Faculty Opinions recommendation of Neoantigen-reactive CD8+ T cells affect clinical outcome of adoptive transfer with tumor-infiltrating lymphocytes in melanoma.

Author(s):  
Paul R Walker
Author(s):  
Nikolaj Pagh Kristensen ◽  
Christina Heeke ◽  
Siri A. Tvingsholm ◽  
Annie Borch ◽  
Arianna Draghi ◽  
...  

2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A822-A822
Author(s):  
Sri Krishna ◽  
Frank Lowery ◽  
Amy Copeland ◽  
Stephanie Goff ◽  
Grégoire Altan-Bonnet ◽  
...  

BackgroundAdoptive T cell therapy (ACT) utilizing ex vivo-expanded autologous tumor infiltrating lymphocytes (TILs) can result in complete regression of human cancers.1 Successful immunotherapy is influenced by several tumor-intrinsic factors.2 3 Recently, T cell-intrinsic factors have been associated with immunotherapy response in murine and human studies.4 5 Analyses of tumor-reactive TILs have concluded that anti-tumor neoantigen-specific TILs are enriched in subsets defined by the expression of PD-1 or CD39.6 7 Thus, there is a lack of consensus regarding the tumor-reactive TIL subset that is directly responsible for successful immunotherapies such as ICB and ACT. In this study, we attempted to define the fitness landscape of TIL-enriched infusion products to specifically understand its phenotypic impact on human immunotherapy responses.MethodsWe compared the phenotypic differences that could distinguish bulk ACT infusion products (I.P.) administered to patients who had complete response to therapy (complete responders, CRs, N = 24) from those whose disease progressed following ACT (non-responders, NRs, N = 30) by high dimensional single cell protein and RNA analysis of the I.P. We further analyzed the phenotypic states of anti-tumor neoantigen specific TILs from patient I.P (N = 26) by flow cytometry and single cell transcriptomics.ResultsWe identified two CD8+ TIL populations associated with clinical outcomes: a memory-progenitor CD39-negative stem-like TIL (CD39-CD69-) in the I.P. associated with complete cancer regression (overall survival, P < 0.0001, HR = 0.217, 95% CI 0.101 to 0.463) and TIL persistence, and a terminally differentiated CD39-positive TIL (CD39+CD69+) population associated with poor TIL persistence post-treatment. Although the majority (>65%) of neoantigen-reactive TILs in both responders and non-responders to ACT were found in the differentiated CD39+ state, CR infusion products also contained a pool of CD39- stem-like neoantigen-specific TILs (median = 8.8%) that was lacking in NR infusion products (median = 23.6%, P = 1.86 x 10-5). Tumor-reactive stem-like T cells were capable of self-renewal, expansion, and persistence, and mediated superior anti-tumor response in vivo.ConclusionsOur results support the hypothesis that responders to ACT received infusion products containing a pool of stem-like neoantigen-specific TILs that are able to undergo prolific expansion, give rise to differentiated subsets, and mediate long-term tumor control and T cell persistence, in line with recent murine ICB studies mediated by TCF+ progenitor T cells.4 5 Our data also suggest that TIL subsets mediating ACT-response (stem-like CD39-) might be distinct from TIL subsets enriched for anti-tumor-reactivity (terminally differentiated CD39+) in human TIL.6 7AcknowledgementsWe thank Don White for curating the melanoma patient cohort, and J. Panopoulos (Flowjo) for helpful discussions on high-dimensional analysis, and NCI Surgery Branch members for helpful insights and suggestions. S. Krishna acknowledges funding support from NCI Director’s Innovation Award from the National Cancer Institute.Trial RegistrationNAEthics ApprovalThe study was approved by NCI’s IRB ethics board.ReferencesGoff SL, et al. Randomized, prospective evaluation comparing intensity of lymphodepletion before adoptive transfer of tumor-infiltrating lymphocytes for patients with metastatic melanoma. J Clin Oncol 2016;34:2389–2397.Snyder A, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med 2014;371:2189–2199.McGranahan N, et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 2016;351:1463–1469.Sade-Feldman M, et al. Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell 2019;176:404.Miller BC, et al. Subsets of exhausted CD8 T cells differentially mediate tumor control and respond to checkpoint blockade. Nat. Immunol 2019;20:326–336.Simoni Y, et al. Bystander CD8 T cells are abundant and phenotypically distinct in human tumour infiltrates. Nature 2018;557:575–579.Gros A, et al. PD-1 identifies the patient-specific CD8+ tumor-reactive repertoire infiltrating human tumors. J Clin Invest 2014;124:2246–2259.


2010 ◽  
Vol 207 (8) ◽  
pp. 1791-1804 ◽  
Author(s):  
Elizabeth D. Thompson ◽  
Hilda L. Enriquez ◽  
Yang-Xin Fu ◽  
Victor H. Engelhard

Studies of T cell responses to tumors have focused on the draining lymph node (LN) as the site of activation. We examined the tumor mass as a potential site of activation after adoptive transfer of naive tumor-specific CD8 T cells. Activated CD8 T cells were present in tumors within 24 h of adoptive transfer and proliferation of these cells was also evident 4–5 d later in mice treated with FTY720 to prevent infiltration of cells activated in LNs. To confirm that activation of these T cells occurred in the tumor and not the tumor-draining LNs, we used mice lacking LNs. Activated and proliferating tumor-infiltrating lymphocytes were evident in these mice 24 h and 4 d after naive cell transfer. T cells activated within tumors acquired effector function that was evident both ex vivo and in vivo. Both cross-presenting antigen presenting cells within the tumor and tumor cells directly presenting antigen activated these functional CD8 effectors. We conclude that tumors support the infiltration, activation, and effector differentiation of naive CD8 T cells, despite the presence of immunosuppressive mechanisms. Thus, targeting of T cell activation to tumors may present a tool in the development of cancer immunotherapy.


2019 ◽  
Vol 4 (31) ◽  
pp. eaap9520 ◽  
Author(s):  
Lelisa F. Gemta ◽  
Peter J. Siska ◽  
Marin E. Nelson ◽  
Xia Gao ◽  
Xiaojing Liu ◽  
...  

In the context of solid tumors, there is a positive correlation between the accumulation of cytotoxic CD8+tumor-infiltrating lymphocytes (TILs) and favorable clinical outcomes. However, CD8+TILs often exhibit a state of functional exhaustion, limiting their activity, and the underlying molecular basis of this dysfunction is not fully understood. Here, we show that TILs found in human and murine CD8+melanomas are metabolically compromised with deficits in both glycolytic and oxidative metabolism. Although several studies have shown that tumors can outcompete T cells for glucose, thus limiting T cell metabolic activity, we report that a down-regulation in the activity of ENOLASE 1, a critical enzyme in the glycolytic pathway, represses glycolytic activity in CD8+TILs. Provision of pyruvate, a downstream product of ENOLASE 1, bypasses this inactivity and promotes both glycolysis and oxidative phosphorylation, resulting in improved effector function of CD8+TILs. We found high expression of both enolase 1 mRNA and protein in CD8+TILs, indicating that the enzymatic activity of ENOLASE 1 is regulated posttranslationally. These studies provide a critical insight into the biochemical basis of CD8+TIL dysfunction.


2020 ◽  
Vol 12 (564) ◽  
pp. eabb2311 ◽  
Author(s):  
Tobias Weiss ◽  
Emanuele Puca ◽  
Manuela Silginer ◽  
Teresa Hemmerle ◽  
Shila Pazahr ◽  
...  

Glioblastoma is a poorly immunogenic cancer, and the successes with recent immunotherapies in extracranial malignancies have, so far, not been translated to this devastating disease. Therefore, there is an urgent need for new strategies to convert the immunologically cold glioma microenvironment into a hot one to enable effective antitumor immunity. Using the L19 antibody, which is specific to a tumor-associated epitope of extracellular fibronectin, we developed antibody-cytokine fusions—immunocytokines—with interleukin-2 (IL2), IL12, or tumor necrosis factor (TNF). We showed that L19 accumulated in the tumor microenvironment of two orthotopic immunocompetent mouse glioma models. Furthermore, intravenous administration of L19-mIL12 or L19-mTNF cured a proportion of tumor-bearing mice, whereas L19-IL2 did not. This therapeutic activity was abolished in RAG−/− mice or upon depletion of CD4 or CD8 T cells, suggesting adaptive immunity. Mechanistically, both immunocytokines promoted tumor-infiltrating lymphocytes and increased the amounts of proinflammatory cytokines within the tumor microenvironment. In addition, L19-mTNF induced tumor necrosis. Systemic administration of the fully human L19-TNF fusion protein to patients with glioblastoma (NCT03779230) was safe, decreased regional blood perfusion within the tumor, and was associated with increasing tumor necrosis and an increase in tumor-infiltrating CD4 and CD8 T cells. The extensive preclinical characterization and subsequent clinical translation provide a robust basis for future studies with immunocytokines to treat malignant brain tumors.


2021 ◽  
Vol 218 (9) ◽  
Author(s):  
Courtney Mowat ◽  
Shayla R. Mosley ◽  
Afshin Namdar ◽  
Daniel Schiller ◽  
Kristi Baker

Colorectal cancers (CRCs) deficient in DNA mismatch repair (dMMR) contain abundant CD8+ tumor-infiltrating lymphocytes (TILs) responding to the abundant neoantigens from their unstable genomes. Priming of such tumor-targeted TILs first requires recruitment of CD8+ T cells into the tumors, implying that this is an essential prerequisite of successful dMMR anti-tumor immunity. We have discovered that selective recruitment and activation of systemic CD8+ T cells into dMMR CRCs strictly depend on overexpression of CCL5 and CXCL10 due to endogenous activation of cGAS/STING and type I IFN signaling by damaged DNA. TIL infiltration into orthotopic dMMR CRCs is neoantigen-independent and followed by induction of a resident memory-like phenotype key to the anti-tumor response. CCL5 and CXCL10 could be up-regulated by common chemotherapies in all CRCs, indicating that facilitating CD8+ T cell recruitment underlies their efficacy. Induction of CCL5 and CXCL10 thus represents a tractable therapeutic strategy to induce TIL recruitment into CRCs, where local priming can be maximized even in neoantigen-poor CRCs.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 860-860 ◽  
Author(s):  
Minjung Lee ◽  
Hongxiang Zeng ◽  
Jia Li ◽  
Wei Han ◽  
Deqiang Sun ◽  
...  

Abstract Background: T cell exhaustion is a dysfunctional state of T cell that occurs during many chronic infections and cancer [1,2]. T cell exhaustion is generally defined by poor effector function, continuous expression of inhibitory receptors and a distinctive transcriptional state when compared with functional effector T cells [2]. Exhaustion prevents optimal control of infection and tumors. Recently, a clearer picture of the functional and phenotypic profiles of exhausted T cells has emerged and T cell exhaustion has been defined in many experimental and clinical settings. Although the involved pathways remain to be fully defined, advances in the molecular delineation of T cell exhaustion are clarifying the underlying causes of this state of differentiation and also suggest promising therapeutic opportunities. A recent study reported disruption of TET2 to promote the therapeutic efficacy of CD19 targeted T cells during cancer immunotherapy [3]. Furthermore, Tet2 deficient macrophages could alter the tumor microenvironment to reduce tumor burden during melanoma progression [4]. Together, these data suggest, contrary to the tacit belief of Tet2 as a tumor suppressor, deletion of Tet2 in specific subsets of immune cells might enhance anti-tumor immunity to benefit cancer therapy. In this study, we set out to explore the role of Tet2 in CD8+ tumor infiltrating lymphocytes (TIL) during melanoma progression. Methods: We intradermally injected B16-OVA mouse melanoma cell lines in B6.SJL-Ptprca Pepcb/BoyJ (CD45.1) mice, and use this as an in vivo model to monitor melanoma progression [5]. In parallel, we injected WT-OTI and Tet2KO-OTI CD8+ T cells into CD45.1 mice injected with B16-OVA cells. Melanoma progression was monitored by measuring the tumor sizes for two weeks. At the end point, spleen and tumor infiltrated CD45.2+CD8+ cells were collected and analyzed. RNA-seq, ATAC-seq and CMS-IP-seq experiments were carried out to examine genome-wide gene expression, chromatin accessibility and DNA hydroxymethylation, with the goal of unveiling the underlying molecular mechanisms. Results: Compared with the control mice injected with WT-OTI CD8+ T cells, we observed a strong delay of melanoma disease progression and up to 80% reduction in tumor sizes in mice injected with Tet2KO-OTI CD8+ T cells. Flow cytometry analysis showed no significant changes in CD8+ T cell populations in major lymphoid organs. However, we detected a pronounced reduction of T cell exhaustion in Tet2KO CD8+ TILs compared with the WT group. Further transcriptome and integrative epigenome analysis revealed that Tet2 deleted TILs showed augmented activation of immune related pathways and reduction of the expression of immunosuppressive genes. Conclusion: Our novel findings demonstrated the therapeutic potential of Tet2 inactivation in immune cells during cancer immunotherapy. In our study, we observed that Tet2 depleted CD8+ TILs displayed increased anti-tumor efficiency in a mouse model of melanoma. Tet2 deletion could effectively alleviate T cell exhaustion to boost CD8+ TIL function. Nonetheless, since Tet2 deficiency is closely associated with various hematology disorders [6,7]; cautions must be taken to balance the tumor promoting and immune-boosting properties of Tet2 during cancer therapy. A temporally controllable system to inactivate Tet2 in specific immune cells might be most desirable for pursuing future therapeutic intervention by targeting Tet2. References 1. Thommen, D. S. & Schumacher, T. N. (2018). Cancer Cell33, 547-562. 2. Wherry, E. J. (2011). Nat Immunol12, 492-499. 3. Fraietta, J. A., Nobles, C. L., Sammons, M. A.et al. (2018). Nature558, 307-312. 4. Pan, W., Zhu, S., Qu, K.et al. (2017). Immunity47, 284-297 e285. 5. Mognol, G. P., Spreafico, R., Wong, V.et al. (2017). Proc Natl Acad Sci U S A114, E2776-E2785. 6. Couronne, L., Bastard, C. & Bernard, O. A. (2012). N Engl J Med366, 95-96. 7. Delhommeau, F., Dupont, S., Della Valle, V.et al. (2009). N Engl J Med360, 2289-2301. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
Author(s):  
Jing Zhang ◽  
Mustapha Abubakar ◽  
pei Yuan ◽  
Hela Koka ◽  
Lei Guo ◽  
...  

Abstract BackgroundTumor-infiltrating lymphocytes (TILs) have strong prognostic values in triple-negative and HER2-enriched breast cancer, but their prognostic roles in luminal breast cancer (LBC) are less clear. Here, we assessed the overall TILs levels and CD8+ T-cells in relation to the prognosis of LBC patients from China.Methods A total of 596 patients with LBC who were premenopausal and treated with adjuvant endocrine therapy were included. Among them, 160 cases were evaluated for CD8 by immunohistochemical (IHC) staining. Whole-section hematoxilyn and eosin and IHC staining were visually assessed for stromal TILs (sTILs), stromal CD8+ T-cells (sCD8) and intratumoral CD8+ T-cells (iCD8). Multivariable Cox proportional hazards model were used to test the associations between TILs and disease-free survival (DFS) and overall survival (OS) with the adjustment of clinicopathologic characteristics and treatment. ResultsHigh sTILs (≥10%) were associated with high histologic grade (p<0.001), luminal B/HER2- (p<0.001), luminal B/HER2+ subtype (p=0.002), and high Ki67 expression (≥25%; p=0.014). Similar associations were observed for sCD8 but not for iCD8. While sTILs and sCD8 were not associated with either DFS or OS, the presence of iCD8 (≥1%) was associated with better DFS in both univariate (HR=0.51, 95%CI 0.26-0.96, p=0.042) and multivariate (HR=0.48, 95%CI 0.25-0.92, p=0.027) analyses. Similar but less significant associations were found for iCD8 and OS (adjusted HR=0.35, 95%CI 0.11-1.10, p=0.073).Conclusions Among Chinese premenopausal patients with LBC, iCD8 demonstrated suggestive associations with favorable outcomes. In contrast, although sTILs and sCD8 were associated with more aggressive tumor features, they did not appear to be associated with clinical outcomes.


Sign in / Sign up

Export Citation Format

Share Document