 5.5e−10OR: 1.3

0.0

0.2

0.4

0.6

0.8

1.0

Non−neoNeo

p: 3.5e−13OR: 1.4

Non-expressedgene

Expressedgene

Non-expressedgene

All neoantigens Strong neoantigens

Lung adeno. Lung squam.

0.8

0.9

1.0

1.1

Imm

unoe

ditin

g sc

ore

3 4 5 6

0.8

0.9

1.0

1.1

Number unique HLAs

Imm

unoe

ditin

g sc

ore

p: 2.1e−05Spearman’srho: 0.43

0.0 0.2 0.4 0.6 0.8 1.0

0

20

40

60

80

Tumor purity

Path

olog

y es

timat

ed T

ILs

Tumors with HLA−C LOH

Tumors without HLA−C LOH

inhibitorysignal

Heterozygous HLA-C C1/C2

inhibitorysignal

inhibitorysignal

inhibitorysignal

Homozygous HLA-C C1/C2

Heterozygous HLA-C C1/C2Homozygous HLA-C C1/C2

no HLA-C LOH

HLA-C LOH

NK cell

XFALSE TRUE

0.0

0.2

0.4

0.6

0.8

1.0

HLA -C C1/C2 heterozygosity

NK

cell

estim

ate

p: 6.2e−07

FALSE TRUE

p: 0.12

B C

0

0.5

1

1.5

p = 0.15 p = 2e-03 p = 0.25 p = 1e-03 p = 0.25 p = 0.04 p = 0.72 p = 0.80All High Hetero. Low

Odd

s ra

tio:

stro

ng n

eoan

tigen

depl

etio

n in

RN

A

HLA LOH HLA intact HLA LOH HLA intact HLA LOH HLA intact HLA LOH HLA intact

n.s.

0 5 10 15 20 25

2

4

6

8

Pairwise cn distance

Pairw

ise

imm

une

dist

ance

p: 1e−05Spearman’s rho: 0.45

p: 0.08Spearman’s rho: 0.23

Lung adeno.

Lung squam.

Page 47: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

HLA−AHLA−BHLA−C

CIITAIRF1

PSME1PSME2PSME3ERAP1ERAP2

CALRPDIA3

B2M

HLA−AHLA−BHLA−C

CIITAIRF1

PSME1PSME2PSME3ERAP1ERAP2

CALRPDIA3

B2M

Lung squam.

Figure S7

Lung adeno.

no alterationmutationcopy number loss

HLA

Antig

en p

res.

HLA

Antig

en p

res.

CRU

K008

6

CRU

K007

4

CRU

K006

8

CRU

K007

9

CRU

K006

9

CRU

K006

4

CRU

K008

3

CRU

K007

2

CRU

K007

3

CRU

K006

2

CRU

K007

6

CRU

K007

1

CRU

K007

0

CRU

K006

5

CRU

K007

8

CRU

K006

7

CRU

K008

2

CRU

K006

6

CRU

K008

4

CRU

K009

0

CRU

K007

5

CRU

K006

3

CRU

K008

9

CRU

K008

5

CRU

K008

8

CRU

K008

7

CRU

K007

7

CRU

K009

2

CRU

K008

1

CRU

K008

0

CRU

K009

3C

RUK0

091

CRU

K002

0C

RUK0

016

CRU

K003

9

CRU

K005

1C

RUK0

027

CRU

K006

0C

RUK0

024

CRU

K003

5C

RUK0

052

CRU

K004

7C

RUK0

026

CRU

K000

8

CRU

K000

1C

RUK0

017

CRU

K000

9

CRU

K000

3

CRU

K002

9C

RUK0

061

CRU

K004

4C

RUK0

004

CRU

K001

2C

RUK0

005

CRU

K004

1

CRU

K003

8

CRU

K001

8

CRU

K004

5

CRU

K004

6

CRU

K004

8C

RUK0

032

CRU

K001

0

CRU

K001

3

CRU

K002

8C

RUK0

002

CRU

K002

2

CRU

K003

7C

RUK0

055

CRU

K002

3

CRU

K003

6C

RUK0

042

CRU

K005

3

CRU

K003

1C

RUK0

034

CRU

K000

6C

RUK0

033

CRU

K005

7C

RUK0

025

CRU

K005

0

CRU

K001

5

CRU

K002

1

CRU

K003

0

CRU

K001

4C

RUK0

019

CRU

K005

8C

RUK0

011

CRU

K004

3C

RUK0

049

CRU

K000

7C

RUK0

040

CRU

K005

4C

RUK0

056

CRU

K005

9

high evasion capacitylow evasion capacityunknown evasion capacity

Page 48: Computational Health Informatics Program, Boston Children ... · associated with improved immunotherapy response 15. 12/57 NSCLC tumors with high TMB had at least one tumor region

Figure S8

100 200 300 400 5000

1

2

3

4

#ClonalNeo used as threshold

HRHR

number patients in high neo group

100 200 300 400 5000

1

2

3

4

#SubclonalNeo used as threshold

number patients in high neo group

100 200 300 4000.0

0.5

1.0

1.5

2.0

#ClonalNeo used as threshold

number patients in high neo group

0 50 100 150 200 250 300 3500

1

2

3

4

5

#SubclonalNeo used as threshold

number patients in high neo group

B

D

Clonal neoantigens, multiple thresholds

Subclonal neoantigens, multiple thresholds

non-significant association

significant association increasing number patients

1Q 3Q2Q

Lung adenocarcinoma Lung squamous cell carcinoma

Lung adenocarcinoma Lung squamous cell carcinoma

Lung adenocarcinoma Lung squamous cell carcinomaE Total neoantigens Multivariate analysis

Time (days) Time (days)0 200 400 600 800 1000 1200 1400

0.0

0.2

0.4

0.6

0.8

1.0

HR = 1.73 (0.58 − 5.17)logrank P = 0.32

15 14 14 14 13 10 4 046 41 35 30 28 18 6 0

0 200 400 600 800 1000 12000.0

0.2

0.4

0.6

0.8

1.0

HR = 1.03 (0.32 − 3.29)logrank P = 0.96

8 8 6 6 6 4 124 19 18 14 12 8 4

< upper quartile neoantigens≥ upper quartile neoantigens

Lung adenocarcinoma Lung squamous cell carcinomaA

Time (days)

Dise

ase

Free

Sur

vival

Dise

ase

Free

Sur

vival

Dise

ase

Free

Sur

vival

Time (days)

Clonal neoantigens

0 200 400 600 800 1000 1200 14000.0

0.2

0.4

0.6

0.8

1.0

HR = 4.7 ( 1.1 − 20.5)logrank P = 0.022

15 14 14 14 13 11 5 046 41 35 30 28 17 5 0

0 200 400 600 800 1000 12000.0

0.2

0.4

0.6

0.8

1.0

HR = 7.35 ( 0.96 − 56.60)logrank P = 0.025

8 8 8 8 8 5 224 19 16 12 10 7 3

Time (days) Time (days)

Lung adenocarcinoma Lung squamous cell carcinomaC Subclonal neoantigens

0 200 400 600 800 1000 1200 14000.0

0.2

0.4

0.6

0.8

1.0

HR = 1.67 (0.56 − 4.97)logrank P = 0.35

15 14 13 12 11 8 3 046 41 36 32 30 20 7 0

0 200 400 600 800 1000 12000.0

0.2

0.4

0.6

0.8

1.0

HR = 0.78 (0.24 − 2.49)logrank P = 0.67

8 8 6 5 3 2 124 19 18 15 15 10 4

F

Low evasion

Adjuvan .therapy

packyears

Stage

Sex

Age

Histology

TRUE

FALSE

Adjuvant

No adjuvant treatment

3

2b

2a

1b

1a

Male

Female

LUSC

LUAD

(N=46)

(N=40)

(N=27)

(N=66)

(N=93)

(N=12)

(N=10)

(N=12)

(N=34)

(N=25)

(N=58)

(N=35)

(N=93)

(N=32)

(N=61)

0.25

reference

0.44

reference

0.99

10.34

9.67

3.78

1.63

reference

0.62

reference

1.02

2.16

reference

(0.11 − 0.58)

(0.15 − 1.34)

(0.98 − 1.01)

(2.01 − 53.27)

(1.77 − 52.83)

(0.72 − 19.78)

(0.40 − 6.57)

(0.26 − 1.49)

(0.97 − 1.07)

(0.92 − 5.05)

0.001 **

0.15

0.541

0.005 **

0.009 **

0.116

0.492

0.289

0.49

0.076

# Events: 31; Global p−value (Log−Rank): 0.00020912 AIC: 224.95; Concordance Index: 0.79 0.1 0.2 0.5 1 2 5 10 20 50

Hazard ratio

** p < 0.01