AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (1 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Original Article | Open Access

DNA damage response-related immune activation signature predicts the response to immune checkpoint inhibitors: from gastrointestinal cancer analysis to pan-cancer validation

Junya Yan1,2,*Shibo Wang1,*Jing Zhang3,*Qiangqiang Yuan1Xianchun Gao1Nannan Zhang1Yan Pan1Haohao Zhang1Kun Liu4Jun Yu1Linbin Lu1Hui Liu5Xiaoliang Gao1Sheng Zhao1Wenyao Zhang1Abudurousuli Reyila1Yu Qi1Qiujin Zhang6Shundong Cang2Yuanyuan Lu1Yanglin Pan1Yan Kong7 ( )Yongzhan Nie1 ( )
State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Air Force Military Medical University, Xi’an 710032, China
Department of Oncology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou 450003, China
Faculty of Life Science, Northwest University, Xi’an 710069, China
Unit 73211 of the People’s Liberation Army, Nanjing 211800, China
Key Laboratory of Resource Biology and Biotechnology in Western China (Ministry of Education), School of Medicine, Northwest University, Xi’an 710069, China
Shaanxi University of Chinese Medicine, Second Clinical Medicine Faculty, Xi’an 712046, China
Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing 100142, China

*These authors contributed equally to this work.

Show Author Information

Abstract

Objective

DNA damage response (DDR) deficiency has emerged as a prominent determinant of tumor immunogenicity. This study aimed to construct a DDR-related immune activation (DRIA) signature and evaluate the predictive accuracy of the DRIA signature for response to immune checkpoint inhibitor (ICI) therapy in gastrointestinal (GI) cancer.

Methods

A DRIA signature was established based on two previously reported DNA damage immune response assays. Clinical and gene expression data from two published GI cancer cohorts were used to assess and validate the association between the DRIA score and response to ICI therapy. The predictive accuracy of the DRIA score was validated based on one ICI-treated melanoma and three pan-cancer published cohorts.

Results

The DRIA signature includes three genes (CXCL10, IDO1, and IFI44L). In the discovery cancer cohort, DRIA-high patients with gastric cancer achieved a higher response rate to ICI therapy than DRIA-low patients (81.8% vs. 8.8%; P < 0.001), and the predictive accuracy of the DRIA score [area under the receiver operating characteristic curve (AUC) = 0.845] was superior to the predictive accuracy of PD-L1 expression, tumor mutational burden, microsatellite instability, and Epstein–Barr virus status. The validation cohort demonstrated that the DRIA score identified responders with microsatellite-stable colorectal and pancreatic adenocarcinoma who received dual PD-1 and CTLA-4 blockade with radiation therapy. Furthermore, the predictive performance of the DRIA score was shown to be robust through an extended validation in melanoma, urothelial cancer, and pan-cancer.

Conclusions

The DRIA signature has superior and robust predictive accuracy for the efficacy of ICI therapy in GI cancer and pan-cancer, indicating that the DRIA signature may serve as a powerful biomarker for guiding ICI therapy decisions.

Electronic Supplementary Material

Download File(s)
cbm-21-3-252_ESM.pdf (832 KB)

References

1

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021; 71: 209-49.

2

Javle M, Hsueh CT. Recent advances in gastrointestinal oncology – updates and insights from the 2009 annual meeting of the American Society of Clinical Oncology. J Hematol Oncol. 2010; 3: 11.

3

Wang Y, Wang M, Wu HX, Xu RH. Advancing to the era of cancer immunotherapy. Cancer Commun (Lond). 2021; 41: 803-29.

4

Sun JM, Shen L, Shah MA, Enzinger P, Adenis A, DoiT, et al. Pembrolizumab plus chemotherapy versus chemotherapy alone for first-line treatment of advanced oesophageal cancer (KEYNOTE-590): a randomised, placebo-controlled, phase 3 study. Lancet. 2021; 398: 759-71.

5

Diaz LA Jr, Shiu KK, Kim TW, Jensen BV, Jensen LH, Punt C, et al. Pembrolizumab versus chemotherapy for microsatellite instabilityhigh or mismatch repair-deficient metastatic colorectal cancer (KEYNOTE-177): final analysis of a randomised, open-label, phase 3 study. Lancet Oncol. 2022; 23: 659-70.

6

Wang ZX, Pan YQ, Li X, Tsubata T, Xu RH. Immunotherapy in gastrointestinal cancers: advances, challenges, and countermeasures. Sci Bull (Beijing). 2023; 68: 763-6.

7

Shitara K, Özgüroğlu M, Bang YJ, Di Bartolomeo M, Mandalà M, Ryu MH, et al. Pembrolizumab versus paclitaxel for previously treated, advanced gastric or gastro-oesophageal junction cancer (KEYNOTE-061): a randomised, open-label, controlled, phase 3 trial. Lancet. 2018; 392: 123-33.

8

Wang F, Wei XL, Wang FH, Xu N, Shen L, Dai GH, et al. Safety, efficacy and tumor mutational burden as a biomarker of overall survival benefit in chemo-refractory gastric cancer treated with toripalimab, a PD-1 antibody in phase Ib/Ⅱ clinical trial NCT02915432. Ann Oncol. 2019; 30: 1479-86.

9

Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 2015; 372: 2509-20.

10

Kim ST, Cristescu R, Bass AJ, Kim KM, Odegaard JI, Kim K, et al. Comprehensive molecular characterization of clinical responses to PD-1 inhibition in metastatic gastric cancer. Nat Med. 2018; 24: 1449-58.

11

Venderbosch S, Nagtegaal ID, Maughan TS, Smith CG, Cheadle JP, Fisher D, et al. Mismatch repair status and BRAF mutation status in metastatic colorectal cancer patients: a pooled analysis of the CAIRO, CAIRO2, COIN, and FOCUS studies. Clin Cancer Res. 2014; 20: 5322-30.

12

Kojima T, Shah MA, Muro K, Francois E, Adenis A, Hsu CH, et al. Randomized Phase Ⅲ KEYNOTE-181 Study of pembrolizumab versus chemotherapy in advanced esophageal cancer. J Clin Oncol. 2020; 38: 4138-48.

13

Ott PA, Bang YJ, Piha-Paul SA, Razak A, Bennouna J, Soria JC, et al. T-cell-inflamed gene-expression profile, programmed death ligand 1 expression, and tumor mutational burden predict efficacy in patients treated with pembrolizumab across 20 cancers: KEYNOTE-028. J Clin Oncol. 2019; 37: 318-27.

14

Samstein RM, Lee CH, Shoushtari AN, Hellmann MD, Shen R, Janjigian YY, et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet. 2019; 51: 202-6.

15

Lawrence MS, Stojanov P, Polak P, Kryukov GV, Cibulskis K, Sivachenko A, et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature. 2013; 499: 214-8.

16

Chabanon RM, Rouanne M, Lord CJ, Soria JC, Pasero P, Postel-Vinay S. Targeting the DNA damage response in immuno-oncology: developments and opportunities. Nat Rev Cancer. 2021; 21: 701-17.

17

Pujade-Lauraine E, Ledermann JA, Selle F, Gebski V, Penson RT, Oza AM, et al. Olaparib tablets as maintenance therapy in patients with platinum-sensitive, relapsed ovarian cancer and a BRCA1/2 mutation (SOLO2/ENGOT-Ov21): a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol. 2017; 18: 1274-84.

18

Hugo W, Zaretsky JM, Sun L, Song C, Moreno BH, Hu-Lieskovan S, et al. Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell. 2016; 165: 35-44.

19

Teo MY, Seier K, Ostrovnaya I, Regazzi AM, Kania BE, Moran MM, et al. Alterations in DNA damage response and repair genes as potential marker of clinical benefit from PD-1/PD-L1 blockade in advanced urothelial cancers. J Clin Oncol. 2018; 36: 1685-94.

20

Wang Y, Jiao X, Li S, Chen H, Wei X, Liu C, et al. Alterations in DNA damage response and repair genes as potential biomarkers for immune checkpoint blockade in gastrointestinal cancer. Cancer Biol Med. 2021; 19: 1139-49.

21

Huang R, Zhou PK. DNA damage repair: historical perspectives, mechanistic pathways and clinical translation for targeted cancer therapy. Signal Transduct Target Ther. 2021; 6: 254.

22

Mulligan JM, Hill LA, Deharo S, Irwin G, Boyle D, Keating KE, et al. Identification and validation of an anthracycline/cyclophosphamide-based chemotherapy response assay in breast cancer. J Natl Cancer Inst. 2014; 106: djt335.

23

Parkes EE, Walker SM, Taggart LE, McCabe N, Knight LA, Wilkinson R, et al. Activation of STING-dependent innate immune signaling by S-phase-specific DNA damage in breast cancer. J Natl Cancer Inst. 2017; 109: djw199.

24

Sharma P, Barlow WE, Godwin AK, Parkes EE, Knight LA, Walker SM, et al. Validation of the DNA damage immune response signature in patients with triple-negative breast cancer from the SWOG 9313c Trial. J Clin Oncol. 2019; 37: 3484-92.

25

Parkes EE, Savage KI, Lioe T, Boyd C, Halliday S, Walker SM, et al. Activation of a cGAS-STING-mediated immune response predicts response to neoadjuvant chemotherapy in early breast cancer. Br J Cancer. 2022; 126: 247-58.

26

Turkington RC, Knight LA, Blayney JK, Secrier M, Douglas R, Parkes EE, et al. Immune activation by DNA damage predicts response to chemotherapy and survival in oesophageal adenocarcinoma. Gut. 2019; 68: 1918-27.

27

Malla SB, Fisher DJ, Domingo E, Blake A, Hassanieh S, Redmond KL, et al. In-depth clinical and biological exploration of DNA damage immune response as a biomarker for oxaliplatin use in colorectal cancer. Clin Cancer Res. 2021; 27: 288-300.

28

Cui C, Xu C, Yang W, Chi Z, Sheng X, Si L, et al. Ratio of the interferon-γ signature to the immunosuppression signature predicts anti-PD-1 therapy response in melanoma. NPJ Genom Med. 2021; 6: 7.

29

Parikh AR, Szabolcs A, Allen JN, Clark JW, Wo JY, Raabe M, et al. Radiation therapy enhances immunotherapy response in microsatellite stable colorectal and pancreatic adenocarcinoma in a phase Ⅱ trial. Nat Cancer. 2021; 2: 1124-35.

30

Gide TN, Quek C, Menzies AM, Tasker AT, Shang P, Holst J, et al. Distinct immune cell populations define response to anti-PD-1 monotherapy and anti-PD-1/anti-CTLA-4 combined therapy. Cancer Cell. 2019; 35: 238-55.e6.

31

Mariathasan S, Turley SJ, Nickles D, Castiglioni A, Yuen K, Wang Y, et al. TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature. 2018; 554: 544-8.

32

Pender A, Titmuss E, Pleasance ED, Fan KY, Pearson H, Brown SD, et al. Genome and transcriptome biomarkers of response to immune checkpoint inhibitors in advanced solid tumors. Clin Cancer Res. 2021; 27: 202-12.

33

Teo MY, Bambury RM, Zabor EC, Jordan E, Al-Ahmadie H, Boyd ME, et al. DNA damage response and repair gene alterations are associated with improved survival in patients with platinum-treated advanced urothelial carcinoma. Clin Cancer Res. 2017; 23: 3610-8.

34

You Z, Lv M, He X, Pan Y, Ge J, Hu X, et al. Homologous recombination repair gene mutations as a predictive biomarker for immunotherapy in patients with advanced melanoma. Front Immunol. 2022; 13: 871756.

35

Wang Z, Zhao J, Wang G, Zhang F, Zhang Z, Zhang F, et al. Comutations in DNA damage response pathways serve as potential biomarkers for immune checkpoint blockade. Cancer Res. 2018; 78: 6486-96.

36

Linger RJ, Kruk PA. BRCA1 16 years later: risk-associated BRCA1 mutations and their functional implications. FEBS J. 2010; 277: 3086-96.

37

Esteller M, Silva JM, Dominguez G, Bonilla F, Matias-Guiu X, Lerma E, et al. Promoter hypermethylation and BRCA1 inactivation in sporadic breast and ovarian tumors. J Natl Cancer Inst. 2000; 92: 564-9.

38

Turner N, Tutt A, Ashworth A. Hallmarks of ‘BRCAness’ in sporadic cancers. Nat Rev Cancer. 2004; 4: 814-9.

39

Vidotto T, Nersesian S, Graham C, Siemens DR, Koti M. DNA damage repair gene mutations and their association with tumor immune regulatory gene expression in muscle invasive bladder cancer subtypes. J Immunother Cancer. 2019; 7: 148.

40

Song Y, Huang J, Liang D, Hu Y, Mao B, Li Q, et al. DNA damage repair gene mutations are indicative of a favorable prognosis in colorectal cancer treated with immune checkpoint inhibitors. Front Oncol. 2020; 10: 549777.

41

Keenan BP, VAN Loon K, Khilnani AD, Fidelman N, Behr SC, Atreya CE, et al. Molecular and radiological features of microsatellite stable colorectal cancer cases with dramatic responses to immunotherapy. Anticancer Res. 2021; 41: 2985-92.

42

Hu J, Chen Z, Bao L, Zhou L, Hou Y, Liu L, et al. Single-cell transcriptome analysis reveals intratumoral heterogeneity in ccRCC, which results in different clinical outcomes. Mol Ther. 2020; 28: 1658-72.

43

Nicoś M, Krawczyk P, Crosetto N, Milanowski J. The role of intratumor heterogeneity in the response of metastatic non-small cell lung cancer to immune checkpoint inhibitors. Front Oncol. 2020; 10: 569202.

Cancer Biology & Medicine
Pages 252-266
Cite this article:
Yan J, Wang S, Zhang J, et al. DNA damage response-related immune activation signature predicts the response to immune checkpoint inhibitors: from gastrointestinal cancer analysis to pan-cancer validation. Cancer Biology & Medicine, 2024, 21(3): 252-266. https://doi.org/10.20892/j.issn.2095-3941.2023.0303

185

Views

1

Downloads

0

Crossref

0

Web of Science

0

Scopus

Altmetrics

Received: 15 August 2023
Accepted: 13 November 2023
Published: 29 December 2023
©2024 The Authors.

Creative Commons Attribution-NonCommercial 4.0 International License

Return