Characterization of neoantigen-specific T cells in cancer resistant to immune checkpoint therapies Shamin Lia,1, Yannick Simonia,b, Summer Zhuanga, Austin Gabelc,d,e,f, Shaokang Mag, Jonathan Cheeg, Laura Islasa, Anthony Cessnaa, Jenette Creaneyg,h,i, Robert K. Bradleyc,d,e, Alec Redwoodg, Bruce W. Robinsong, and Evan W. Newella,1 aVaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109; bImmunoSCAPE, Pte Ltd, Singapore, 228208 Singapore; cComputational Biology Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109; dBasic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109; eDepartment of Genome Sciences, University of Washington, Seattle, WA 98195; fMedical Scientist Training Program, University of Washington, Seattle, WA 98195; gNational Centre for Asbestos Related Disease, Faculty of Health and Medical Science, University of Western Australia, 6009 Perth, Australia; hDepartment of Respiratory Medicine, Sir Charles Gairdner Hospital, 6009 Perth, Australia; and iInstitute for Respiratory Health, University of Western Australia, 6009 Perth, Australia Edited by Rafi Ahmed, Emory University, Atlanta, GA, and approved June 16, 2021 (received for review December 14, 2020) Neoantigen-specific T cells are strongly implicated as being critical for (MCA) sarcoma for instance, two immunodominant neoantigens effective immune checkpoint blockade treatment (ICB) (e.g., anti–PD-1 have been identified and showed to be reactivated upon anti–PD-1 and anti–CTLA-4) and are being targeted for vaccination-based thera- and/or anti–CTLA-4 blockade, enabling an effective tumor rejec- pies. However, ICB treatments show uneven responses between pa- tion that could be boosted via neopeptide-based vaccination (21). tients, and neoantigen vaccination efficiency has yet to be established. Notably, using mass cytometry, CD8+ tumor-infiltrating lympho- Here, we characterize neoantigen-specific CD8+ Tcellsinatumorthat cytes (TILs) that are specific for these neoantigens showed a dra- is resistant to ICB and neoantigen vaccination. Leveraging the use of matic phenotypic change between treated versus untreated tumors mass cytometry combined with multiplex major histocompatibility (22, 24), leading to the question as to whether the phenotypes of complex (MHC) class I tetramer staining, we screened and identified neoantigen-specific CD8+ TILs could accurately predict treatment + tumor neoantigen–specific CD8 T cells in the Lewis Lung carcinoma outcome. This concept has been extensively studied in the recent (LLC) tumor model (mRiok1). We observed an expansion of mRiok1- years to identify reliable biomarkers for immunotherapy response, + specific CD8 tumor-infiltrating lymphocytes (TILs) after ICB targeting and numerous works have addressed the paradigm of “exhausted” IMMUNOLOGY AND INFLAMMATION PD-1 or CTLA-4 with no sign of tumor regression. The expanded versus “precursor exhausted” Tcells(25–29). While “exhausted” + neoantigen-specific CD8 TILs remained phenotypically and function- Tcellsareoftenassociatedwithdysfunction and reduced capacity ally exhausted but displayed cytotoxic characteristics. When combin- “ ” + to control tumor growth, precursor exhausted refers to T cells ing both ICB treatments, mRiok1-specific CD8 TILs showed a stem-like capable of proliferation after immunotherapy that maintain phenotype and a higher capacity to produce cytokines, but tumors did memory characteristics and a long-term effective T cell response not show signs of regression. Furthermore, combining both ICB treat- (27). As each of these populations is well characterized with a ments with neoantigen vaccination did not induce tumor regression either despite neoantigen-specific CD8+ TIL expansion. Overall, this Significance work provides a model for studying neoantigens in an immunother- apy nonresponder model. We showed that a robust neoantigen- specific T-cell response in the LLC tumor model could fail in tumor Strongly implicated in effective antitumor immune responses, – response to ICB, which will have important implications in designing tumor mutation derived antigens, or neoantigens, are one of the future immunotherapeutic strategies. main targets for cancer vaccines despite uncertainty of the efficacy of this approach. Using a high-throughput screening method, we LLC | CyTOF | immunotherapies | neoantigen | vaccination identify an endogenously immunogenic neoantigen in a com- monly used mouse lung tumor model. We also found that the endogenous CD8 T cells specific for this neoantigen expand urrently, one of the most successful immunotherapeutic in- greatly upon treatment with immune checkpoint inhibitors or Cterventions for treating cancers is targeting T cell checkpoints vaccination despite the lack of associated tumor regression in this such as PD-1 or CTLA-4, that improve the effector T cell response model. In addition to informing neoantigen vaccination strategies through distinct pathways (1, 2). Both inhibitors have showed and providing an accessible system for testing alternative thera- promising results, improving both patient outcomes and overall peutics, our results provide insights into the mechanisms for the survival. However, with an unpredictable and uneven response lack of response observed for a majority of patients treated with across different cancer types and treatment settings (3, 4), the checkpoint blockade immunotherapies. efficacy of these therapies needs to be improved. — In this context, presence of neoantigens in the tumor cells Author contributions: S.L. designed research; S.L., S.Z., A.G., S.M., L.I., and A.C. performed unique mutated proteins derived from either viral oncogenic pro- research; E.W.N. contributed new reagents/analytic tools; S.L., Y.S., J. Chee, J. Creaney, teins or nonsynonymous somatic mutations—elicits a potent im- R.K.B., A.R., and B.W.R. analyzed data; S.L. and E.W.N. wrote the paper; Y.S. discussed the mune response that improves treatment efficacy (5–11). While this data and reviewed the paper; S.Z. and A.G. reviewed the paper; J. Chee, J. Creaney, R.K.B., A.R., and B.W.R. discussed the data; and E.W.N. led the project. is especially true in tumors with a high tumor mutational burden Competing interest statement: E.W.N is a cofounder, advisor, and shareholder of Immu- (TMB) and thus a high neoantigen load, recent studies showed a noScape Pte. Ltd. E.W.N. is an advisor for Neogene Therapeutics and Nanostring likely better correlation between neoantigen immunogenicity and Technologies. treatment response (8, 12, 13). Although neoantigen discovery This article is a PNAS Direct Submission. could lead to some very attractive therapeutics (including adoptive This open access article is distributed under Creative Commons Attribution-NonCommercial- T cell therapy and neoantigen vaccine) (14–16), the challenges of NoDerivatives License 4.0 (CC BY-NC-ND). predicting and identifying neoantigens remain (8, 17). In addition, 1To whom correspondence may be addressed. Email: [email protected] or since TMB is not perfectly predictive, more is to be learned about [email protected]. how to efficiently tackle neoantigen-associated therapies (18, 19). This article contains supporting information online at https://www.pnas.org/lookup/suppl/ In mice, neoantigens have been studied mostly in immunother- doi:10.1073/pnas.2025570118/-/DCSupplemental. apy responder models (20–23). In d42m1-T3 methylcholanthrene Published July 20, 2021. PNAS 2021 Vol. 118 No. 30 e2025570118 https://doi.org/10.1073/pnas.2025570118 | 1of10 Downloaded by guest on September 29, 2021 unique protein, transcriptomic, and epigenetic signature, assessing lungs (SI Appendix,Fig.S1D and E). Our observations indicated the profile of tumor-specific CD8+ T cells in this setting would be the presence of exhausted neoantigen-specific CD8+ TILs that important to understand their role and reshape their response in could potentially play a role in LLC antitumor response. immunotherapy (30). Here, we use a syngeneic model of lung cancer in mice widely Neoantigen-Specific T Cells Expand in Tumors Resistant to Anti–PD-1 described as nonresponsive to immune checkpoint blockade and –CTLA-4 Therapies. Since neoantigen mRiok1-specific CD8+ (31–34). With the advantage of being derived from a spontane- TILs were exhausted and expressed PD-1, we next assessed the ous tumor, we hypothesize that neoantigen-specific T cells in efficacy of anti–PD-1 treatments in our experimental settings. We Lewis Lung carcinoma (LLC) are elicited despite a relatively low measured the tumor volume between day 7 and 16 in the control tumor mutational burden and nonresponsiveness to checkpoint group (treated with isotype IgG2a) and in mice treated with blockade immunotherapy. We first validated the neoantigen anti–PD-1 antibody (Fig. 2A). We did not observe any significant prediction performed with bioinformatic approaches by ex vivo difference in tumor growth between both groups, indicating that screening of the neopeptides using mass cytometry and further LLC tumor was resistant to anti–PD-1 therapy. Interestingly, we analyzed the role of tumor-specific CD8+ T cells in tumors re- detected a significant expansion of mRiok1-specific CD8+ TILs in sistant to immune checkpoint blockade therapy. anti–PD-1–treated mice, reaching up to 20% of total CD8+ TILs (mean 11 ± 5%) and 8% of total TILs (mean 3.8 ± 2.5%) Results (Fig. 2B). As anti–CTLA-4 blockade has also been shown to en- Identification
Details
-
File Typepdf
-
Upload Time-
-
Content LanguagesEnglish
-
Upload UserAnonymous/Not logged-in
-
File Pages10 Page
-
File Size-