Purpose:

PD-1 blockade plus chemotherapy has become the new standard of care in patients with untreated advanced non–small cell lung cancer (NSCLC), whereas predictive biomarkers remain undetermined.

Experimental Design:

We integrated clinical, genomic, and survival data of 427 NSCLC patients treated with first-line PD-1 blockade plus chemotherapy or chemotherapy from two phase III trials (CameL and CameL-sq) and investigated the predictive and prognostic value of HLA class I evolutionary divergence (HED).

Results:

High HED could predict significantly improved objective response rate (ORR), progression-free survival (PFS), and overall survival (OS) in those who received PD-1 blockade plus chemotherapy [in the CameL trial, ORR: 81.8% vs. 53.2%; P = 0.032; PFS: hazard ratio (HR), 0.47; P = 0.012; OS: HR, 0.40; P = 0.014; in the CameL-sq trial, ORR: 89.2% vs. 62.3%; P = 0.007; PFS: HR, 0.49; P = 0.005; OS: HR, 0.38; P = 0.002], but not chemotherapy. In multivariate analysis adjusted for PD-L1 expression and tumor mutation burden, high HED was independently associated with markedly better ORR, PFS, and OS in both trials. Moreover, the joint utility of HED and PD-L1 expression showed better performance than either alone in predicting treatment benefit from PD-1 blockade plus chemotherapy. Single-cell RNA sequencing of 58,977 cells collected from 11 patients revealed that tumors with high HED had improved antigen presentation and T cell–mediated antitumor immunity, indicating an inflamed tumor microenvironment phenotype.

Conclusions:

These findings suggest that high HED could portend survival benefit in advanced NSCLC treated with first-line PD-1 blockade plus chemotherapy.

See related commentary by Dimou, p. 4706

Translational Relevance

Despite the huge success observed in clinical trials evaluating first-line PD-1 blockade plus chemotherapy in advanced NSCLC, nearly half of patients do not respond. This divergence in terms of treatment benefit highlights a critical need to identify robust predictive biomarkers. In this study, we integrated clinical, genomic, and survival data of 427 patients with advanced NSCLC who received first-line PD-1 blockade plus chemotherapy from two previous phase III trials and investigated the predictive value of HED to PD-1 blockade plus chemotherapy. The findings demonstrate a strong association of high HED with better treatment outcomes in patients treated with first-line PD-1 blockade plus chemotherapy, but not with chemotherapy, indicating high HED would represent a potential biomarker to predict treatment outcomes of patients with untreated advanced NSCLC who received PD-1 blockade plus chemotherapy.

Immune-checkpoint inhibitors (ICI) targeting PD-1 or PD-L1 (PD-1 blockade) plus cytotoxic chemotherapy have become the first-line standard of care for patients with advanced non–small cell lung cancer (NSCLC) without driver gene alterations (1). Several global or regional phase III trials reported that the objective response rate (ORR) of PD-1 blockade plus chemotherapy was approximately 45% to 60% in the intention-to-treat (ITT) population. Yet, not all of them can benefit from this regimen (2). There are three officially approved predictive biomarkers to guide the clinical application of PD-1 blockade monotherapy, including PD-L1 expression, tumor mutational burden (TMB), and microsatellite instability status (3, 4). However, they failed to show the feasibility to predict response to PD-1 blockade plus chemotherapy in advanced NSCLC. Therefore, the identification of robust predictive biomarkers for PD-1 blockade plus chemotherapy is urgently needed in current clinical practice.

Unlike PD-1 blockade monotherapy, the application of PD-1 blockade plus chemotherapy is mainly based on the undetermined hypothesis that some chemotherapeutic drugs may stimulate T cell–mediated immunity via increasing antigen release from tumor cell death as well as elimination of some immunosuppressive cells, such as regulatory T cells (Treg) and myeloid-derived suppressor cell (MDSC; refs. 5–7). As an essential component of antigen presentation, human leukocyte antigen class I (HLA-I) plays a pivotal role in T cell–mediated antitumor immunity (8). Theoretically, a more diverse HLA-I repertoire would lead to the presentation of broader antigens, increasing the odds of presenting more immunogenic antigens and increasing the likelihood of response to ICIs. In fact, several elegant studies have reported that the germline HED, a measure of sequence divergence between alleles of HLA-I genotype that is associated with the diversity of immunopeptidomes and intratumoral T-cell receptor clonality, could predict the outcomes of PD-1 blockade monotherapy in patients with various solid tumors including NSCLC, melanoma, and gastrointestinal cancer (9, 10). However, some studies did not support its predictive value for immunotherapy alone (11, 12). Under this circumstance, we supposed that a more diverse HLA-I repertoire would be more potent to predict response to PD-1 blockade plus chemotherapy in advanced NSCLC.

In this study, we integrated clinical and HLA-I genotype data of 427 patients with advanced NSCLC who received first-line PD-1 blockade plus chemotherapy from two previous phase III trials and investigated the predictive value of HED to PD-1 blockade plus chemotherapy. To delineate the immune profiles of tumors with distinct HED, we conducted single-cell RNA sequencing (scRNA-seq) of 58,977 cells collected from an independent cohort of 11 individuals with NSCLC.

Study design

Two previously published cohorts (CameL and CameL-sq) of patients from phase III trials were identified. Briefly, CameL is a randomized, open-label, multicenter, phase III trial to compare the efficacy and safety of camrelizumab plus chemotherapy with chemotherapy alone as first-line treatment for patients with non-squamous NSCLC without EGFR and ALK alteration (13). CameL-sq is a randomized, placebo-controlled, double-blind phase III trial to investigate the efficacy and safety of camrelizumab or placebo plus chemotherapy as first-line treatment for patients with advanced squamous NSCLC (14). Both studies met the primary endpoints and have predefined the biomarker analysis (13, 14). In the CameL study, patients with the following criteria were eligible: ages 18 to 70 years, histologically or cytologically confirmed stage IIIB–IV non-squamous NSCLC without EGFR or ALK alterations, Eastern Cooperative Oncology Group performance status (ECOG PS) of 0 or 1, no previous systemic chemotherapy, at least one measurable lesion per Response Evaluation Criteria in Solid Tumors version 1.1 (RECIST v1.1), and a life expectancy of ≥3 months. Patients were excluded if they had untreated central nervous system (CNS) metastases and corticosteroid use within two weeks before study treatment was recorded. In the CameL-sq study, eligible patients were: ages 18 to 75 years, pathologically confirmed stage IIIB–IV squamous NSCLC, no previously systemic therapy for metastatic disease, at least one measurable lesion per RECIST v1.1, ECOG PS of 0 or 1, archival (within 12 months from first study dose) or fresh tumor tissues for biomarker testing. Key exclusion criteria were: patients with active or symptomatic CNS metastases, EGFR or ALK alterations, history or presence of autoimmune disease or interstitial pneumonia, and use of immunosuppressants within two weeks before study treatment. Patients with adequate pretreatment fresh or archival formalin-fixed paraffin-embedded (FFPE) tissue and peripheral blood samples were eligible. This study was approved by independent ethics committees/institutional review boards at each study site and conducted according to the Declaration of Helsinki, Guidelines for Good Clinical Practice, and local laws and regulations of China.

Sample collection

Fresh or archival FFPE baseline samples from the CameL and CameL-sq studies were collected before the protocol-defined treatments. Fresh biopsy tissues were snap-frozen in liquid nitrogen within 30 minutes. Pretreatment blood samples (8–10 mL) were collected in ethylene diamine tetraacetic acid (EDTA)-coated tubes (RRID:SCR_013311, BD Biosciences) and centrifuged at 1,800 × g for 10 minutes within two hours of collection to separate white blood cells.

DNA extraction

Fresh or FFPE tumor samples with ≥20% tumor cell contents were qualified and included. Genomic DNA from tumor tissues and peripheral blood cells were extracted with the QIAamp DNA FFPE Tissue Kit (RRID:SCR_008539, Qiagen) and DNeasy Blood and Tissue Kit (Qiagen) or GeneRead DNA FFPE Kit (Qiagen 180134) and Tguide S32 Magnetic Blood Genomic DNA Kit (RRID:SCR_023688, Tiangen) following the manufacturers’ protocols. DNA concentration was measured by Qubit dsDNA High-Sensitivity Assay Kit (RRID:SCR_008452, Thermo Fisher), whereas the quality of DNA was assessed by Agilent 2100 BioAnalyzer (RRID:SCR_019715, Agilent).

HLA-I genotyping and HED calculation

HLA-I typing of normal blood samples was performed using the OptiType tool (version 1.3.5; RRID:SCR_022279; ref. 15) with default parameter settings. HED was calculated according to previous studies (10, 16). Briefly, we first extracted the protein sequences of exons 2 and 3 of each allele of each individual HLA-I (HLA-A, HLA-B, and HLA-C) genotype from the IMGT/HLA database (RRID:SCR_002971), and then calculated divergence between allele sequences using Grantham distance metric (17), implemented by HLA-HED software (https://github.com/sunhuaibo/HLA-HED).
where |$i$| and |$j$| are the amino acids at a given position in the alignment; D is the distance between given two amino acids; |$c$|⁠, |$p$|⁠, and |$v$| represent composition, polarity, and molecular volume of the amino acid, respectively; |$a$|⁠, |$\beta$|⁠, and |$\gamma $| are constants. The final HED score was calculated by normalizing the value by the length of the alignment of two alleles. The mean HED was calculated as the mean divergence at HLA-A, HLA-B, and HLA-C. The HLA alleles data were collected from the published literature pertaining to The Cancer Genome Atlas (TCGA) cohort (18). HEDs were calculated as previously described above. Patients’ survival outcomes of TCGA lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC) cohorts were obtained from the cBioPortal database.

Loss of heterozygosity (LOH) in HLA

Following a previous report, we analyzed the LOH of HLA event of each tumor sample using the LOHHLA tool (RRID:SCR_023690, https://bitbucket.org/mcgranahanlab/lohhla/src/master/) with default parameter settings (19).

Library preparation of whole-exome sequencing

Thirty to 300 ng of genomic DNA was sheared to a length of approximately 200 base pairs (bp) by Covaris LE220 and library preparations were performed with KAPA Hyper Prep Kit (KAPA Biosystems). Libraries were quantified with quantitative PCR using the KAPA Library Quantification kit (KAPA Biosystems), and the size was determined using the Bioanalyzer 2100 (Agilent Technologies). Sequencing was performed on the Illumina HiSeq4000 platform using PE150 sequencing chemistry (Illumina).

Data processing and variants calling

Base calling was performed on bcl2fastq (RRID:SCR_015058) V.2.16.0.10 (20) to generate sequence reads in the FASTQ format. After removing low-quality reads by Trimmomatic (v0.36; RRID:SCR_011848; ref. 21), clean reads were aligned to the human reference genome (hg19, NCBI Build 37.5; RRID:SCR_012917) with the Burrows–Wheeler Aligner (BWA, version 0.7.17; RRID:SCR_010910). Then, the Picard toolkit (version 2.23.0, RRID:SCR_006525; ref. 22) was used to convert SAM files to compressed BAM files, which were then sorted according to chromosome coordinates. The Genome Analysis Toolkit (GATK, RRID:SCR_001876; ref. 23) was used for realignment. Single-nucleotide variants (SNV) and small insertions/deletions (InDel) were called via MuTect2 (RRID:SCR_000559; ref. 24) with tumor–normal mode. To avoid false-positive results, SNVs and InDels that appeared on the blacklist (including sequence-specific errors, repeat regions, segmental duplications, and lowly mappable regions recorded in ENCODE; RRID:SCR_006793) were removed. After annotation by ANNOVAR (RRID:SCR_012821; ref. 25), we filtered out variants in either introns or synonymous mutations. Somatic mutations with following criteria were used for analysis: (i) the sequencing depth was more than 100×; (ii) the variant allele frequency threshold of SNV was 4% and that of InDels was 5%. Furthermore, variants with a minor allele frequency ≥1% in the Exome Aggregation Consortium (ExAC, RRID:SCR_004068) and Genome Aggregation Database (gnomAD, RRID:SCR_014964) were removed. Mutations not recorded in the COSMIC (RRID:SCR_002260) database were filtered out. Somatic copy-number variations were identified using the CNVkit (v0.9.5, RRID:SCR_021917).

TMB calculation

As previously described (26), TMB was calculated by integrating the total number of somatic, base substitutions, coding, and InDel mutations per megabase of the genome examined. We first quantified the number of somatic nonsynonymous SNVs. Then, the value was extrapolated to the whole-exome using a validated algorithm (27). Only the regions with sequencing depth >100× after deduplication were used for TMB calculation. Germline genomic alterations in the Single-Nucleotide Polymorphism database or occurring with ≥2 counts in the ExAC database were not recorded.

Neoantigen prediction

Similar to our previous study (26), we calculated the neoantigen burden by using the following algorithm. First, we called the HLA-I alleles from the matched normal exome sequencing data using HLA-HD (v1.2.0.1, RRID:SCR_022285; ref. 28). Neoepitope presentation was then predicted for tumor-specific peptides of length 9–11 using the eluted-ligand mode of NetMHCpan-4.0 (RRID:SCR_018182; ref. 29). The following criteria were used to select the potential neoantigens for subsequent analysis: (i) derived from tumor-specific genomic alterations (including missense, inframe indels, frameshift, and fusions); (ii) high predicted affinity to HLA-I alleles [half maximal inhibitory concentration (IC50) < 500 nmol/L] with k-mer of 9–11 length; (iii) fold change >10 comparing to wild-type binding affinity. IEDB-recommended model (RRID:SCR_006604) was conducted to predict the HLA binding affinity using all variant-containing 9–11 mer for HLA-A/B/C binding estimations. HLA typing for patients was performed in silico using HLA-ATHLATES (RRID:SCR_023689; ref. 30) per the recommended algorithm.

scRNA-seq data integrating and clustering

Our previously published raw scRNA-seq data of patients with NSCLC were collected and integrated for comparison of immune cell landscape between tumors with high and low mean HED (31, 32). Their matched para-carcinoma normal tissues were used for HED calculation as previously described. Briefly, filtering of low-quality cells, data normalization, sample integration, clustering, and identification of cell types were performed using Seurat (version 4.1.1; ref. 33): (i) scRNA-seq data sets from 11 patients with NSCLC were loaded into R (version 4.1.3) using the CreateSeuratObject function of Seurat (RRID:SCR_016341) and merged together. (ii) Low-quality cells where >20% of transcripts derived from the mitochondria and either <500 expressed genes or >5,000 were filtered out. (iii) The NormalizeData function was performed to normalize raw counts. The top 2,000 highly variable genes were used for the following principal component analysis (PCA). (iv) The IntegrateData function was applied to remove batch effects among 11 patients. (v) The RunPCA and RunUMAP functions were used to reduce dimension. Subsequently, clustering cells by executing the FindNeighbors and FindClusters functions.

Proportion analysis

The Wilcoxon test was used to identify changes in the proportion of each cell type between tumors with high and low mean HED in total cells or immune cells in our cohorts. Significantly different cell types were then validated by using scRNA-seq data of a published cohort of 47 tumor samples from 36 patients with NSCLC who received PD-1–based treatments (34). A two-tailed Student t test was performed to calculate the significant differences of immune cells among pretreatment, posttreatment nonresponsive, and posttreatment responsive tumors.

Definition of differentially expressed genes (DEG)

DEGs between tumors with high and low mean HED in specific cell types were defined by performing the FindMarkers procedure of Seurat, remaining top 100 DEGs with both |Log2FC| > 0.5 and adjusted P < 0.01.

Gene ontology (GO) enrichment analyses

GO enrichments were performed by using the enrichGO function in the clusterProfiler package (version 3.12.0; refs. 35, 36) based on the highly expressed DEGs for the HEDhigh arm in specific cell types (P value cutoff = 0.01, q-value cutoff = 0.05). The AddModuleScore function of Seurat was applied to calculate the activity of each GO term in each cell.

Statistical analyses

The Kruskal–Wallis test was used for comparisons of HED distributions across individual HLA-I loci. Of the CameL and CameL-sq cohorts, cutoffs for high mean HED were determined using the top quartile, and low mean HED were defined as values less than the top quartile. For TCGA cohort, the absolute cutoff value of the CameL cohort was applied to the LUAD cohort, and the absolute cutoff value of CameL-sq was used for the LUSC cohort. High TMB was defined as values greater than or equal to 10 Muts/Mb, and positive PD-L1 expression was determined using the tumor proportional score greater than or equal to 1%. When combining mean HED and PD-L1 expression levels, patients with both high mean HED and positive PD-L1 expression were defined as the “Both high” group, whereas the other patients belonged to the “Others” group. When combining mean HED and TMB, patients were defined as the “Both high” group if their mean HED and TMB were both greater than or equal to the cutoff points for mean HED and TMB, and the others were defined as the “Others” group.

Survival curves were performed using Kaplan–Meier analysis. P values were calculated using the log-rank test. Hazard ratios (HR) and corresponding 95% confidence intervals (CI) were analyzed using stratified Cox proportional hazards model. For multivariable analyses, P values, HRs, and corresponding 95% CIs were analyzed using the Cox proportional hazards model and were visualized by “function ggforest.” The Fisher exact test was introduced to analyze the difference in response rate between high and low mean HED groups. The Spearman coefficients method was used for the correlation analysis. Two-side P < 0.05 was considered statistically significant. All statistical analyses were performed with R version 4.0.5 (RRID:SCR_001905).

Data availability

The data that support the findings of this study are available from the corresponding author upon reasonable request. All requests for raw data will be reviewed by the leading clinical site, Shanghai Pulmonary Hospital, Tongji University School of Medicine, and the study sponsor, Jiangsu Hengrui Pharmaceuticals, to check whether the request is subject to any intellectual property or confidentiality obligations. A signed data access agreement with the sponsor is required before accessing shared data. Source data are provided in this paper. All the relevant raw data were uploaded to the National Genomics Data Center. The accession numbers are HRA001180 and HRA004337 (https://ngdc.cncb.ac.cn/gsa-human/). We also listed the research resource identifier in Supplementary Table S4.

Study overview

To investigate the predictive value of HED to PD-1 blockade plus chemotherapy, we conducted integrated analyses using the CameL study (n = 174; ref. 13), and the CameL-sq study (n = 253; ref. 14; Fig. 1A; Supplementary Fig. S1 and Table 1). The clinical data of two studies used for biomarker association analyses were based on the updated analysis of both trials. Demographic and baseline parameters in the biomarker-evaluable population were generally analogous to those in the ITT population (Supplementary Tables S1 and S2). Treatment outcomes were also representative of the ITT population in both cohorts.

Figure 1.

Study overview and HED landscape. A, The biomarker data sets, experimental design, and key analysis objectives performed in the current study are summarized. The biomarker analyses are based on HED. To investigate the predictive value of HED, we used the HLA-I genotypes’ data of patients from previous phase III trials. High-quality pretreatment tumor tissues and peripheral blood samples were collected. The association between mean HED and treatment outcomes was evaluated. To better understand the molecular and immune profiles of tumors with distinct mean HED, we used whole-exome sequencing, multiplex immunofluorescence staining, and single-cell RNA sequencing to delineate the mutational and immune infiltration landscape. B, Distribution of patient mean HED across CameL cohort (CameL study mainly included patients with lung adenocarcinoma) and TCGA LUAD. C, Distribution of HED between patients treated with PD-1 blockade plus chemotherapy and chemotherapy in CameL cohort. D, Distributions of HED for each HLA-A, HLA-B, and HLA-C heterozygous genotype. In PD-1 blockade plus chemotherapy arm, HLA-A versus HLA-B (P = 0.490); HLA-A versus HLA-C (P < 0.001); HLA-B versus HLA-C (P < 0.001; two-sided Mann–Whitney test). In the chemotherapy arm, HLA-A versus HLA-B (P = 0.411); HLA-A versus HLA-C (P < 0.001); HLA-B versus HLA-C (P < 0.001; two-sided Mann–Whitney test). HED, HLA class I evolutionary divergence; PD-1 + chemo, PD-1 blockade plus chemotherapy; Chemo, chemotherapy; LUAD, lung adenocarcinoma.

Figure 1.

Study overview and HED landscape. A, The biomarker data sets, experimental design, and key analysis objectives performed in the current study are summarized. The biomarker analyses are based on HED. To investigate the predictive value of HED, we used the HLA-I genotypes’ data of patients from previous phase III trials. High-quality pretreatment tumor tissues and peripheral blood samples were collected. The association between mean HED and treatment outcomes was evaluated. To better understand the molecular and immune profiles of tumors with distinct mean HED, we used whole-exome sequencing, multiplex immunofluorescence staining, and single-cell RNA sequencing to delineate the mutational and immune infiltration landscape. B, Distribution of patient mean HED across CameL cohort (CameL study mainly included patients with lung adenocarcinoma) and TCGA LUAD. C, Distribution of HED between patients treated with PD-1 blockade plus chemotherapy and chemotherapy in CameL cohort. D, Distributions of HED for each HLA-A, HLA-B, and HLA-C heterozygous genotype. In PD-1 blockade plus chemotherapy arm, HLA-A versus HLA-B (P = 0.490); HLA-A versus HLA-C (P < 0.001); HLA-B versus HLA-C (P < 0.001; two-sided Mann–Whitney test). In the chemotherapy arm, HLA-A versus HLA-B (P = 0.411); HLA-A versus HLA-C (P < 0.001); HLA-B versus HLA-C (P < 0.001; two-sided Mann–Whitney test). HED, HLA class I evolutionary divergence; PD-1 + chemo, PD-1 blockade plus chemotherapy; Chemo, chemotherapy; LUAD, lung adenocarcinoma.

Close modal
Table 1.

Baseline characteristics of all included patients.

CameL trialCameL-sq trial
PD-1 + chemo groupChemo groupPD-1 + chemo groupChemo group
Mean HED high (n = 22)Mean HED low (n = 66)Mean HED high (n = 22)Mean HED low (n = 64)Mean HED high (n = 37)Mean HED low (n = 81)Mean HED high (n = 29)Mean HED low (n = 106)
Age, years (%) 
 <65 20 (90.9) 56 (84.8) 16 (72.7) 42 (65.6) 22 (59.5) 41 (50.6) 18 (62.1) 65 (61.3) 
 ≥65 2 (9.1) 10 (15.2) 6 (27.3) 22 (34.4) 15 (40.5) 40 (49.4) 11 (37.9) 41 (38.7) 
Sex (%) 
 Male 11 (50.0) 50 (75.8) 19 (86.4) 49 (76.6) 35 (94.6) 78 (96.3) 26 (89.7) 101 (95.3) 
 Female 11 (50.0) 16 (24.2) 3 (13.6) 15 (23.4) 2 (5.4) 3 (3.7) 3 (10.3) 5 (4.7) 
ECOG PS (%) 
 0 7 (31.8) 17 (25.8) 2 (9.1) 15 (23.4) 3 (8.1) 18 (22.2) 7 (24.1) 23 (21.7) 
 1 15 (68.2) 49 (74.2) 20 (90.9) 49 (76.6) 34 (91.9) 63 (77.8) 22 (75.9) 83 (78.3) 
Smoking history (%) 
 <400 cigarette-years or never 11 (50.0) 15 (22.7) 5 (22.7) 24 (37.5) 5 (13.5) 9 (11.1) 4 (13.8) 20 (18.9) 
 ≥400 cigarette-years 11 (50.0) 51 (77.3) 17 (77.3) 40 (62.5) 32 (86.5) 72 (88.9) 25 (86.2) 86 (81.1) 
Histopathologic type (%) 
 Squamous cell carcinomas 0 (0.0) 0 (0.0) 0 (0.0) 0 (0.0) 37 (100.0) 81 (100.0) 29 (100.0) 106 (100.0) 
 Adenocarcinoma 21 (95.5) 65 (98.5) 21 (95.5) 63 (98.4) 0 (0.0) 0 (0.0) 0 (0.0) 0 (0.0) 
 Other 1 (4.5) 1 (1.5) 1 (4.5) 1 (1.6) 0 (0.0) 0 (0.0) 0 (0.0) 0 (0.0) 
Disease stage (%) 
 III 3 (13.6) 12 (18.2) 5 (22.7) 8 (12.5) 12 (32.4) 22 (27.2) 10 (34.5) 30 (28.3) 
 IV 19 (86.4) 54 (81.8) 17 (77.3) 56 (87.5) 25 (67.6) 59 (72.8) 19 (65.5) 76 (71.7) 
 Brain metastasis 0 (0.0) 2 (3.0) 2 (9.1) 1 (1.6) 0 (0.0) 1 (1.2) 2 (6.9) 1 (0.9) 
PD-L1 TPS (%) 
 <1% 3 (13.6) 16 (24.2) 3 (13.6) 18 (28.1) 15 (40.5) 40 (49.4) 15 (51.7) 53 (50.0) 
 ≥1% 19 (86.4) 47 (71.2) 16 (72.7) 40 (62.5) 22 (59.5) 41 (50.6) 14 (48.3) 53 (50.0) 
 Not evaluable 0 (0.0) 3 (4.5) 3 (13.6) 6 (9.4) 0 (0.0) 0 (0.0) 0 (0.0) 0 (0.0) 
TMB, Muts/Mb (%) 
 ≥10 1 (4.5) 4 (6.1) 5 (22.7) 6 (9.4) 15 (40.5) 31 (38.3) 9 (31.0) 50 (47.2) 
 <10 21 (95.5) 62 (93.9) 17 (77.3) 58 (90.6) 22 (59.5) 50 (61.7) 20 (69.0) 56 (52.8) 
CameL trialCameL-sq trial
PD-1 + chemo groupChemo groupPD-1 + chemo groupChemo group
Mean HED high (n = 22)Mean HED low (n = 66)Mean HED high (n = 22)Mean HED low (n = 64)Mean HED high (n = 37)Mean HED low (n = 81)Mean HED high (n = 29)Mean HED low (n = 106)
Age, years (%) 
 <65 20 (90.9) 56 (84.8) 16 (72.7) 42 (65.6) 22 (59.5) 41 (50.6) 18 (62.1) 65 (61.3) 
 ≥65 2 (9.1) 10 (15.2) 6 (27.3) 22 (34.4) 15 (40.5) 40 (49.4) 11 (37.9) 41 (38.7) 
Sex (%) 
 Male 11 (50.0) 50 (75.8) 19 (86.4) 49 (76.6) 35 (94.6) 78 (96.3) 26 (89.7) 101 (95.3) 
 Female 11 (50.0) 16 (24.2) 3 (13.6) 15 (23.4) 2 (5.4) 3 (3.7) 3 (10.3) 5 (4.7) 
ECOG PS (%) 
 0 7 (31.8) 17 (25.8) 2 (9.1) 15 (23.4) 3 (8.1) 18 (22.2) 7 (24.1) 23 (21.7) 
 1 15 (68.2) 49 (74.2) 20 (90.9) 49 (76.6) 34 (91.9) 63 (77.8) 22 (75.9) 83 (78.3) 
Smoking history (%) 
 <400 cigarette-years or never 11 (50.0) 15 (22.7) 5 (22.7) 24 (37.5) 5 (13.5) 9 (11.1) 4 (13.8) 20 (18.9) 
 ≥400 cigarette-years 11 (50.0) 51 (77.3) 17 (77.3) 40 (62.5) 32 (86.5) 72 (88.9) 25 (86.2) 86 (81.1) 
Histopathologic type (%) 
 Squamous cell carcinomas 0 (0.0) 0 (0.0) 0 (0.0) 0 (0.0) 37 (100.0) 81 (100.0) 29 (100.0) 106 (100.0) 
 Adenocarcinoma 21 (95.5) 65 (98.5) 21 (95.5) 63 (98.4) 0 (0.0) 0 (0.0) 0 (0.0) 0 (0.0) 
 Other 1 (4.5) 1 (1.5) 1 (4.5) 1 (1.6) 0 (0.0) 0 (0.0) 0 (0.0) 0 (0.0) 
Disease stage (%) 
 III 3 (13.6) 12 (18.2) 5 (22.7) 8 (12.5) 12 (32.4) 22 (27.2) 10 (34.5) 30 (28.3) 
 IV 19 (86.4) 54 (81.8) 17 (77.3) 56 (87.5) 25 (67.6) 59 (72.8) 19 (65.5) 76 (71.7) 
 Brain metastasis 0 (0.0) 2 (3.0) 2 (9.1) 1 (1.6) 0 (0.0) 1 (1.2) 2 (6.9) 1 (0.9) 
PD-L1 TPS (%) 
 <1% 3 (13.6) 16 (24.2) 3 (13.6) 18 (28.1) 15 (40.5) 40 (49.4) 15 (51.7) 53 (50.0) 
 ≥1% 19 (86.4) 47 (71.2) 16 (72.7) 40 (62.5) 22 (59.5) 41 (50.6) 14 (48.3) 53 (50.0) 
 Not evaluable 0 (0.0) 3 (4.5) 3 (13.6) 6 (9.4) 0 (0.0) 0 (0.0) 0 (0.0) 0 (0.0) 
TMB, Muts/Mb (%) 
 ≥10 1 (4.5) 4 (6.1) 5 (22.7) 6 (9.4) 15 (40.5) 31 (38.3) 9 (31.0) 50 (47.2) 
 <10 21 (95.5) 62 (93.9) 17 (77.3) 58 (90.6) 22 (59.5) 50 (61.7) 20 (69.0) 56 (52.8) 

Abbreviations: HED, HLA class I evolutionary divergence; PD-1 + chemo, PD-1 blockade plus chemotherapy; ECOG PS, Eastern Cooperative Oncology Group performance status; PD-L1, programmed cell death ligand 1; TPS, tumor proportion score; TMB, tumor mutational burden.

HED landscape

We first delineated the landscape of HEDs at HLA-A, HLA-B, and HLA-C in the CameL study. Hierarchical clustering of HEDs showed that there were different clusters of high and low divergence between alleles (Supplementary Fig. S2), in line with previous reports (9, 10) and known interrelationships of the three loci. Mean HED distributions were similar between patients of the CameL and TCGA LUAD cohorts (Fig. 1B), and patients from PD-1 blockade plus chemotherapy and chemotherapy groups (Fig. 1C). Comparison of distribution patterns of HEDs for each HLA-A, HLA-B, and HLA-C heterozygous genotype showed that HLA-C pairwise divergences were significantly lower to HLA-A and HLA-B pairwise divergences (Fig. 1D), in accordance with previous studies reported HLA-C as the most recently evolved gene (37). There was no obvious difference in pairwise divergences between HLA-A and HLA-B alleles (Fig. 1D).

HEDhigh predicts outcomes of PD-1 blockade plus chemotherapy

Similar to the previous definition (10, 38), we defined “HEDhigh” as the mean HED greater than or equal to the top quartile, and “HEDlow” as the mean HED less than the top quartile. The demographic and baseline parameters in the HED-high group were generally analogous with those in the HED-low group in both treatment cohorts. First, we excluded the mean HED as a generally prognostic factor in both LUAD and LUSC by using TCGA data sets (Supplementary Fig. S3). Then, we surveyed the associations between mean HED and outcomes of PD-1 blockade plus chemotherapy in all included patients. The results showed that HEDhigh was correlated with better OS and PFS than HEDlow in patients who received PD-1 blockade plus chemotherapy, but not chemotherapy, in both cohorts (Supplementary Fig. S4). Meanwhile, HEDhigh also associated with numerically higher ORR to PD-1 blockade plus chemotherapy in the CameL study (P = 0.113; Supplementary Fig. S4).

Given the impact of HLA-I heterozygosity on the diversity of immunopeptidomes, we hypothesized that the predictive performance of mean HED could be improved in patients with fully heterozygous HLA-I genotypes. In fully heterozygous patients of the CameL cohort (n = 143), patients with HEDhigh had significantly longer OS (HR = 0.40, 95%CI = 0.19–0.85; P = 0.014) and PFS (HR = 0.47, 95% CI = 0.26–0.86; P = 0.012) than those with HEDlow in PD-1 blockade plus chemotherapy arm (Fig. 2A and B). In the HEDhigh group, patients treated with PD-1 blockade plus chemotherapy had significantly better OS (HR = 0.47, 95% CI = 0.20–1.09; P = 0.071) and PFS (HR = 0.36, 95% CI = 0.18–0.72; P = 0.003) than those treated with chemotherapy, whereas no significant difference on OS and PFS was observed in patients with HEDlow (Fig. 2A and B). The associations between HEDhigh and treatment outcomes were absent in the chemotherapy arm (OS: HR = 1.00, 95% CI = 0.53–1.86; P = 0.987; PFS: HR = 0.80, 95% CI = 0.46–1.39; P = 0.431). Moreover, patients with HEDhigh had a markedly better ORR than those with HEDlow in the PD-1 blockade plus chemotherapy arm (ORR: 81.8% vs. 53.2%; P = 0.032; Fig. 2C), whereas it was similar in the chemotherapy arm (ORR: 27.3% vs. 47.8%; P = 0.123; Fig. 2D). The predictive value of HEDhigh for PD-1 blockade plus chemotherapy held in multivariate analysis (OS: HR = 0.39, 95% CI = 0.18–0.85; P = 0.018; PFS: HR = 0.47, 95% CI = 0.26–0.88; P = 0.0.18; Fig. 2E) when adjusted for PD-L1 expression and TMB level, confirming that the effect of HEDhigh on survival benefit may be specific and independent of PD-L1 expression and TMB.

Figure 2.

HEDhigh predicts outcomes of PD-1 blockade plus chemotherapy. A, Association of HEDhigh with OS after PD-1 blockade plus chemotherapy or chemotherapy in the CameL cohort of patients fully heterozygous at HLA-I (two-sided log-rank test). Density plots show the distribution and cut point (top quartile) for mean HED used in Kaplan–Meier analysis. B, Association of HEDhigh with PFS after PD-1 blockade plus chemotherapy or chemotherapy in the CameL cohort of patients fully heterozygous at HLA-I (two-sided log-rank test). C, Comparison of response rates between HEDhigh and HEDlow groups in PD-1 blockade plus chemotherapy arm in the CameL cohort of patients fully heterozygous at HLA-I (two-sided Fisher exact test). D, Comparison of response rates between HEDhigh and HEDlow groups in chemotherapy arm in the CameL cohort of patients fully heterozygous at HLA-I (two-sided Fisher exact test). E, Forest plots depicting the significance of HEDhigh with PD-L1 expression and tissue TMB in the CameL cohort of patients fully heterozygous at HLA-I. P values were estimated by means of the Cox model in the multivariate analysis. F, Association of HEDhigh with OS after PD-1 blockade plus chemotherapy or chemotherapy in the CameL-sq cohort of patients fully heterozygous at HLA-I (two-sided log-rank test). Density plots show the distribution and cutoff point (top quartile) for mean HED used in Kaplan–Meier analysis. G, Association of HEDhigh with PFS after PD-1 blockade plus chemotherapy or chemotherapy in the CameL-sq cohort of patients fully heterozygous at HLA-I (two-sided log-rank test). H, Comparison of response rates between HEDhigh and HEDlow groups in PD-1 blockade plus chemotherapy arm in the CameL-sq cohort of patients fully heterozygous at HLA-I (two-sided Fisher exact test). I, Comparison of response rates between HEDhigh and HEDlow groups in the chemotherapy arm in the CameL-sq cohort of patients fully heterozygous at HLA-I (two-sided Fisher exact test). J, Forest plots depicting the significance of HEDhigh with PD-L1 expression and tissue TMB in the CameL-sq cohort of patients fully heterozygous at HLA-I. P values were estimated by means of the Cox model in the multivariate analysis. HR, hazard ratio; CI, confidence interval; PD-1+chemo, PD-1 blockade plus chemotherapy; Chemo, chemotherapy; TPS, tumor proportional score; TMB, tumor mutational burden. R, response, included patients with complete and partial response per RECIST v1.1; NR, not response included patients with stable disease and disease progression per RECIST v1.1.

Figure 2.

HEDhigh predicts outcomes of PD-1 blockade plus chemotherapy. A, Association of HEDhigh with OS after PD-1 blockade plus chemotherapy or chemotherapy in the CameL cohort of patients fully heterozygous at HLA-I (two-sided log-rank test). Density plots show the distribution and cut point (top quartile) for mean HED used in Kaplan–Meier analysis. B, Association of HEDhigh with PFS after PD-1 blockade plus chemotherapy or chemotherapy in the CameL cohort of patients fully heterozygous at HLA-I (two-sided log-rank test). C, Comparison of response rates between HEDhigh and HEDlow groups in PD-1 blockade plus chemotherapy arm in the CameL cohort of patients fully heterozygous at HLA-I (two-sided Fisher exact test). D, Comparison of response rates between HEDhigh and HEDlow groups in chemotherapy arm in the CameL cohort of patients fully heterozygous at HLA-I (two-sided Fisher exact test). E, Forest plots depicting the significance of HEDhigh with PD-L1 expression and tissue TMB in the CameL cohort of patients fully heterozygous at HLA-I. P values were estimated by means of the Cox model in the multivariate analysis. F, Association of HEDhigh with OS after PD-1 blockade plus chemotherapy or chemotherapy in the CameL-sq cohort of patients fully heterozygous at HLA-I (two-sided log-rank test). Density plots show the distribution and cutoff point (top quartile) for mean HED used in Kaplan–Meier analysis. G, Association of HEDhigh with PFS after PD-1 blockade plus chemotherapy or chemotherapy in the CameL-sq cohort of patients fully heterozygous at HLA-I (two-sided log-rank test). H, Comparison of response rates between HEDhigh and HEDlow groups in PD-1 blockade plus chemotherapy arm in the CameL-sq cohort of patients fully heterozygous at HLA-I (two-sided Fisher exact test). I, Comparison of response rates between HEDhigh and HEDlow groups in the chemotherapy arm in the CameL-sq cohort of patients fully heterozygous at HLA-I (two-sided Fisher exact test). J, Forest plots depicting the significance of HEDhigh with PD-L1 expression and tissue TMB in the CameL-sq cohort of patients fully heterozygous at HLA-I. P values were estimated by means of the Cox model in the multivariate analysis. HR, hazard ratio; CI, confidence interval; PD-1+chemo, PD-1 blockade plus chemotherapy; Chemo, chemotherapy; TPS, tumor proportional score; TMB, tumor mutational burden. R, response, included patients with complete and partial response per RECIST v1.1; NR, not response included patients with stable disease and disease progression per RECIST v1.1.

Close modal

We next validate the predictive value of HEDhigh in fully heterozygous patients of the CameL-sq study (n = 196; ref. 14). Patients with HEDhigh had dramatically prolonged OS (HR = 0.38, 95% CI = 0.21–0.71; P = 0.002) and PFS (HR = 0.49, 95% CI = 0.29–0.81; P = 0.005) than those with HEDlow in PD-1 blockade plus chemotherapy arm (Fig. 2F and G). Patients with HEDhigh received PD-1 blockade plus chemotherapy had markedly prolonged OS (HR = 0.40, 95% CI = 0.20–0.80; P = 0.007) and PFS (HR = 0.08, 95% CI = 0.04–0.18; P < 0.001) than those treated with chemotherapy, whereas it was similar between PD-1 blockade plus chemotherapy and chemotherapy groups in patients with HEDlow (Fig. 2F and G). Superior ORR was observed in patients with HEDhigh than in those with HEDlow in PD-1 blockade plus chemotherapy arm (89.2% vs. 62.3%; P = 0.007; Fig. 2H) whereas it was similar in the chemotherapy arm (58.6% vs. 47.1%; P = 0.378; Fig. 2I). Multivariate analysis also showed the significantly predictive performance of HEDhigh for PD-1 blockade plus chemotherapy (Fig. 2J), further supporting that HEDhigh is a reliable predictive biomarker to PD-1 blockade plus chemotherapy in untreated advanced NSCLC, especially those with fully heterozygous HLA-I genotypes.

Previously, Lu and colleagues reported that HED of HLA-A, HLA-B, or HLA-C had a distinct impact on predicting ICI-treated outcomes in gastrointestinal cancer, and HLA-B HED was correlated with improved clinical benefit after ICI treatment (9). Therefore, we evaluated the effect of HED of HLA-A, HLA-B, or HLA-C on treatment outcomes. As shown in Supplementary Figs. S5 and S6, each of HLA-A, HLA-B, or HLA-C HED cannot independently and perfectly predict the treatment response and outcomes of PD-1 blockade plus chemotherapy in both cohorts.

Considering the predictive robustness of mean HED, we then assessed the effect of distinct cutoffs of mean HED on treatment outcome prediction. Generally, a negative relationship was observed between mean HED and HR of PFS, suggesting that an increase in mean HED corresponds to improved survival benefit (Supplementary Fig. S7A and S7B). Interestingly, although several cutoffs of mean HED showed statistical significance to predict treatment outcomes, the top quartile seemed to be the optimal cutoff to stratify patients into groups with different clinical outcomes in both cohorts. In addition, previous studies revealed that HLA-I LOH is one of the key immune-evade mechanisms and negative predictors of outcomes after ICI treatment in NSCLC (19, 39). We also explored the associations between HLA-I LOH and response to PD-1 blockade plus chemotherapy. The results showed that HLA-I LOH cannot predict treatment response and outcomes of PD-1 blockade plus chemotherapy in both the cohorts (Supplementary Fig. S7C and S7F).

Taken together, these data highlight the importance of mean HED and suggest that HEDhigh may portend survival benefit in advanced NSCLC patients treated with first-line PD-1 blockade plus chemotherapy.

HEDhigh associated with long-term benefit of PD-1 blockade plus chemotherapy

As we mentioned above, the increasing mean HED was correlated with longer PFS, suggesting a “dose-dependent manner” between mean HED and treatment benefit. To confirm this association, we utilized the STEPP approach to investigate the 2-year PFS probability along the mean HED range according to a previous publication (38). In both cohorts, we observed the nearly linear association between mean HED and improved 2-year PFS probability in the PD-1 blockade plus chemotherapy arm (Fig. 3AB). However, higher mean HED was not associated with improved outcome in the chemotherapy arm. Notably, the threshold of these benefits was slightly different between CameL and CameL-sq cohorts. Collectively, these data suggest that HEDhigh could predict long-term treatment benefits of PD-1 blockade plus chemotherapy in advanced NSCLC.

Figure 3.

HEDhigh predicts the long-term benefit of PD-1 blockade plus chemotherapy. A, STEPP showing the 2-year progression-free probability across the mean HED range in the CameL cohort. Higher HED was correlated with improved outcomes in the PD-1 blockade plus chemotherapy arm but not in the chemotherapy arm. B, STEPP showing the 2-year progression-free probability across the mean HED range in the CameL-sq cohort. Higher HED was correlated with improved outcomes in PD-1 blockade plus chemotherapy arm. Because few patients in the chemotherapy arm of the CameL-sq cohort had more than 2-year PFS, STEPP cannot be conducted. PFS, progression-free survival; PD-1 + chemo, PD-1 blockade plus chemotherapy; Chemo, chemotherapy.

Figure 3.

HEDhigh predicts the long-term benefit of PD-1 blockade plus chemotherapy. A, STEPP showing the 2-year progression-free probability across the mean HED range in the CameL cohort. Higher HED was correlated with improved outcomes in the PD-1 blockade plus chemotherapy arm but not in the chemotherapy arm. B, STEPP showing the 2-year progression-free probability across the mean HED range in the CameL-sq cohort. Higher HED was correlated with improved outcomes in PD-1 blockade plus chemotherapy arm. Because few patients in the chemotherapy arm of the CameL-sq cohort had more than 2-year PFS, STEPP cannot be conducted. PFS, progression-free survival; PD-1 + chemo, PD-1 blockade plus chemotherapy; Chemo, chemotherapy.

Close modal

The predictive value of HEDhigh was irrespective of TMB

To evaluate the combined effect of mean HED and other known predictive biomarkers for immunotherapy, including PD-L1 expression or TMB on response to PD-1 blockade plus chemotherapy, we first examined the correlation between mean HED and PD-L1 expression or TMB. Mean HED did not correlate with PD-L1 expression or TMB (Fig. 4A and B) in the CameL cohort, whereas mean HED showed mild correlation with PD-L1 expression but not TMB in the CameL-sq cohort (Supplementary Fig. S8A and S8B). No significant correlation was detected between mean HED and neoantigen burden (Fig. 4C). We also checked the associations between mean HED at each HLA-I locus and PD-L1 expression, TMB, or neoantigen burden, and found no correlation among them (Supplementary Fig. S9). When stratifying patients with fully heterozygous HLA-I genotypes by mean HED and TMB level, we found that the joint utility of both HEDhigh and high TMB showed an improved effect on the survival benefit of PD-1 blockade plus chemotherapy in the CameL cohort but not in the CameL-sq cohort, and no improved effect on treatment response in both cohorts (Supplementary Fig. S10). Of note, HEDhigh could predict the survival benefit in both high and low TMB groups (Supplementary Fig. S11), indicating that the predictive value of HEDhigh for PD-1 blockade plus chemotherapy was greatly irrespective of the TMB level.

Figure 4.

Combination of mean HED and PD-L1 expression showed better predictive performance. A, Correlation of mean HED with PD-L1 TPS (P = 0.157; two-sided Spearman correlation). B, Correlation of mean HED with TMB (P = 0.229; two-sided Kendall rank correlation). C, Correlation of mean HED with neoantigen burden (P = 0.938; two-sided Kendall rank correlation). D, OS comparisons between patients with both high and others after PD-1 blockade plus chemotherapy or chemotherapy among fully heterozygous patients (two-sided log-rank test). When combining mean HED and PD-L1 expression levels, patients with both HEDhigh and positive PD-L1 expression were defined as the “Both high” group, whereas the other patients belonged to the “Others” group. E, PFS comparisons between patients with both high and others after PD-1 blockade plus chemotherapy or chemotherapy among fully heterozygous patients (two-sided log-rank test). F, Response rates comparison between patients with both high and others after PD-1 blockade plus chemotherapy among fully heterozygous patients (two-sided Fisher exact test). G, Response rates comparison between patients with both high and others after chemotherapy among fully heterozygous patients (two-sided Fisher exact test). TPS, tumor proportional score; TMB, tumor mutational burden; PD-1 + chemo, PD-1 blockade plus chemotherapy; Chemo, chemotherapy; R, response, included patients with complete and partial response per RECIST v1.1; NR, not response included patients with stable disease and disease progression per RECIST v1.1.

Figure 4.

Combination of mean HED and PD-L1 expression showed better predictive performance. A, Correlation of mean HED with PD-L1 TPS (P = 0.157; two-sided Spearman correlation). B, Correlation of mean HED with TMB (P = 0.229; two-sided Kendall rank correlation). C, Correlation of mean HED with neoantigen burden (P = 0.938; two-sided Kendall rank correlation). D, OS comparisons between patients with both high and others after PD-1 blockade plus chemotherapy or chemotherapy among fully heterozygous patients (two-sided log-rank test). When combining mean HED and PD-L1 expression levels, patients with both HEDhigh and positive PD-L1 expression were defined as the “Both high” group, whereas the other patients belonged to the “Others” group. E, PFS comparisons between patients with both high and others after PD-1 blockade plus chemotherapy or chemotherapy among fully heterozygous patients (two-sided log-rank test). F, Response rates comparison between patients with both high and others after PD-1 blockade plus chemotherapy among fully heterozygous patients (two-sided Fisher exact test). G, Response rates comparison between patients with both high and others after chemotherapy among fully heterozygous patients (two-sided Fisher exact test). TPS, tumor proportional score; TMB, tumor mutational burden; PD-1 + chemo, PD-1 blockade plus chemotherapy; Chemo, chemotherapy; R, response, included patients with complete and partial response per RECIST v1.1; NR, not response included patients with stable disease and disease progression per RECIST v1.1.

Close modal

Combination of mean HED and PD-L1 expression showed better predictive performance

To evaluate the impact of HEDhigh combined with PD-L1 expression on survival benefit and treatment response, we then stratified patients with fully heterozygous HLA-I genotypes by mean HED and PD-L1 expression. The results showed that the combination of HEDhigh and positive PD-L1 expression showed better predictive performance for both OS (HR = 0.29, 95% CI = 0.12–0.69; P = 0.003; Fig. 4D) and PFS (HR = 0.41, 95% CI = 0.22–0.80; P = 0.006; Fig. 4E) in the PD-1 blockade plus chemotherapy arm, as evidenced by reduced HR and corresponding P values. The combined effect was absent in the chemotherapy arm (Fig. 4D and E). Furthermore, ORR was significantly better in patients with HEDhigh and positive PD-L1 expression in the PD-1 blockade plus chemotherapy arm (ORR: 84.2% vs. 52.1%; P = 0.025; Fig. 4F) whereas it was similar in the chemotherapy arm (ORR: 37.5% vs. 45.7%; P = 0.771; Fig. 4G). The combined effect of mean HED and PD-L1 expression on survival benefit after PD-1 blockade plus chemotherapy was further confirmed in the CameL-sq cohort, which also showed reduced HR and corresponding P values of both OS (HR = 0.24, 95% CI = 0.10–0.62; P = 0.001; Supplementary Fig. S8C) and PFS (HR = 0.34, 95% CI = 0.18–0.65; P = 0.001; Supplementary Fig. S8D). Analogously, ORR was dramatically higher in patients with HEDhigh and positive PD-L1 expression in PD-1 blockade plus chemotherapy arm (ORR: 89.2% vs. 62.2%; P = 0.007; Supplementary Fig. S8E) and similar in the chemotherapy arm (ORR: 58.6% vs. 47.1%; P = 0.378; Supplementary Fig. S8F).

Immune profiles of tumors with HEDhigh versus HEDlow

To determine the immune profiles of tumors with distinct HEDs, we analyzed a large scRNA-seq data of 58,977 cells collected from 11 individuals with NSCLC (Supplementary Table S3). Dimensionality reduction with t-distributed stochastic neighborhood embedding (t-SNE) and graph-based clustering identified eight major cell subsets, including T, B, natural killer like T, myeloid, fibroblast, endothelial, mast, and cancer cells (Fig. 5A; Supplementary Fig. S12A). We found an increased number of T and B cells in the HEDhigh group, whereas the proportion of myeloid and cancer cells was significantly lower in the HEDhigh group than in the HEDlow group (Fig. 5B). We further identified 14 immune cell subsets using graph-based clustering (Fig. 5C; Supplementary Figs. S12B and S13A and S13B). We observed that patients with HEDhigh had a significantly higher proportion of CD4+ central memory T (Tcm), CD4+ Th1-like, and follicular B cells (Fig. 5D). Pathway enrichment analyses showed a strong enrichment of antigen presentation and immune response in CD4+ Tcm, CD4+ Th1-like, and follicular B cells from the HEDhigh group (Fig. 5EG and I). However, several immunosuppressive cells, including classic dendritic cell type 2 (cDC2), monocyte, macrophage, neutrophil, and mast cells, was significantly lower in the HEDhigh group (Fig. 5D and H). These significantly different cells between HEDhigh and HEDlow groups had distinct transcriptomic features and enriched pathways (Supplementary Fig. S13E–S13H).

Figure 5.

Immune profiles of tumors with HEDhigh versus HEDlow. A, t-SNE plots show 58,977 single cells from 11 NSCLC patients. Points are color-coded by cell type (left), HED (top right), and included patients (bottom right). B, Comparisons of the proportion of specific cell type in total cells between patients with HEDhigh and HEDlow (median ± quartiles for 4 HEDhigh and 7 HEDlow patients; two-sided Wilcoxon test). C, t-SNE plot shows cell types identified from 40,400 immune cells. D, Comparisons of the proportion of specific cell type in immune cells between patients with HEDhigh and HEDlow (median ± quartiles for 4 HEDhigh and 7 HEDlow patients; two-sided Wilcoxon test). E–H, Bubble charts show the expression of selected differentially expressed genes (DEGs) between HEDhigh and HEDlow patients (left). Bar charts show 15 significantly enriched GO terms of DEGs highly expressed in CD4+ Tcm (E), CD4+ Th1-like (F), Follicular B (G), and cDC2 (H) in HEDhigh patients. I, Heat maps show the activity of 50 GO terms associated with immunity in CD4+ Tcm, CD4+ Th1-like, Follicular B, and cDC2, respectively. t-SNE, t-distributed stochastic neighborhood embedding; cDC2, classic dendritic cell type 2; pDC, plasmacytoid DCs; NKT, natural killer like T; GO, Gene Ontology.

Figure 5.

Immune profiles of tumors with HEDhigh versus HEDlow. A, t-SNE plots show 58,977 single cells from 11 NSCLC patients. Points are color-coded by cell type (left), HED (top right), and included patients (bottom right). B, Comparisons of the proportion of specific cell type in total cells between patients with HEDhigh and HEDlow (median ± quartiles for 4 HEDhigh and 7 HEDlow patients; two-sided Wilcoxon test). C, t-SNE plot shows cell types identified from 40,400 immune cells. D, Comparisons of the proportion of specific cell type in immune cells between patients with HEDhigh and HEDlow (median ± quartiles for 4 HEDhigh and 7 HEDlow patients; two-sided Wilcoxon test). E–H, Bubble charts show the expression of selected differentially expressed genes (DEGs) between HEDhigh and HEDlow patients (left). Bar charts show 15 significantly enriched GO terms of DEGs highly expressed in CD4+ Tcm (E), CD4+ Th1-like (F), Follicular B (G), and cDC2 (H) in HEDhigh patients. I, Heat maps show the activity of 50 GO terms associated with immunity in CD4+ Tcm, CD4+ Th1-like, Follicular B, and cDC2, respectively. t-SNE, t-distributed stochastic neighborhood embedding; cDC2, classic dendritic cell type 2; pDC, plasmacytoid DCs; NKT, natural killer like T; GO, Gene Ontology.

Close modal

Despite the huge success observed in clinical trials evaluating first-line PD-1 blockade plus chemotherapy in advanced NSCLC, not all of them can benefit from this regimen. This divergence in terms of treatment benefit highlights a critical need to identify robust predictive biomarkers. Given the potential mechanism of the synergistic antitumor effect of PD-1 blockade plus chemotherapy that builds upon the increased tumor-related antigen release by some chemotherapeutic drugs (5), this study investigated the predictive significance of HED for PD-1 blockade plus chemotherapy in untreated advanced NSCLC from two phase III trials. Our results showed that HEDhigh was associated with significantly better treatment response, survival and 2-year PFS probability in NSCLC patients treated with first-line PD-1 blockade plus chemotherapy, especially in those with fully heterozygous HLA-I genotypes, but not in the chemotherapy group. We also found that a combination of mean HED and PD-L1 expression showed better predictive performance. Using scRNA-seq of untreated NSCLC, we found that patients with HEDhigh were associated with improved antigen presentation and antitumor immunity, further supporting HEDhigh as a predictive biomarker in this setting.

Previously, several studies investigated the predictive value of HED for PD-1 blockade monotherapy. In 2019, Chowell and colleagues reported the first evidence that patients with HEDhigh received PD-1 blockade monotherapy showed a longer OS in melanoma and NSCLC (10). They further found that HEDhigh was associated with both improved clinical benefit and durability of response in advanced renal cell carcinoma patients treated with PD-1 blockade plus lenvatinib (38). Then, Lu and colleagues reported that high HLA-B HED was correlated with a better OS compared with the low HLA-B HED subgroup in gastrointestinal cancer (9). Nevertheless, controversial findings were reported in two studies with large samples. The first group of investigators integrated patients across 17 pembrolizumab clinical trials and reported that HLA-I diversity including HED was not associated with response to pembrolizumab (11). The second one performed a meta-analysis investigating the association between germline HLA-I heterozygosity and HED and outcomes with PD-1 blockade among various solid tumors and also reported no association with response (12). This inconsistency might be due to the different patient distribution. For example, nearly all of the patients are of European ancestry and a majority of them with PD-L1–positive expression in the pembrolizumab clinical trials (11). Moreover, most of them received pembrolizumab monotherapy in the later-line setting, which could not represent the whole population. In addition, they included a mixture of patients from distinct trials across a long-time interval, which would have an impact on the homogeneous estimation of survival benefit. Nowadays, PD-1 blockade plus chemotherapy have been established as a new standard of care in various solid tumors including NSCLC. Considering chemotherapy could enhance immunogenicity via increasing tumor antigen release, the predictive value of HED needs to be further reevaluated for PD-1 blockade plus chemotherapy in advanced NSCLC.

To our best knowledge, the current study first investigated the predictive significance of HED for PD-1 blockade plus chemotherapy in patients with untreated advanced NSCLC from two phase III trials. Our results showed that HEDhigh was associated with significantly better response and survival benefit in NSCLC patients treated with first-line PD-1 blockade plus chemotherapy in both trials, especially in patients with fully heterozygous HLA-I genotypes. We also found that HEDhigh was associated higher landmark time probability of 2-year PFS. However, it did not affect the treatment outcome in the chemotherapy group. Multivariate analysis demonstrated that HEDhigh was a strong determinant of efficacy for PD-1 blockade plus chemotherapy. Currently, both the approved companion diagnostics of PD-L1 expression and TMB cannot be used for predicting response to PD-1 blockade plus chemotherapy in advanced NSCLC. Our study was the first one to reveal HED as the potential predictor for first-line PD–1 blockade plus chemotherapy in advanced NSCLC, though the robustness of its predictive value remains to be investigated. More importantly, our finding highlights the necessity of identifying distinct predictive biomarkers regarding different immunotherapeutic regimens.

The effect of the combinatorial utility of mean HED and TMB on survival benefits from ICI treatment has been demonstrated to be superior to either alone in melanoma and gastrointestinal cancer (10, 40), suggesting a rational combination of different potential biomarkers could improve the predictive performance. We also explored the combined effect of mean HED and PD-L1 expression or TMB on response to PD-1 blockade plus chemotherapy. The results showed that the joint utility of HEDhigh and PD-L1 expression showed better performance than either alone in predicting treatment benefits from this combination. Patients with both high HED and positive PD-L1 expression are most likely to benefit from PD-1 blockade plus chemotherapy, underscoring the rationale of the combinatorial utility of mean HED and PD-L1 expression in advanced NSCLC patients treated with first-line PD-1 blockade plus chemotherapy. Herein, both HED and PD-L1 expression should be taken into consideration in NSCLC in future clinical trials.

Having noticed the potential predictive value of mean HED, we are eager to understand the immune profiles of tumors with HEDhigh versus HEDlow. We adopted the scRNA-seq analysis of pretreated tumors with available HLA-I genotypes’ data and found that tumors with HEDhigh had markedly higher infiltration of CD4+ Tcm, CD4+ Th1-like, and follicular B cells, whereas cDC2, monocyte, macrophage, neutrophil, and mast cells were significantly lower in the HEDhigh group. CD4+ Tcm, CD4+ Th1-like, and follicular B cells were previously found to be associated with improved T cell–mediated immunity (41, 42), whereas cDC2, monocyte, macrophage, neutrophil, and mast cells were immunosuppressive in most cases (43–45). These findings indicate that tumors with HEDhigh would possess an immunosupportive TME phenotype. Interestingly, our further analysis revealed strong enrichment of antigen presentation and immune response in CD4+ Tcm, CD4+ Th1-like, and follicular B cells from the HEDhigh group. Importantly, CD4+ Tcm and CD4+ Th1-like cells were found to be significantly increased in responsive tumors after PD-1 blockade plus chemotherapy in a recent study (Supplementary Fig. S13C and S13D; ref. 34). Collectively, these data revealed that tumors with HEDhigh were associated with improved antigen presentation and T cell–mediated antitumor immunity, supporting it as a potential predictor for PD-1 blockade plus chemotherapy in NSCLC.

There were several limitations of this work. First, despite we analyzed the largest cohort of NSCLC patients treated with first-line PD-1 blockade plus chemotherapy to identify the potential biomarker hitherto, the current study was a retrospective analysis with hypothesis generating in nature. Hence, we should cautiously interpret these findings until they are validated in prospective, standardized biomarker studies. Second, all of the included patients were Asians. Whether HEDhigh was also a reliable predictor of PD-1 blockade plus chemotherapy in non-Asian ancestry patients with distinct HLA-I genotypes and immunophenotypes remained unknown. Third, we utilized the top quartile as the cutoff to define HEDhigh versus HEDlow groups. Although the cutoff is popular in prior publications and helpful for us to clarify the relevant investigations due to its briefness, it may not be optimal. Fourth, we only focused on the genetic variables to predict treatment benefit and did not evaluate the tumor immune microenvironment features. The integration of multiple-dimensional information would be valuable for the identification of robust biomarkers. Fifth, we did not observe the robust predictive or prognostic value of TMB in this cohort by using 10 Muts/Mb as the cutoff. Although the definition and cutoff of TMB are popular, the use of 10 Muts/Mb cutoff was mostly applied to commercially targeted gene panels, and the data of this study were generated by whole-exome sequencing. Considering the great discrepancy of the test platform, process, analysis algorithm, and definitions, we still need larger populations to validate the predictive and/or prognostic value of TMB in NSCLC received first-line PD-1 blockade plus chemotherapy. Finally, although we performed mIF and scRNA-seq to depict the immune profiles of tumor with distinct HED, we did not unravel the detailed biological explanations. The scRNA-seq data were generated mainly from patients with early-stage NSCLC without driver gene status, PD-L1 expression, and TMB level, so there may be significant confounders in terms of late-stage lung cancers with HED high and immune cell landscapes. Future clinical and translational research in this field would provide mechanistic insights into how HED could improve the T cell–mediated tumor control and predict the survival benefit of NSCLC patients who received first-line PD-1 blockade plus chemotherapy.

In conclusion, HEDhigh represents a potential biomarker to predict the response and survival outcomes of patients with untreated advanced NSCLC who received PD-1 blockade plus chemotherapy, especially in those with fully heterozygous HLA-I genotypes. Further investigation in prospective studies would be helpful to optimize PD-1 blockade plus chemotherapy in patients with untreated advanced NSCLC who are more likely to derive benefit. Considering HED can be analyzed from normal tissue (e.g., peripheral blood cells) DNA sequencing, it would be more convenient for daily clinical practice when ample tissue samples were difficult to obtain.

C. Zhou reports honoraria as a speaker from Roche, Lily China, Boehringer Ingelheim, Merck, Hengrui, Qilu, Sanofi, Merck Sharp & Dohme, Innovent Biologics, C-Stone, Luye Pharma, TopAlliance Biosciences, and Amoy Diagnostics and advisor fees from Innovent Biologics, Hengrui, Qilu, and TopAlliance Biosciences. No disclosures were reported by the other authors.

T. Jiang: Conceptualization, resources, data curation, software, formal analysis, supervision, funding acquisition, validation, investigation, visualization, methodology, writing–original draft, project administration, writing–review and editing. Q. Jin: Resources, data curation, software, formal analysis, funding acquisition, validation, visualization, methodology. J. Wang: Resources, data curation, software, formal analysis, supervision, visualization, methodology. F. Wu: Resources, data curation, software, formal analysis, supervision, investigation, visualization. J. Chen: Resources, data curation, software, formal analysis, supervision. G. Chen: Resources, data curation, software, formal analysis. Y. Huang: Resources, data curation, software, formal analysis. J. Chen: Resources, data curation, software, formal analysis. Y. Cheng: Resources, data curation, software, formal analysis. Q. Wang: Resources, data curation, software, formal analysis. Y. Pan: Resources, data curation, software, formal analysis. J. Zhou: Resources, data curation, software, formal analysis. J. Shi: Resources, data curation, software, formal analysis. X. Xu: Resources, data curation, software, formal analysis. L. Lin: Resources, data curation, software, formal analysis. W. Zhang: Resources, data curation, software, formal analysis. Y. Zhang: Resources, data curation, software, formal analysis. Y. Liu: Resources, data curation, software, formal analysis. Y. Fang: Resources, data curation, software, formal analysis. J. Feng: Resources, data curation, software, formal analysis. Z. Wang: Resources, data curation, software, formal analysis. S. Hu: Resources, data curation, software, formal analysis. J. Fang: Resources, data curation, software, formal analysis. Y. Shu: Resources, data curation, software, formal analysis. J. Cui: Resources, data curation, software, formal analysis. Y. Hu: Resources, data curation, software, formal analysis. W. Yao: Resources, data curation, software, formal analysis. X. Li: Resources, data curation, software, formal analysis. X. Lin: Resources, data curation, software, formal analysis. R. Wang: Resources, data curation, software, formal analysis. Y. Wang: Resources, data curation, software, formal analysis. W. Shi: Resources, data curation, software, formal analysis. G. Feng: Resources, data curation, software, formal analysis. J. Ni: Resources, data curation, software, formal analysis. B. Mao: Resources, data curation, software, formal analysis. D. Ren: Resources, data curation, software, formal analysis. H. Sun: Resources, data curation, software, formal analysis. H. Zhang: Resources, data curation, software, formal analysis. L. Chen: Conceptualization, resources, data curation, software, formal analysis, validation, visualization, methodology, project administration. C. Zhou: Conceptualization, resources, data curation, software, formal analysis, funding acquisition, validation, project administration, writing–review and editing. S. Ren: Conceptualization, resources, data curation, software, formal analysis, supervision, funding acquisition, validation, investigation, visualization, writing–original draft, project administration, writing–review and editing.

We are grateful to all patients and their families and all staff at the study centers. This study was also supported in part by grants from the National Natural Science Foundation of China (No. 82102859, 12126605, T2341007, 12131020, 31930022, 12026608), JST Moonshot R&D Grant (No. JPMJMS2021), Shanghai Public Health Committee Foundation (2020CXJQ02), Shanghai Rising-Star Program (23QA1408000), Oncology development incentive program of Shanghai Pulmonary Hospital, Cultivation project for National Natural Science Foundation of China Youth Fund of Shanghai Pulmonary Hospital (fkzr2306), Innovation Group of Shanghai Pulmonary Hospital (FKCX1903), Shanghai Hospital Development Center Three-year Action Plan to Promote Clinical Skills and Clinical Innovation in Municipal Hospitals (SHDC2020CR1036B), and the Project of Shanghai Municipal Health Commission: Establishment, Promotion and Application of Multidisciplinary Collaborative Diagnosis and Treatment System for Pulmonary Non-infectious Diseases.

The publication costs of this article were defrayed in part by the payment of publication fees. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/).

1.
Grant
MJ
,
Herbst
RS
,
Goldberg
SB
.
Selecting the optimal immunotherapy regimen in driver-negative metastatic NSCLC
.
Nat Rev Clin Oncol
2021
;
18
:
625
44
.
2.
Liu
L
,
Bai
H
,
Wang
C
,
Seery
S
,
Wang
Z
,
Duan
J
, et al
.
Efficacy and safety of first-line immunotherapy combinations for advanced NSCLC: a systematic review and network meta-analysis
.
J Thorac Oncol
2021
;
16
:
1099
117
.
3.
Havel
JJ
,
Chowell
D
,
Chan
TA
.
The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy
.
Nat Rev Cancer
2019
;
19
:
133
50
.
4.
Sharma
P
,
Siddiqui
BA
,
Anandhan
S
,
Yadav
SS
,
Subudhi
SK
,
Gao
J
, et al
.
The next decade of immune checkpoint therapy
.
Cancer Discov
2021
;
11
:
838
57
.
5.
Inoue
H
,
Tani
K
.
Multimodal immunogenic cancer cell death as a consequence of anticancer cytotoxic treatments
.
Cell Death Differ
2014
;
21
:
39
49
.
6.
Wang
Z
,
Till
B
,
Gao
Q
.
Chemotherapeutic agent-mediated elimination of myeloid-derived suppressor cells
.
Oncoimmunology
2017
;
6
:
e1331807
.
7.
Lesterhuis
WJ
,
Punt
CJ
,
Hato
SV
,
Eleveld-Trancikova
D
,
Jansen
BJ
,
Nierkens
S
, et al
.
Platinum-based drugs disrupt STAT6-mediated suppression of immune responses against cancer in humans and mice
.
J Clin Invest
2011
;
121
:
3100
8
.
8.
Leone
P
,
Shin
EC
,
Perosa
F
,
Vacca
A
,
Dammacco
F
,
Racanelli
V
.
MHC class I antigen processing and presenting machinery: organization, function, and defects in tumor cells
.
J Natl Cancer Inst
2013
;
105
:
1172
87
.
9.
Lu
Z
,
Chen
H
,
Jiao
X
,
Wang
Y
,
Wu
L
,
Sun
H
, et al
.
Germline HLA-B evolutionary divergence influences the efficacy of immune checkpoint blockade therapy in gastrointestinal cancer
.
Genome Med
2021
;
13
:
175
.
10.
Chowell
D
,
Krishna
C
,
Pierini
F
,
Makarov
V
,
Rizvi
NA
,
Kuo
F
, et al
.
Evolutionary divergence of HLA class I genotype impacts efficacy of cancer immunotherapy
.
Nat Med
2019
;
25
:
1715
20
.
11.
Chhibber
A
,
Huang
L
,
Zhang
H
,
Xu
J
,
Cristescu
R
,
Liu
X
, et al
.
Germline HLA landscape does not predict efficacy of pembrolizumab monotherapy across solid tumor types
.
Immunity
2022
;
55
:
56
64
.
12.
Litchfield
K
,
Reading
JL
,
Puttick
C
,
Thakkar
K
,
Abbosh
C
,
Bentham
R
, et al
.
Meta-analysis of tumor- and T cell-intrinsic mechanisms of sensitization to checkpoint inhibition
.
Cell
2021
;
184
:
596
614
.
13.
Zhou
C
,
Chen
G
,
Huang
Y
,
Zhou
J
,
Lin
L
,
Feng
J
, et al
.
Camrelizumab plus carboplatin and pemetrexed versus chemotherapy alone in chemotherapy-naive patients with advanced non-squamous non-small-cell lung cancer (CameL): a randomised, open-label, multicentre, phase 3 trial
.
Lancet Respir Med
2021
;
9
:
305
14
.
14.
Ren
S
,
Chen
J
,
Xu
X
,
Jiang
T
,
Cheng
Y
,
Chen
G
, et al
.
Camrelizumab plus carboplatin and paclitaxel as first-line treatment for advanced squamous NSCLC (CameL-Sq): a phase 3 trial
.
J Thorac Oncol
2022
;
17
:
544
57
.
15.
Szolek
A
,
Schubert
B
,
Mohr
C
,
Sturm
M
,
Feldhahn
M
,
Kohlbacher
O
.
OptiType: precision HLA typing from next-generation sequencing data
.
Bioinformatics
2014
;
30
:
3310
6
.
16.
Pierini
F
,
Lenz
TL
.
Divergent allele advantage at human MHC genes: signatures of past and ongoing selection
.
Mol Biol Evol
2018
;
35
:
2145
58
.
17.
Grantham
R
.
Amino acid difference formula to help explain protein evolution
.
Science
1974
;
185
:
862
4
.
18.
Yarmarkovich
M
,
Farrel
A
,
Sison
A
, 3rd, di
Marco
M
,
Raman
P
,
Parris
JL
, et al
.
Immunogenicity and immune silence in human cancer
.
Front Immunol
2020
;
11
:
69
.
19.
McGranahan
N
,
Rosenthal
R
,
Hiley
CT
,
Rowan
AJ
,
Watkins
TBK
,
Wilson
GA
, et al
.
Allele-specific HLA loss and immune escape in lung cancer evolution
.
Cell
2017
;
171
:
1259
71
.
20.
Chen
S
,
Zhou
Y
,
Chen
Y
,
Gu
J
.
fastp: an ultra-fast all-in-one FASTQ preprocessor
.
Bioinformatics
2018
;
34
:
i884
90
.
21.
Bolger
AM
,
Lohse
M
,
Usadel
B
.
Trimmomatic: a flexible trimmer for Illumina sequence data
.
Bioinformatics
2014
;
30
:
2114
20
.
22.
Li
H
,
Handsaker
B
,
Wysoker
A
,
Fennell
T
,
Ruan
J
,
Homer
N
, et al
.
The sequence alignment/map format and SAMtools
.
Bioinformatics
2009
;
25
:
2078
9
.
23.
McKenna
A
,
Hanna
M
,
Banks
E
,
Sivachenko
A
,
Cibulskis
K
,
Kernytsky
A
, et al
.
The genome analysis toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data
.
Genome Res
2010
;
20
:
1297
303
.
24.
Cibulskis
K
,
Lawrence
MS
,
Carter
SL
,
Sivachenko
A
,
Jaffe
D
,
Sougnez
C
, et al
.
Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples
.
Nat Biotechnol
2013
;
31
:
213
9
.
25.
Wang
K
,
Li
M
,
Hakonarson
H
.
ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data
.
Nucleic Acids Res
2010
;
38
:
e164
.
26.
Wu
F
,
Jiang
T
,
Chen
G
,
Huang
Y
,
Zhou
J
,
Lin
L
, et al
.
Multiplexed imaging of tumor immune microenvironmental markers in locally advanced or metastatic non-small-cell lung cancer characterizes the features of response to PD-1 blockade plus chemotherapy
.
Cancer Commun (Lond)
2022
;
42
:
1331
46
.
27.
Chalmers
ZR
,
Connelly
CF
,
Fabrizio
D
,
Gay
L
,
Ali
SM
,
Ennis
R
, et al
.
Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden
.
Genome Med
2017
;
9
:
34
.
28.
Kawaguchi
S
,
Higasa
K
,
Shimizu
M
,
Yamada
R
,
Matsuda
F
.
HLA-HD: An accurate HLA typing algorithm for next-generation sequencing data
.
Hum Mutat
2017
;
38
:
788
97
.
29.
Jurtz
V
,
Paul
S
,
Andreatta
M
,
Marcatili
P
,
Peters
B
,
Nielsen
M
.
NetMHCpan-4.0: improved peptide-MHC class I interaction predictions integrating eluted ligand and peptide binding affinity data
.
J Immunol
2017
;
199
:
3360
8
.
30.
Liu
C
,
Yang
X
,
Duffy
B
,
Mohanakumar
T
,
Mitra
RD
,
Zody
MC
, et al
.
ATHLATES: accurate typing of human leukocyte antigen through exome sequencing
.
Nucleic Acids Res
2013
;
41
:
e142
.
31.
Wu
F
,
Fan
J
,
He
Y
,
Xiong
A
,
Yu
J
,
Li
Y
, et al
.
Single-cell profiling of tumor heterogeneity and the microenvironment in advanced non-small cell lung cancer
.
Nat Commun
2021
;
12
:
2540
.
32.
Chen
J
,
Tan
Y
,
Sun
F
,
Hou
L
,
Zhang
C
,
Ge
T
, et al
.
Single-cell transcriptome and antigen-immunoglobin analysis reveals the diversity of B cells in non-small cell lung cancer
.
Genome Biol
2020
;
21
:
152
.
33.
Hao
Y
,
Hao
S
,
Andersen-Nissen
E
,
Mauck
WM
, 3rd,
Zheng
S
,
Butler
A
, et al
.
Integrated analysis of multimodal single-cell data
.
Cell
2021
;
184
:
3573
87
.
34.
Liu
B
,
Hu
X
,
Feng
K
,
Gao
R
,
Xue
Z
,
Zhang
S
, et al
.
Temporal single-cell tracing reveals clonal revival and expansion of precursor exhausted T cells during anti-PD-1 therapy in lung cancer
.
Nat Cancer
2022
;
3
:
108
21
.
35.
Wu
T
,
Hu
E
,
Xu
S
,
Chen
M
,
Guo
P
,
Dai
Z
, et al
.
clusterProfiler 4.0: A universal enrichment tool for interpreting omics data
.
Innovation (Camb)
2021
;
2
:
100141
.
36.
Yu
G
,
Wang
LG
,
Han
Y
,
He
QY
.
clusterProfiler: an R package for comparing biological themes among gene clusters
.
OMICS
2012
;
16
:
284
7
.
37.
Anderson
SK
.
Molecular evolution of elements controlling HLA-C expression: adaptation to a role as a killer-cell immunoglobulin-like receptor ligand regulating natural killer cell function
.
HLA
2018
;
92
:
271
8
.
38.
Lee
CH
,
DiNatale
RG
,
Chowell
D
,
Krishna
C
,
Makarov
V
,
Valero
C
, et al
.
High response rate and durability driven by HLA genetic diversity in patients with kidney cancer treated with lenvatinib and pembrolizumab
.
Mol Cancer Res
2021
;
19
:
1510
21
.
39.
Montesion
M
,
Murugesan
K
,
Jin
DX
,
Sharaf
R
,
Sanchez
N
,
Guria
A
, et al
.
Somatic HLA class I loss is a widespread mechanism of immune evasion which refines the use of tumor mutational burden as a biomarker of checkpoint inhibitor response
.
Cancer Discov
2021
;
11
:
282
92
.
40.
Goodman
AM
,
Castro
A
,
Pyke
RM
,
Okamura
R
,
Kato
S
,
Riviere
P
, et al
.
MHC-I genotype and tumor mutational burden predict response to immunotherapy
.
Genome Med
2020
;
12
:
45
.
41.
Oh
DY
,
Fong
L
.
Cytotoxic CD4(+) T cells in cancer: Expanding the immune effector toolbox
.
Immunity
2021
;
54
:
2701
11
.
42.
Laumont
CM
,
Banville
AC
,
Gilardi
M
,
Hollern
DP
,
Nelson
BH
.
Tumour-infiltrating B cells: immunological mechanisms, clinical impact and therapeutic opportunities
.
Nat Rev Cancer
2022
;
22
:
414
30
.
43.
Kvedaraite
E
,
Ginhoux
F
.
Human dendritic cells in cancer
.
Sci Immunol
2022
;
7
:
eabm9409
.
44.
Pittet
MJ
,
Michielin
O
,
Migliorini
D
.
Clinical relevance of tumour-associated macrophages
.
Nat Rev Clin Oncol
2022
;
19
:
402
21
.
45.
De Sanctis
F
,
Adamo
A
,
Cane
S
,
Ugel
S
.
Targeting tumour-reprogrammed myeloid cells: the new battleground in cancer immunotherapy
.
Semin Immunopathol
2023
;
45
:
163
86
.

Supplementary data