Abstract
Introduction
Methods
Results
Conclusions
Keywords
Introduction
Methods
MDACC Cohort
CM012 and Chowell Cohorts
HLA Typing and Supertypes
Statistical Analysis
Results
Outcome Analysis: MDACC Cohort
Characteristic | MDACC Cohort (n = 200) | CM012 Cohort (n = 75) | Chowell Cohort (n = 371) |
---|---|---|---|
Age, y, n (%) | |||
≤64 | 80 (40) | 36 (48) | 261 (70) |
>64 | 120 (60) | 39 (52) | 110 (30) |
Sex, n (%) | |||
Female | 96 (48) | 38 (51) | 51 (14) |
Male | 104 (52) | 37 (49) | 49 (13) |
NA | 0 (0) | 0 (0) | 271 (73) |
Smoking status, n (%) | |||
Never | 45 (22) | 15 (20) | NA |
Ever | 155 (78) | 60 (80) | NA |
Histologic type, n (%) | |||
Nonsquamous | 160 (80) | 59 (79) | NA |
Squamous | 40 (20) | 16 (21) | NA |
PD-L1 expression, n (%) | |||
Negative | 51 (26) | 25 (33) | 0 (0) |
Positive | 82 (41) | 45 (60) | 0 (0) |
NA | 67 (34) | 5 (7) | 371 (100) |
Targetable driver mutation, n (%) | |||
No | 164 (82) | 67 (89) | NA |
Yes | 36 (18) | 8 (11) | NA |
STK11 mutation, n (%) | |||
No | 177 (89) | 67 (89) | NA |
Yes | 20 (10) | 7 (9) | NA |
NA | 3 (2) | 1 (1) | NA |
Prior radiation therapy, n (%) | |||
none/≥6 mo | 141 (70) | NA | NA |
<6 mo | 59 (30) | NA | NA |
Prior lines of therapy, n (%) | |||
0–1 | 150 (75) | NA | NA |
≥2 | 50 (25) | NA | NA |
Time from prior systemic therapy, n (%) | |||
none/≥6 mo | 81 (40) | NA | NA |
<6 mo | 119 (60) | NA | NA |
Concurrent chemotherapy, n (%) | |||
No | 183 (92) | 75 (100) | 371 (100) |
Yes | 17 (8) | 0 (0) | 0 (0) |
Tumor mutational burden, n (%) | |||
≥ median | 0 (0) | 38 (51) | 161 (43) |
< median | 0 (0) | 37 (49) | 144 (39) |
NA | 200 (100) | 0 (0) | 66 (18) |
Overall HLA class I zygosity, n (%) | |||
Heterozygous | 157 (78) | 55 (73) | 291 (78) |
Homozygous | 43 (22) | 20 (27) | 80 (22) |
HLA-A zygosity, n (%) | |||
Heterozygous | 174 (87) | 62 (83) | 328 (88) |
Homozygous | 26 (13) | 13 (17) | 43 (12) |
HLA-B zygosity, n (%) | |||
Heterozygous | 183 (92) | 65 (87) | 350 (94) |
Homozygous | 17 (8) | 10 (13) | 21 (6) |
HLA-C zygosity, n (%) | |||
Heterozygous | 180 (90) | 66 (88) | 331 (89) |
Homozygous | 20 (10) | 9 (12) | 40 (11) |

Characteristic | HR (95% CI) | p Value |
---|---|---|
Progression-free survival | ||
Univariate analysis | ||
Zygosity (homozygous vs. heterozygous) | 0.87 (0.59–1.28) | 0.480 |
HLA-A homozygous (yes vs. no) | 0.79 (0.49–1.26) | 0.315 |
HLA-B homozygous (yes vs. no) | 1.21 (0.70–2.09) | 0.504 |
HLA-C homozygous (yes vs. no) | 0.94 (0.55–1.61) | 0.832 |
Age (>64 vs. ≤64) | 0.89 (0.64–1.22) | 0.461 |
Sex (male vs. female) | 1.05 (0.76–1.43) | 0.781 |
Smoking status (ever vs. never) | 0.65 (0.46–0.93) | 0.019 |
Histological type (nonsquamous vs. squamous) | 0.71 (0.49–1.05) | 0.083 |
PD-L1 expression (positive vs. negative) | 0.82 (0.55–1.22) | 0.326 |
Targetable driver mutation (yes vs. no) | 2.08 (1.42–3.05) | <0.001 |
STK11 mutation (yes vs. no) | 1.12 (0.65–1.91) | 0.689 |
Prior radiation therapy (none/≥6 mo vs. <6 mo) | 0.74 (0.53–1.03) | 0.076 |
Prior lines of therapy (≥2 vs. 0 or 1) | 0.97 (0.68–1.39) | 0.884 |
Time from prior systemic therapy (none/≥6 mo vs. <6 mo) | 0.96 (0.70–1.33) | 0.826 |
Concurrent agents (yes vs. no) | 0.61 (0.33–1.12) | 0.112 |
Multivariate analysis | ||
Histologic type (nonsquamous vs. squamous) | 0.59 (0.40–0.88) | 0.010 |
Prior radiation therapy (none/≥6 mo vs. <6 mo) | 0.70 (0.50–0.99) | 0.042 |
Targetable driver mutation (yes vs. no) | 2.43 (1.63–3.63) | <0.001 |
Overall survival | ||
Univariate analysis | ||
Zygosity (homozygous vs. heterozygous) | 0.67 (0.36–1.26) | 0.217 |
HLA-A homozygous (yes vs. no) | 0.37 (0.15–0.92) | 0.033 |
HLA-B homozygous (yes vs. no) | 1.20 (0.48–2.98) | 0.700 |
HLA-C homozygous (yes vs. no) | 1.08 (0.49–2.38) | 0.845 |
Age (>64 vs. ≤64) | 1.09 (0.66–1.79) | 0.744 |
Sex (male vs. female) | 1.00 (0.61–1.63) | 0.996 |
Smoking status (ever vs. never) | 1.10 (0.62–1.97) | 0.735 |
Histologic type (nonsquamous vs. squamous) | 0.73 (0.41–1.30) | 0.280 |
PD-L1 expression (positive vs. negative) | 0.35 (0.19–0.66) | 0.001 |
Targetable driver mutation (yes vs. no) | 1.19 (0.63–2.23) | 0.591 |
STK11 mutation (yes vs. no) | 1.78 (0.84–3.78) | 0.130 |
Prior radiation therapy (none/≥6 mo vs. <6 mo) | 0.73 (0.44–1.22) | 0.235 |
Prior lines of therapy (≥2 vs. 0 or 1) | 1.53 (0.92–2.54) | 0.100 |
Time from prior systemic therapy (none/≥6 mo vs. <6 mo) | 0.63 (0.37–1.07) | 0.085 |
Concurrent agents (yes vs. no) | 0.89 (0.36–2.23) | 0.810 |
Multivariate analysis | ||
HLA-A homozygous (yes vs. no) | 0.44 (0.10–1.82) | 0.256 |
Time from prior systemic therapy (none/≥6 mo vs. <6 mo) | 0.69 (0.35–1.34) | 0.271 |
PD-L1 expression (positive vs. negative) | 0.39 (0.20–0.75) | 0.005 |
HLA Class I Supertypes and Alleles: MDACC Cohort
Characteristic | HR (95% CI) | p Value |
---|---|---|
HLA supertype multivariate analysis | ||
Progression-free survival | ||
Histologic type (nonsquamous vs. squamous) | 0.59 (0.40–0.88) | 0.010 |
Targetable driver mutation (yes vs. no) | 2.45 (1.64–3.66) | <0.001 |
Prior radiation therapy (none/≥6 mo vs. <6 mo) | 0.71 (0.50–0.99) | 0.046 |
A24 (present vs. absent) | 1.38 (0.95–2.00) | 0.088 |
Overall survival | ||
PD-L1 expression (positive vs. negative) | 0.41 (0.21–0.78) | 0.007 |
A24 (present vs. absent) | 1.66 (0.78–3.53) | 0.191 |
HLA allele multivariate analysis | ||
Progression-free survival | ||
Histologic type (nonsquamous vs. squamous) | 0.54 (0.35–0.82) | 0.004 |
Targetable driver mutation (yes vs. no) | 2.70 (1.77–4.12) | <0.001 |
Prior radiation therapy (none/≥6 mo vs. <6 mo) | 0.68 (0.47–0.99) | 0.043 |
A23:01 (present vs. absent) | 1.88 (0.91–3.89) | 0.089 |
C03:04 (present vs. absent) | 2.30 (1.35–3.91) | 0.002 |
Overall survival | ||
PD-L1 expression (positive vs. negative) | 0.45 (0.23–0.88) | 0.021 |
A23:01 (present vs. absent) | 0.85 (0.11–6.46) | 0.876 |
C05:01 (present vs. absent) | 0.52 (0.18–1.52) | 0.229 |
Outcome Analysis: CM012 Cohort
Characteristic | HR (95% CI) | P value |
---|---|---|
Univariate analysis | ||
Zygosity (homozygous vs. heterozygous) | 0.86 (0.45–1.65) | 0.649 |
HLA-A homozygous (yes vs. no) | 0.83 (0.39–1.77) | 0.627 |
HLA-B homozygous (yes vs. no) | 0.76 (0.32–1.79) | 0.529 |
HLA-C homozygous (yes vs. no) | 0.73 (0.29–1.85) | 0.508 |
Age (>64 vs. ≤64) | 0.89 (0.51–1.55) | 0.674 |
Sex (male vs. female) | 1.03 (0.59–1.79) | 0.921 |
Smoking status (ever vs. never) | 0.70 (0.36–1.36) | 0.288 |
Histologic type (nonsquamous vs. squamous) | 0.85 (0.42–1.70) | 0.648 |
PD-L1 expression (positive vs. negative) | 0.86 (0.47–1.59) | 0.634 |
Targetable driver mutation (yes vs. no) | 3.43 (1.56–7.54) | 0.002 |
STK11 mutation (yes vs. no) | 3.05 (1.27–7.34) | 0.013 |
Tumor mutational burden (≥ median vs. < median) | 0.45 (0.25–0.79) | 0.006 |
Multivariate analysis | ||
Tumor mutational burden (≥ median vs. < median) | 0.45 (0.24–0.84) | 0.011 |
STK11 mutation (yes vs. no) | 4.31 (1.73–10.77) | 0.002 |
Targetable driver mutation (yes vs. no) | 2.62 (1.12–6.12) | 0.026 |
HLA supertype multivariate analysis | ||
Tumor mutational burden (≥ median vs. < median) | 0.43 (0.23–0.80) | 0.008 |
STK11 mutation (yes vs. no) | 3.59 (1.41–9.15) | 0.008 |
Targetable driver mutation (yes vs. no) | 2.44 (1.04–5.70) | 0.040 |
A02 (present vs. absent) | 0.61 (0.34–1.09) | 0.094 |
HLA Class I Supertypes and Alleles: CM012 Cohort
Outcomes Analysis: Chowell Cohort
Discussion
Acknowledgments
Supplementary Data
- Supplemental Table 1
- Supplemental Figure 1
HLA zygosity and outcomes for each HLA class I loci – MDACC cohort. A) Progression-free survival for HLA-A zygosity; B) Progression-free survival for HLA-B zygosity; C) Progression-free survival for HLA-C zygosity; D) Overall survival for HLA-A zygosity; E) Overall survival for HLA-B zygosity; F) Overall survival for HLA-C zygosity.
- Supplemental Figure 2
HLA zygosity and outcomes for each HLA class I loci – CM012 cohort. A) Progression-free survival for HLA-A zygosity; B) Progression-free survival for HLA-B zygosity; C) Progression-free survival for HLA-C zygosity.
References
- Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer.N Engl J Med. 2015; 373: 1627-1639
- Five-year follow-up of nivolumab in previously treated advanced non–small-cell lung cancer: results from the CA209-003 study.J Clin Oncol. 2018; 36: 1675-1684
- Atezolizumab for first-line treatment of metastatic nonsquamous NSCLC.N Engl J Med. 2018; 378: 2288-2301
- Pembrolizumab plus chemotherapy in metastatic non–small-cell lung cancer.N Engl J Med. 2018; 378: 2078-2092https://doi.org/10.1056/NEJMoa1801005
- Pembrolizumab plus chemotherapy for squamous non–small-cell lung cancer.N Engl J Med. 2018; 379: 2040-2051
- Pembrolizumab versus chemotherapy for PD-L1–positive non–small-cell lung cancer.N Engl J Med. 2016; 19: 1823-1833
- Mutational landscape determines sensitivity to PD-1 blockade in non–small cell lung cancer.Science. 2015; 348: 124-128
- Genetic basis for clinical response to CTLA-4 blockade in melanoma.N Engl J Med. 2014; : 2189-2199
- Genomic features of response to combination immunotherapy in patients with advanced non-small-cell lung cancer.Cancer Cell. 2018; 33: 843-852.e4
- Overall survival with combined nivolumab and ipilimumab in advanced melanoma.N Engl J Med. 2017; 377: 1345-1356
- Durable clinical benefit with nivolumab plus ipilimumab in DNA mismatch repair-deficient/microsatellite instability-high metastatic colorectal cancer.J Clin Oncol. 2018; 36: 773-779
- Mismatch-repair deficiency predicts response of solid tumors to PD-1 blockade.Science. 2017; 357: 409-413
- Nivolumab versus docetaxel in advanced squamous-cell non–small-cell lung cancer.N Engl J Med. 2015; 373: 123-135
- Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): a multicentre, open-label, phase 2 randomised controlled trial.Lancet. 2016; 387: 1837-1846
- Robust prediction of response to immune checkpoint blockade therapy in metastatic melanoma.Nat Med. 2018; 24: 1545-1549
- Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response.Nat Med. 2018; 24: 1550-1558
- Allele-specific HLA loss and immune escape in lung cancer evolution.Cell. 2017; 171: 1259-1271.e11https://doi.org/10.1016/j.cell.2017.10.001
- Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade.Science. 2016; 351: 1463-1469
- Patient HLA class I genotype influences response to immune checkpoint blockade therapy.Science. 2017; 359: 582-587
- Pembrolizumab for the treatment of non–small-cell lung cancer.N Engl J Med. 2015; 372: 2018-2028
- Development of a companion diagnostic for pembrolizumab in non-small cell lung cancer using immunohistochemistry for programmed death ligand-1.Arch Pathol Lab Med. 2016; 140: 1243-1249
- Nivolumab plus ipilimumab as first-line treatment for advanced non-small-cell lung cancer (CheckMate 012): results of an open-label, phase 1, multicohort study.Lancet Oncol. 2017; 18: 31-41
- Development of an automated PD-L1 immunohistochemistry (IHC) assay for non-small cell lung cancer.Appl Immunohistochem Mol Morphol. 2015; 23: 541-549
- HLA class I supertypes: a revised and updated classification.BMC Immunol. 2008; 9: 1-15
- Molecular determinants of response to anti–programmed cell death (PD)-1 and anti–programmed death-ligand (PD-L)-ligand 1 blockade in patients with non–small-cell lung cancer profiled with targeted next-generation sequencing.J Clin Oncol. 2018; 36: 633-641
- Blood-based tumor mutational burden as a predictor of clinical benefit in non-small-cell lung cancer patients treated with atezolizumab.Nat Med. 2018; 24: 1441-1448
- Genomic correlates of response to immune checkpoint blockade in microsatellite-stable solid tumors.Nat Genet. 2018; 50: 1271-1281
- TP53 , STK11 and EGFR mutations predict tumor immune profile and the response to anti-PD-1 in lung adenocarcinoma.Clin Cancer Res. 2018; 24: 5710-5723
- EGFR mutations and ALK rearrangements are associated with low response rates to PD-1 pathway blockade in non-small cell lung cancer: a retrospective analysis.Clin Cancer Res. 2016; 22: 4585-4593https://doi.org/10.1158/1078-0432.CCR-15-3101
- Phase 1/2 study of the safety and tolerability of nivolumab plus crizotinib for the first-line treatment of anaplastic lymphoma kinase translocation—positive advanced non–small cell lung cancer (CheckMate 370).J Thorac Oncol. 2018; 13: 682-688
- Mutations associated with acquired resistance to PD-1 blockade in melanoma.N Engl J Med. 2016; 375: 819-829
- Impaired HLA class I antigen processing and presentation as a mechanism of acquired resistance to immune checkpoint inhibitors in lung cancer.Cancer Discov. 2017; 7: 1420-1435
- Comprehensive analysis of cancer-associated somatic mutations in class i HLA genes.Nat Biotechnol. 2015; 33: 1152-1158
- Major histocompatibility complex class II and programmed death ligand 1 expression predict outcome after programmed death 1 blockade in classic Hodgkin lymphoma.J Clin Oncol. 2018; 36: 942-950
- MHC proteins confer differential sensitivity to CTLA-4 and PD-1 blockade in untreated metastatic melanoma.Sci Transl Med. 2018; 10: eaar3342
- Durvalumab as third-line or later treatment for advanced non-small-cell lung cancer (ATLANTIC): an open-label, single-arm, phase 2 study.Lancet Oncol. 2018; 19: 521-536
- Nivolumab plus ipilimumab in lung cancer with a high tumor mutational burden.N Engl J Med. 2018; 373: 2093-2104
- STK11/LKB1 mutations and PD-1 inhibitor resistance in KRAS-mutant lung adenocarcinoma.Cancer Discov. 2018; 8: 822-835
Article info
Publication history
Footnotes
Dr. Negrao and Dr. Lam equally contributed to this work.
Disclosure: Dr. Lam reports grants and personal fees from Takeda, personal fees from BMS, and grants from Guardant Health and Adaptimmune outside the submitted work. Dr. Swisher reports personal fees from Ethicon and Peter MacCallum Cancer Center outside the submitted work. Dr. Gibbons reports grants and personal fees from AstraZeneca and Janssen, personal fees from Sanofi and GSK, and grants from Takeda outside the submitted work. Dr. Wistuba reports grants and personal fees from Genentech/Roche, Bristol-Myers Squibb, Astra Zeneca/Medimmune, Pfizer, HTG Molecular, Asuragen, and Merck; grants from EMD Serono, Oncoplex, DepArray, Adaptive, Adaptimmune, Takeda, Amgen, Karus, Johnson & Johnson, Bayer, and 4D; and personal fees from GlaxoSmithKline and MSD outside the submitted work. Dr. Papadimitrakopoulou reports grants and personal fees from Nektar Therapeutics, AstraZeneca, Merck, F. Hoffman-La Roche, Bristol-Myers Squibb, Eli Lilly, and Novartis; personal fees from Arrys Therapeutics, LOXO Oncology, Araxes Pharma, Jannsen Research Foundation, Clovis Oncology, Takeda, Abbvie, TRM Oncology, Tesaro, Exelixis, and Gritstone, and grants from Janssen, Checkmate, and Incyte outside the submitted work. Dr. Glisson reports grants from Bristol Myers Squibb, Pfizer, and Medimmune outside the submitted work. Dr. Blumenschein reports personal fees from Abbvie, Adicet, Amgen, ARIAD, Clovis, and Genentech; grants and personal fees from Bayer, BMS, Celgene, Merck, Novartis, and Xcovery; and grants from Adaptimmune, Exelixis, Genentech, GlaxoSmithKline, Hoffman-La Roche, Immatics, Incyte, KITE, Macrogenetics, MedImmune, and Torque outside the submitted work. Dr. Heymach reports grants and personal fees from AstraZeneca, Spectrum, and GlaxoSmithKline; personal fees from Boehringer Ingelheim, Exelixis, Genentech, Guardant Health, Hengrui, Lilly, Novartis, EMD Serono, and Synta; and grants from Bayer outside the submitted work; in addition, Dr. Heymach has a patent held by Spectrum with royalties paid. Dr. Zhang reports personal fees from BMS, AstraZeneca, Geneplus, OrigMed, and Innovent outside the submitted work. The remaining authors declare no conflict of interest.
Identification
Copyright
User license
Elsevier user license |
Permitted
For non-commercial purposes:
- Read, print & download
- Text & data mine
- Translate the article
Not Permitted
- Reuse portions or extracts from the article in other works
- Redistribute or republish the final article
- Sell or re-use for commercial purposes
Elsevier's open access license policy