Genomic predictors of response to PD-1 inhibition in children with germline DNA replication repair deficiency
Language English Country United States Media print-electronic
Document type Journal Article, Observational Study, Research Support, Non-U.S. Gov't
Grant support
PJT-156006
CIHR - Canada
PubMed
34992263
PubMed Central
PMC8799468
DOI
10.1038/s41591-021-01581-6
PII: 10.1038/s41591-021-01581-6
Knihovny.cz E-resources
- MeSH
- Survival Analysis MeSH
- B7-H1 Antigen antagonists & inhibitors MeSH
- Child MeSH
- Adult MeSH
- Immune Checkpoint Inhibitors pharmacology therapeutic use MeSH
- Humans MeSH
- Adolescent MeSH
- Young Adult MeSH
- Biomarkers, Tumor MeSH
- Tumor Microenvironment MeSH
- Neoplasms drug therapy MeSH
- DNA Repair genetics MeSH
- Prospective Studies MeSH
- DNA Replication genetics MeSH
- Retrospective Studies MeSH
- Germ-Line Mutation * MeSH
- Check Tag
- Child MeSH
- Adult MeSH
- Humans MeSH
- Adolescent MeSH
- Young Adult MeSH
- Male MeSH
- Female MeSH
- Publication type
- Journal Article MeSH
- Observational Study MeSH
- Research Support, Non-U.S. Gov't MeSH
- Names of Substances
- B7-H1 Antigen MeSH
- CD274 protein, human MeSH Browser
- Immune Checkpoint Inhibitors MeSH
- Biomarkers, Tumor MeSH
Cancers arising from germline DNA mismatch repair deficiency or polymerase proofreading deficiency (MMRD and PPD) in children harbour the highest mutational and microsatellite insertion-deletion (MS-indel) burden in humans. MMRD and PPD cancers are commonly lethal due to the inherent resistance to chemo-irradiation. Although immune checkpoint inhibitors (ICIs) have failed to benefit children in previous studies, we hypothesized that hypermutation caused by MMRD and PPD will improve outcomes following ICI treatment in these patients. Using an international consortium registry study, we report on the ICI treatment of 45 progressive or recurrent tumors from 38 patients. Durable objective responses were observed in most patients, culminating in a 3 year survival of 41.4%. High mutation burden predicted response for ultra-hypermutant cancers (>100 mutations per Mb) enriched for combined MMRD + PPD, while MS-indels predicted response in MMRD tumors with lower mutation burden (10-100 mutations per Mb). Furthermore, both mechanisms were associated with increased immune infiltration even in 'immunologically cold' tumors such as gliomas, contributing to the favorable response. Pseudo-progression (flare) was common and was associated with immune activation in the tumor microenvironment and systemically. Furthermore, patients with flare who continued ICI treatment achieved durable responses. This study demonstrates improved survival for patients with tumors not previously known to respond to ICI treatment, including central nervous system and synchronous cancers, and identifies the dual roles of mutation burden and MS-indels in predicting sustained response to immunotherapy.
Atrium Health Levine Children's Hospital Charlotte NC USA
Biotechnology and Food Engineering Technion Israel Institute of Technology Tel Aviv Israel
Broad Institute of Harvard and MIT Cambridge MA USA
Dalla Lana School of Public Health University of Toronto Toronto Ontario Canada
Department of Basic Medical Sciences Faculty of Medicine The Hashemite University Zarqa Jordan
Department of Diagnostic Imaging The Hospital for Sick Children Toronto Ontario Canada
Department of Immunology University of Toronto Toronto Ontario Canada
Department of Laboratory Medicine and Pathobiology University of Toronto Toronto Ontario Canada
Department of Medical Biophysics University of Toronto Toronto Ontario Canada
Department of Neurosurgery Neurological Institute Taipei Veterans General Hospital Taipei Taiwan
Department of Oncology Sahlgrenska University Hospital Gothenburg Sweden
Department of Paediatric Haematology Oncology Tata Medical Centre Kolkata India
Department of Paediatric Laboratory Medicine The Hospital for Sick Children Toronto Ontario Canada
Department of Paediatrics University of Melbourne Parkville Victoria Australia
Department of Paediatrics University of Toronto Toronto Ontario Canada
Department of Pediatric Hematology Oncology Rambam Health Care Campus Haifa Israel
Department of Pediatric Hematology Oncology Sheba Medical Centre Ramat Gan Israel
Department of Pediatric Hematology Oncology Tel Aviv Sourasky Medical Centre Tel Aviv Israel
Department of Pediatric Neurosurgery Dana Children's Hospital Tel Aviv Israel
Department of Pediatric Oncology Valley Children's Hospital Madera CA USA
Department of Pediatrics Anschutz Medical Campus Children's Hospital of Colorado Aurora CO USA
Department of Pediatrics J W Ruby Memorial Hospital West Virginia University Morgantown WV USA
Department of Pediatrics The University of Texas Southwestern Medical School Dallas TX USA
Department of Pediatrics University of Pittsburgh School of Medicine Pittsburgh PA USA
Departments of Neurology and Pediatrics University of California San Francisco CA USA
Developmental and Stem Cell Biology Program The Hospital for Sick Children Toronto Ontario Canada
Division of Haematology Oncology The Hospital for Sick Children Toronto Ontario Canada
Division of Neurosurgery The Hospital for Sick Children Toronto Ontario Canada
Division of Pediatric Hematology Oncology BMT Medical College of Wisconsin Milwaukee WI USA
Institute of Medical Science Faculty of Medicine University of Toronto Toronto Ontario Canada
Kids Cancer Centre Sydney Children's Hospital Randwick New South Wales Australia
Lux Med Onkologia Warsaw Poland
Massachusetts General Hospital Cancer Center and Department of Pathology Charlestown MA USA
Ontario Institute for Cancer Research Toronto Ontario Canada
Paediatric Haematology and Oncology University Hospital Frankfurt Frankfurt Germany
Paediatric Unit Fondazione IRCCS Istituto Nazionale dei Tumori Milan Italy
Pediatric Hematology Oncology Helen DeVos Children's Hospital Grand Rapids MI USA
Phoenix Children's Hospital Phoenix AZ USA
Princess Margaret Cancer Centre University Health Network Toronto Ontario Canada
Program in Cell Biology The Hospital for Sick Children Toronto Ontario Canada
Program in Genetics and Genome Biology The Hospital for Sick Children Toronto Ontario Canada
Queen Silvia Children's Hospital Sahlgrenska University Hospital Gothenburg Sweden
Radiation Medicine Program Princess Margaret Cancer Centre Toronto Ontario Canada
Women's and Children's Hospital North Adelaide South Australia Australia
Zane Cohen Centre for Digestive Diseases Mount Sinai Hospital Toronto Ontario Canada
See more in PubMed
Cortez D. Replication-coupled DNA repair. Mol. Cell. 2019;74:866–876. PubMed PMC
Campbell BB, et al. Comprehensive analysis of hypermutation in human cancer. Cell. 2017;171:1042–1056. PubMed PMC
Chung J, et al. DNA polymerase and mismatch repair exert distinct microsatellite instability signatures in normal and malignant human cells. Cancer Discov. 2021;11:1176–1191. PubMed PMC
Mur P, et al. Role of POLE and POLD1 in familial cancer. Genet. Med. 2020;22:2089–2100. PubMed PMC
Seppälä TT, et al. European guidelines from the EHTG and ESCP for Lynch syndrome: an updated third edition of the Mallorca guidelines based on gene and gender. Br. J. Surg. 2021;108:484–498. PubMed PMC
Tabori U, et al. Clinical management and tumor surveillance recommendations of inherited mismatch repair deficiency in childhood. Clin. Cancer Res. 2017;23:e32–e37. PubMed
Amayiri N, et al. High frequency of mismatch repair deficiency among pediatric high grade gliomas in Jordan. Int. J. Cancer. 2016;138:380–385. PubMed
Shlien A, et al. Combined hereditary and somatic mutations of replication error repair genes result in rapid onset of ultra-hypermutated cancers. Nat. Genet. 2015;47:257–262. PubMed
Liu D, et al. Integrative molecular and clinical modeling of clinical outcomes to PD1 blockade in patients with metastatic melanoma. Nat. Med. 2019;25:1916–1927. PubMed PMC
Garon EB, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N. Engl. J. Med. 2015;372:2018–2028. PubMed
Topalian SL’, et al. Five-year survival and correlates among patients with advanced melanoma, renal cell carcinoma, or non-small cell lung cancer treated with nivolumab. JAMA Oncol. 2019;5:1411–1420. PubMed PMC
McGrail DJ, et al. High tumor mutation burden fails to predict immune checkpoint blockade response across all cancer types. Ann. Oncol. 2021;32:661–672. PubMed PMC
Rousseau B, et al. The spectrum of benefit from checkpoint blockade in hypermutated tumors. N. Engl. J. Med. 2021;384:1168–1170. PubMed PMC
Gromeier M, et al. Very low mutation burden is a feature of inflamed recurrent glioblastomas responsive to cancer immunotherapy. Nat. Commun. 2021;12:352. PubMed PMC
Geoerger B, et al. Pembrolizumab in paediatric patients with advanced melanoma or a PD-L1-positive, advanced, relapsed, or refractory solid tumour or lymphoma (KEYNOTE-051): interim analysis of an open-label, single-arm, phase 1-2 trial. Lancet Oncol. 2020;21:121–133. PubMed
Le DT, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N. Engl. J. Med. 2015;372:2509–2520. PubMed PMC
Le DT, et al. Phase II open-label study of pembrolizumab in treatment-refractory, microsatellite instability-high/mismatch repair-deficient metastatic colorectal cancer: KEYNOTE-164. J. Clin. Oncol. 2020;38:11–19. PubMed PMC
André T, et al. Pembrolizumab in microsatellite-instability-high advanced colorectal cancer. N. Engl. J. Med. 2020;383:2207–2218. PubMed
Marabelle A, et al. Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study. Lancet Oncol. 2020;21:1353–1365. PubMed
Schrock AB, et al. Tumor mutational burden is predictive of response to immune checkpoint inhibitors in MSI-high metastatic colorectal cancer. Ann. Oncol. 2019;30:1096–1103. PubMed
Marabelle A, et al. Efficacy of pembrolizumab in patients with noncolorectal high microsatellite instability/mismatch repair-deficient cancer: results from the phase II KEYNOTE-158 study. J. Clin. Oncol. 2020;38:1–10. PubMed PMC
Sampson JH, Gunn MD, Fecci PE, Ashley DM. Brain immunology and immunotherapy in brain tumours. Nat. Rev. Cancer. 2020;20:12–25. PubMed PMC
Geoerger B, et al. Atezolizumab for children and young adults with previously treated solid tumours, non-Hodgkin lymphoma, and Hodgkin lymphoma (iMATRIX): a multicentre phase 1-2 study. Lancet Oncol. 2020;21:134–144. PubMed
Merchant MS, et al. Phase I clinical trial of ipilimumab in pediatric patients with advanced solid tumors. Clin. Cancer Res. 2016;22:1364–1370. PubMed PMC
Davis KL, et al. Nivolumab in children and young adults with relapsed or refractory solid tumours or lymphoma (ADVL1412): a multicentre, open-label, single-arm, phase 1-2 trial. Lancet Oncol. 2020;21:541–550. PubMed PMC
Martin-Romano P, et al. Evidence of pseudoprogression in patients treated with PD1/PDL1 antibodies across tumor types. Cancer Med. 2020;9:2643–2652. PubMed PMC
Bouffet E, et al. Immune checkpoint inhibition for hypermutant glioblastoma multiforme resulting from germline biallelic mismatch repair deficiency. J. Clin. Oncol. 2016;34:2206–2211. PubMed
Wen PY, et al. Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group. J. Clin. Oncol. 2010;28:1963–1972. PubMed
Eisenhauer EA, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1) Eur. J. Cancer. 2009;45:228–247. PubMed
Yoshida T, Furuta H, Hida T. Risk of tumor flare after nivolumab treatment in patients with irradiated field recurrence. Med. Oncol. 2017;34:34. PubMed
Alexandrov LB, et al. The repertoire of mutational signatures in human cancer. Nature. 2020;578:94–101. PubMed PMC
Touat M, et al. Mechanisms and therapeutic implications of hypermutation in gliomas. Nature. 2020;580:517–523. PubMed PMC
McGranahan N, et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science. 2016;351:1463–1469. PubMed PMC
Le DT, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357:409–413. PubMed PMC
Mandal R, et al. Genetic diversity of tumors with mismatch repair deficiency influences anti-PD-1 immunotherapy response. Science. 2019;364:485–491. PubMed PMC
Maruvka YE, et al. Analysis of somatic microsatellite indels identifies driver events in human tumors. Nat. Biotechnol. 2017;35:951–959. PubMed PMC
Llosa NJ, et al. The vigorous immune microenvironment of microsatellite instable colon cancer is balanced by multiple counter-inhibitory checkpoints. Cancer Discov. 2015;5:43–51. PubMed PMC
Huang AC, et al. T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature. 2017;545:60–65. PubMed PMC
Watts TH. TNF/TNFR family members in costimulation of T cell responses. Annu. Rev. Immunol. 2005;23:23–68. PubMed
Clouthier DL, et al. An interim report on the investigator-initiated phase 2 study of pembrolizumab immunological response evaluation (INSPIRE) J. Immunother. Cancer. 2019;7:72. PubMed PMC
Blumenthal DT, et al. Pembrolizumab: first experience with recurrent primary central nervous system (CNS) tumors. J. Neurooncol. 2016;129:453–460. PubMed
Haworth KB, et al. Going back to class I: MHC and immunotherapies for childhood cancer. Pediatr. Blood Cancer. 2015;62:571–576. PubMed PMC
Majzner RG, et al. Assessment of programmed death-ligand 1 expression and tumor-associated immune cells in pediatric cancer tissues. Cancer. 2017;123:3807–3815. PubMed
Khasraw M, Reardon DA, Weller M, Sampson JH. PD-1 inhibitors: do they have a future in the treatment of glioblastoma? Clin. Cancer Res. 2020;26:5287–5296. PubMed PMC
Kato S, et al. Expression of TIM3/VISTA checkpoints and the CD68 macrophage-associated marker correlates with anti-PD1/PDL1 resistance: implications of immunogram heterogeneity. Oncoimmunology. 2020;9:1708065. PubMed PMC
Stahl M, et al. Risk of recurrence and survival after relapse in patients with Ewing sarcoma. Pediatr. Blood Cancer. 2011;57:549–553. PubMed
London WB, et al. Historical time to disease progression and progression-free survival in patients with recurrent/refractory neuroblastoma treated in the modern era on Children’s Oncology Group early-phase trials. Cancer. 2017;123:4914–4923. PubMed PMC
Hayes-Jordan AA, et al. Colon cancer in patients under 25 years old: a different disease? J. Am. Coll. Surg. 2020;230:648–656. PubMed
Akinkuotu AC, Maduekwe UN, Hayes-Jordan A. Surgical outcomes and survival rates of colon cancer in children and young adults. Am. J. Surg. 2021;221:718–724. PubMed
Wang J, et al. Metastatic patterns and survival outcomes in patients with stage IV colon cancer: a population-based analysis. Cancer Med. 2020;9:361–373. PubMed PMC
Bajorin DF, et al. Long-term survival in metastatic transitional-cell carcinoma and prognostic factors predicting outcome of therapy. J. Clin. Oncol. 1999;17:3173–3181. PubMed
Azad NS, et al. Nivolumab Is effective in mismatch repair-deficient noncolorectal cancers: results from arm Z1D-A subprotocol of the NCI-MATCH (EAY131) Study. J. Clin. Oncol. 2020;38:214–222. PubMed PMC
Thomas R, Wang W, Su DM. Contributions of age-related thymic involution to immunosenescence and inflammaging. Immun. Ageing. 2020;17:2. PubMed PMC
Campbell BB, et al. Mutations in the RAS/MAPK pathway drive replication repair deficient hypermutated tumors and confer sensitivity to MEK inhibition. Cancer Discov. 2021;11:1454–1467. PubMed PMC
Cloughesy TF, et al. Neoadjuvant anti-PD-1 immunotherapy promotes a survival benefit with intratumoral and systemic immune responses in recurrent glioblastoma. Nat. Med. 2019;25:477–486. PubMed PMC
Bai R, Lv Z, Xu D, Cui J. Predictive biomarkers for cancer immunotherapy with immune checkpoint inhibitors. Biomark. Res. 2020;8:34. PubMed PMC
Turajlic S, et al. Insertion-and-deletion-derived tumour-specific neoantigens and the immunogenic phenotype: a pan-cancer analysis. Lancet Oncol. 2017;18:1009–1021. PubMed
Terry RL, et al. Immune profiling of pediatric solid tumors. J. Clin. Invest. 2020;130:3391–3402. PubMed PMC
Platten M, et al. A vaccine targeting mutant IDH1 in newly diagnosed glioma. Nature. 2021;592:463–468. PubMed PMC
Antonios JP, et al. Detection of immune responses after immunotherapy in glioblastoma using PET and MRI. Proc. Natl Acad. Sci. USA. 2017;114:10220–10225. PubMed PMC
Kim KH, et al. The first-week proliferative response of peripheral blood PD-1(+)CD8(+) T cells predicts the response to anti-PD-1 therapy in solid tumors. Clin. Cancer Res. 2019;25:2144–2154. PubMed
Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25:1754–1760. PubMed PMC
DePristo MA, et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat. Genet. 2011;43:491–498. PubMed PMC
McKenna A, et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 2010;20:1297–1303. PubMed PMC
Van der Auwera GA, et al. From FastQ data to high confidence variant calls: the Genome Analysis Toolkit best practices pipeline. Curr. Protoc. Bioinformatics. 2013;43:11.10.1–11.10.33. PubMed PMC
Yang SYC, et al. Landscape of genomic alterations in high-grade serous ovarian cancer from exceptional long- and short-term survivors. Genome Med. 2018;10:81. PubMed PMC
Cibulskis K, et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat. Biotechnol. 2013;31:213–219. PubMed PMC
Saunders CT, et al. Strelka: accurate somatic small-variant calling from sequenced tumor-normal sample pairs. Bioinformatics. 2012;28:1811–1817. PubMed
Koboldt DC, et al. VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res. 2012;22:568–576. PubMed PMC
Danecek P, et al. The variant call format and VCFtools. Bioinformatics. 2011;27:2156–2158. PubMed PMC
McLaren W, et al. The Ensembl Variant Effect Predictor. Genome Biol. 2016;17:122. PubMed PMC
Rosenthal R, McGranahan N, Herrero J, Taylor BS, Swanton C. DeconstructSigs: delineating mutational processes in single tumors distinguishes DNA repair deficiencies and patterns of carcinoma evolution. Genome Biol. 2016;17:31. PubMed PMC
Warren RL, et al. Derivation of HLA types from shotgun sequence datasets. Genome Med. 2012;4:95. PubMed PMC
Nariai N, et al. HLA-VBSeq: accurate HLA typing at full resolution from whole-genome sequencing data. BMC Genomics. 2015;16(Suppl. 2):S7. PubMed PMC
Iafolla MAJ, et al. Predicting toxicity and response to pembrolizumab through germline genomic HLA class 1 analysis. JNCI Cancer Spectr. 2021;5:pkaa115. PubMed PMC
Bjerregaard AM, Nielsen M, Hadrup SR, Szallasi Z, Eklund AC. MuPeXI: prediction of neo-epitopes from tumor sequencing data. Cancer Immunol. Immunother. 2017;66:1123–1130. PubMed PMC
Jurtz V, et al. NetMHCpan-4.0: improved peptide-MHC class I interaction predictions integrating eluted ligand and peptide binding affinity data. J. Immunol. 2017;199:3360–3368. PubMed PMC
Hundal J, et al. pVAC-Seq: a genome-guided in silico approach to identifying tumor neoantigens. Genome Med. 2016;8:11. PubMed PMC
Shen R, Seshan VE. FACETS: allele-specific copy number and clonal heterogeneity analysis tool for high-throughput DNA sequencing. Nucleic Acids Res. 2016;44:e131. PubMed PMC
Carter SL, et al. Absolute quantification of somatic DNA alterations in human cancer. Nat. Biotechnol. 2012;30:413–421. PubMed PMC
Ionescu DN, Downes MR, Christofides A, Tsao MS. Harmonization of PD-L1 testing in oncology: a Canadian pathology perspective. Curr. Oncol. 2018;25:e209–e216. PubMed PMC
Dobin A, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29:15–21. PubMed PMC
Li B, Dewey CN. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics. 2011;12:323. PubMed PMC
Newman AM, et al. Robust enumeration of cell subsets from tissue expression profiles. Nat. Methods. 2015;12:453–457. PubMed PMC
Newman AM, et al. Determining cell type abundance and expression from bulk tissues with digital cytometry. Nat. Biotechnol. 2019;37:773–782. PubMed PMC
Chen B, Khodadoust MS, Liu CL, Newman AM, Alizadeh AA. Profiling tumor infiltrating immune cells with CIBERSORT. Methods Mol. Biol. 2018;1711:243–259. PubMed PMC
Sturm G, et al. Comprehensive evaluation of transcriptome-based cell-type quantification methods for immuno-oncology. Bioinformatics. 2019;35:i436–i445. PubMed PMC
Rohr-Udilova N, et al. Deviations of the immune cell landscape between healthy liver and hepatocellular carcinoma. Sci. Rep. 2018;8:6220. PubMed PMC
Gu Z, Eils R, Schlesner M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics. 2016;32:2847–2849. PubMed
Racle J, Gfeller D. EPIC: a tool to estimate the proportions of different cell types from bulk gene expression data. Methods Mol. Biol. 2020;2120:233–248. PubMed
Mulder DT, et al. CapTCR-seq: hybrid capture for T-cell receptor repertoire profiling. Blood Adv. 2018;2:3506–3514. PubMed PMC
Bolotin DA, et al. MiXCR: software for comprehensive adaptive immunity profiling. Nat. Methods. 2015;12:380–381. PubMed
Kaplinsky J, Arnaout R. Robust estimates of overall immune-repertoire diversity from high-throughput measurements on samples. Nat. Commun. 2016;7:11881. PubMed PMC
Overt and covert genetic causes of pediatric acute lymphoblastic leukemia