Pollack, I. F. Mind tumors in kids. N. Engl. J. Med. 331, 1500–1507 (1994).
Jones, C. & Baker, S.J. Distinctive genetic and epigenetic mechanisms driving paediatric diffuse high-grade glioma. Nat. Rev. Most cancers 14, 651–661 (2014).
Chevignard, M., Câmara-Costa, H., Doz, F. & Dellatolas, G. Core deficits and high quality of survival after childhood medulloblastoma: a evaluate. Neurooncol. Pract. 4, 82–97 (2017).
Makale, M. T., McDonald, C. R., Hattangadi-Gluth, J. A. & Kesari, S. Mechanisms of radiotherapy-associated cognitive incapacity in sufferers with mind tumours. Nat. Rev. Neurol. 13, 52–64 (2017).
Hwang, E. I. et al. The present panorama of immunotherapy for pediatric mind tumors. Nat. Most cancers 3, 11–24 (2022).
Dunkel, I. J. et al. Nivolumab with or with out ipilimumab in pediatric sufferers with high-grade CNS malignancies: security, efficacy, biomarker, and pharmacokinetics: checkMate 908. Neuro. Oncol. 25, 1530–1545 (2023).
Bouffet, E. et al. Immune checkpoint inhibition for hypermutant glioblastoma multiforme ensuing from germline biallelic mismatch restore deficiency. J. Clin. Oncol. 34, 2206–2211 (2016).
Fried, I. et al. Preliminary outcomes of immune modulating antibody MDV9300 (pidilizumab) therapy in kids with diffuse intrinsic pontine glioma. J. Neurooncol. 136, 189–195 (2018).
Binnewies, M. et al. Understanding the tumor immune microenvironment (TIME) for efficient remedy. Nat. Med. 24, 541–550 (2018).
Bruni, D., Angell, H. Ok. & Galon, J. The immune contexture and Immunoscore in most cancers prognosis and therapeutic efficacy. Nat. Rev. Most cancers 20, 662–680 (2020).
Tumeh, P. C. et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 515, 568–571 (2014).
Patel, R. R., Ramkissoon, S. H., Ross, J. & Weintraub, L. Tumor mutational burden and driver mutations: characterizing the genomic panorama of pediatric mind tumors. Pediatr. Blood Most cancers 67, e28338 (2020).
Mok, T. S. Ok. et al. Pembrolizumab versus chemotherapy for beforehand untreated, PD-L1-expressing, regionally superior or metastatic non-small-cell lung most cancers (KEYNOTE-042): a randomised, open-label, managed, section 3 trial. Lancet 393, 1819–1830 (2019).
Schumacher, T. N. & Schreiber, R. D. Neoantigens in most cancers immunotherapy. Science 348, 69–74 (2015).
Gubin, M. M. et al. Checkpoint blockade most cancers immunotherapy targets tumour-specific mutant antigens. Nature 515, 577–581 (2014).
Robbins, P. F. et al. Mining exomic sequencing information to determine mutated antigens acknowledged by adoptively transferred tumor-reactive T cells. Nat. Med. 19, 747–752 (2013).
Tran, E. et al. T-cell switch remedy concentrating on mutant KRAS in most cancers. N. Engl. J. Med. 375, 2255–2262 (2016).
Carreno, B. M. et al. Most cancers immunotherapy. A dendritic cell vaccine will increase the breadth and variety of melanoma neoantigen-specific T cells. Science 348, 803–808 (2015).
Ott, P. A. et al. An immunogenic private neoantigen vaccine for sufferers with melanoma. Nature 547, 217–221 (2017).
Lowery, F. J. et al. Molecular signatures of antitumor neoantigen-reactive T cells from metastatic human cancers. Science 375, 877–884 (2022).
Kvistborg, P. & Yewdell, J. W. Enhancing responses to most cancers immunotherapy. Science 359, 516–517 (2018).
Chiou, S. H. et al. World evaluation of shared T cell specificities in human non-small cell lung most cancers permits HLA inference and antigen discovery. Immunity 54, 586–602.e8 (2021).
Ganesan, A. P. et al. Tissue-resident reminiscence options are linked to the magnitude of cytotoxic T cell responses in human lung most cancers. Nat. Immunol. 18, 940–950 (2017).
Clarke, J. et al. Single-cell transcriptomic evaluation of tissue-resident reminiscence T cells in human lung most cancers. J. Exp. Med. 216, 2128–2149 (2019).
Savas, P. et al. Single-cell profiling of breast most cancers T cells reveals a tissue-resident reminiscence subset related to improved prognosis. Nat. Med. 24, 986–993 (2018).
Han, J. et al. Resident and circulating reminiscence T cells persist for years in melanoma sufferers with sturdy responses to immunotherapy. Nat. Most cancers 2, 300–311 (2021).
Liu, B. et al. Temporal single-cell tracing reveals clonal revival and growth of precursor exhausted T cells throughout anti-PD-1 remedy in lung most cancers. Nature Most cancers 3, 108–121 (2022).
Li, H. et al. Dysfunctional CD8 T cells type a proliferative, dynamically regulated compartment inside human melanoma. Cell 176, 775–789.e18 (2019).
Sade-Feldman, M. et al. Defining T cell states related to response to checkpoint immunotherapy in melanoma. Cell 175, 998–1013.e20 (2018).
Yost, Ok. E. et al. Clonal substitute of tumor-specific T cells following PD-1 blockade. Nat. Med. 25, 1251–1259 (2019).
Gide, T. N. et al. Distinct immune cell populations outline response to anti-PD-1 monotherapy and anti-PD-1/anti-CTLA-4 mixed remedy. Most cancers Cell 35, 238–255.e6 (2019).
Kurtulus, S. et al. Checkpoint blockade immunotherapy induces dynamic adjustments in PD-1−CD8+ tumor-Infiltrating T cells. Immunity 50, 181–194.e6 (2019).
Siddiqui, I. et al. Intratumoral Tcf1+PD-1+CD8+ T cells with stem-like properties promote tumor management in response to vaccination and checkpoint blockade immunotherapy. Immunity 50, 195–211.e10 (2019).
Castellino, F. et al. Chemokines improve immunity by guiding naive CD8+ T cells to websites of CD4+ T cell-dendritic cell interplay. Nature 440, 890–895 (2006).
Brewitz, A. et al. CD8+ T cells orchestrate pDC-XCR1+ dendritic cell spatial and practical cooperativity to optimize priming. Immunity 46, 205–219 (2017).
Mackay, L. Ok. et al. Hobit and Blimp1 instruct a common transcriptional program of tissue residency in lymphocytes. Science 352, 459–463 (2016).
Mackay, L. Ok. et al. The developmental pathway for CD103+CD8+ tissue-resident reminiscence T cells of pores and skin. Nat. Immunol. 14, 1294–1301 (2013).
Lim, M. et al. Part III trial of chemoradiotherapy with temozolomide plus nivolumab or placebo for newly recognized glioblastoma with methylated MGMT promoter. Neuro. Oncol. 24, 1935–1949 (2022).
Omuro, A. et al. Radiotherapy mixed with nivolumab or temozolomide for newly recognized glioblastoma with unmethylated MGMT promoter: a world randomized section III trial. Neuro. Oncol. 25, 123–134 (2022).
Mathewson, N. D. et al. Inhibitory CD161 receptor recognized in glioma-infiltrating T cells by single-cell evaluation. Cell 184, 1281–1298.e26 (2021).
Corridoni, D. et al. Single-cell atlas of colonic CD8+ T cells in ulcerative colitis. Nat. Med. 26, 1480–1490 (2020).
Jonsson, A. H. et al. Granzyme Ok+ CD8 T cells type a core inhabitants in infected human tissue. Sci. Transl. Med. 14, eabo0686 (2022).
Xu, T. et al. Single-cell profiling reveals pathogenic position and differentiation trajectory of granzyme Ok+CD8+ T cells in major Sjögren’s syndrome. JCI Perception 8, e167490 (2023).
Trapnell, C. et al. The dynamics and regulators of cell destiny selections are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol. 32, 381–386 (2014).
Baitsch, L. et al. Exhaustion of tumor-specific CD8+ T cells in metastases from melanoma sufferers. J. Clin. Make investments. 121, 2350–2360 (2011).
Gros, A. et al. PD-1 identifies the patient-specific CD8+ tumor-reactive repertoire infiltrating human tumors. J. Clin. Make investments. 124, 2246–2259 (2014).
Thommen, D. S. et al. A transcriptionally and functionally distinct PD-1+ CD8+ T cell pool with predictive potential in non-small-cell lung most cancers handled with PD-1 blockade. Nat. Med. 24, 994–1004 (2018).
Chen, R. et al. Antigen presentation equipment signature-derived CALR mediates migration, polarization of macrophages in glioma and predicts immunotherapy response. Entrance. Immunol. 13, 833792 (2022).
Sledzinska, A. et al. Regulatory T cells restrain Interleukin-2- and Blimp-1-dependent acquisition of cytotoxic perform by CD4+ T cells. Immunity 52, 151–166.e6 (2020).
Eschweiler, S. et al. Intratumoral follicular regulatory T cells curtail anti-PD-1 therapy efficacy. Nat. Immunol. 22, 1052–1063 (2021).
Awad, M. M. et al. Customized neoantigen vaccine NEO-PV-01 with chemotherapy and anti-PD-1 as first-line therapy for non-squamous non-small cell lung most cancers. Most cancers Cell 40, 1010–1026.e11 (2022).
Ott, P. A. et al. A section Ib trial of personalised neoantigen remedy plus Anti-PD-1 in sufferers with superior melanoma, non-small cell lung most cancers, or bladder most cancers. Cell 183, 347–362.e24 (2020).
Pittet, M. J. et al. Excessive frequencies of naive Melan-A/MART-1-specific CD8+ T cells in a big proportion of human histocompatibility leukocyte antigen (HLA)-A2 people. J. Exp. Med. 190, 705–715 (1999).
Schmidt, N., Flecken, T. & Thimme, R. Tumor-associated antigen particular CD8+ T cells in hepatocellular carcinoma—a promising goal for immunotherapy. Oncoimmunology 3, e954919 (2014).
Moustaki, A. et al. Antigen cross-presentation in younger tumor-bearing hosts promotes CD8+ T cell terminal differentiation. Sci. Immunol. 7, eabf6136 (2022).
Sekine, T. et al. TOX is expressed by exhausted and polyfunctional human effector reminiscence CD8+ T cells. Sci. Immunol. 5, eaba7918 (2020).
Alfei, F. et al. TOX reinforces the phenotype and longevity of exhausted T cells in power viral an infection. Nature 571, 265–269 (2019).
Alspach, E. et al. MHC-II neoantigens form tumour immunity and response to immunotherapy. Nature 574, 696–701 (2019).
Gorsi, H. S. et al. Nivolumab within the therapy of recurrent or refractory pediatric mind tumors: a single institutional expertise. J. Pediatr. Hematol. Oncol. 41, e235–e241 (2019).
Mascarenhas, L. et al. Part 1 scientific trial of durvalumab in kids with strong and central nervous system tumors. J. Clin. Oncol. 40, 10029 (2022).
Meckiff, B. J. et al. Imbalance of regulatory and cytotoxic SARS-CoV-2-reactive CD4+ T cells in COVID-19. Cell 183, 1340–1353.e16 (2020).
Wolock, S. L., Lopez, R. & Klein, A. M. Scrublet: computational identification of cell doublets in single-cell transcriptomic information. Cell Syst. 8, 281–291.e89 (2019).
Stuart, T. et al. Complete integration of single-cell information. Cell 177, 1888–1902.e21 (2019).
Tran, H. T. N. et al. A benchmark of batch-effect correction strategies for single-cell RNA sequencing information. Genome Biol. 21, 12 (2020).
Korsunsky, I. et al. Quick, delicate and correct integration of single-cell information with Concord. Nat. Strategies. 16, 1289–1296 (2019).
Denisenko, E. et al. Systematic evaluation of tissue dissociation and storage biases in single-cell and single-nucleus RNA-seq workflows. Genome Biol. 21, 130 (2020).
van den Brink, S. C. et al. Single-cell sequencing reveals dissociation-induced gene expression in tissue subpopulations. Nat. Strategies 14, 935–936 (2017).
Behr, F. M. et al. Blimp-1 fairly than Hobit drives the formation of tissue-resident reminiscence CD8+ T cells within the lungs. Entrance. Immunol. 10, 400 (2019).
Hänzelmann, S., Castelo, R. & Guinney, J. GSVA: gene set variation evaluation for microarray and RNA-seq information. BMC Bioinf. 14, 7 (2013).
Conway, J. R., Lex, A. & Gehlenborg, N. UpSetR: an R bundle for the visualization of intersecting units and their properties. Bioinformatics 33, 2938–2940 (2017).
Shugay, M. et al. VDJtools: unifying post-analysis of T cell receptor repertoires. PLoS Comput. Biol. 11, e1004503 (2015).
Csárdi, G. & Nepusz, T. The igraph software program bundle for complicated community analysis. InterJ. Complicated Syst. 1695, 1–9 (2006).
McCann, Ok. et al. Concentrating on the tumor mutanome for personalised vaccination in a TMB low non-small cell lung most cancers. J. Immunother. Most cancers 10, e003821 (2022).
Therneau, T. M. & Grambsch, P. M. in Modeling Survival Information: Extending the Cox Mannequin (eds Therneau T. M. & Grambsch P. M.) 39–77 (Springer, 2000).