scholarly journals 321 PD-L1 checkpoint blockade eradicates residual leukemia in a mouse model of acute lymphoblastic leukemia by countering exhaustion of cytotoxic and effector CD4+ T-cells

2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A345-A346
Author(s):  
Sean Tracy ◽  
Hrishi Venkatesh ◽  
Lynn Heltemes-Harris ◽  
Gregory Hubbard ◽  
Can Hekim ◽  
...  

BackgroundPhenotypic exhaustion of CD4+ T-cells is a strong negative prognostic factor in acute lymphoblastic leukemia (ALL).1–3 Despite this, PD1/PD-L1 immune checkpoint therapy has shown little activity in this disease setting to date. Factors influencing the responsiveness of the T-cell compartment to checkpoint blockade are unknown.MethodsAn established murine model of BCR-ABL+ ALL was used. Leukemia was established by tail vein injection, and mice were treated with the BCR-ABL tyrosine kinase inhibitor nilotinib with or without PD-L1 mAb therapy. scRNAseq/TCRseq was performed using multiple treatment groups.ResultsTreatment of leukemia-bearing mice with a combination of the BCR-ABL tyrosine kinase inhibitor nilotinib and PD-L1 immune checkpoint blockade led to eradication of leukemia in 70% of treated mice (figure 1). Efficacy was dependent on the presence of CD4+ T-cells, while CD8+ T-cells appeared to play a lesser role. Direct cytotoxicity by CD4+ T-cells was confirmed in live cell-killing assays (figure 2). Mice that were treated with PD-L1 blockade and survived to day 100 were found to have no detectable residual leukemia. They were also protected from leukemia rechallenge, suggesting the elicitation of a memory response. scRNAseq analysis revealed that CD44hi CD4+ T-cells were highly heterogeneous, with regulatory, effector, and stem-like TCF7+ precursor subsets present (figures 3–4). A unique population of CD4+ T-cells was elicited by live leukemia challenge (clusters 6 and 7 in figure 3) but not by vaccination with heat-killed leukemia cells. This subset was characterized by relatively low levels of expression of TCF7, but high levels of expression of Granzyme B, TOX, the effector cytokines IFNγ and TNFα, the inhibitory receptors PD1, TIM3, and LAG3, and the chemokine CCL5 (figure 5). PD-L1 checkpoint blockade was associated with early narrowing of the clonality of this population (figure 6), decreased markers of exhaustion, and more robust synthesis of TNFα.Abstract 321 Figure 1Survival analysis. BCR-ABL+ ALL was established by tail vein injection on day 0. Nilotinib (75 mg/kg) was administered via oral gavage. mAbs targeting PD-L1 with or without depleting antibodies towards CD4 or CD8 were administered via intraperitoneal injection. p-value derived by log-rank analysisAbstract 321 Figure 2Analysis of the increase in the number of dead cells (y-axis) over time (x-axis) from a live killing assay (Incucyte) using splenic CD4+ or CD8+ T-cells from experimental arms as treated in figure 3. Control traces from separate wells with LM138 target cells only are included. Experiments were done using Cytotox NIRAbstract 321 Figure 3(Left) Experimental approach. 5 groups (n=4 mice/group) were treated in parallel with the indicated conditions. CD44hi CD4+ T-cells from the spleen and bone marrow of mice in each group were labelled with oligo-conjugated hashtag antibodies (Biolegend) and CITE-SEQ antibodies towards PD1, TIM3, LAG3, CD25, and TIGIT, prior to FACs-sorting. scRNAseq/TCRseq analysis (10x Genomics) was performed on 5,349 individual cells after multiplet removal. (Right) UMAP plots of all cells combined. Clusters were identified by differential expression of canonical gene productsAbstract 321 Figure 4Feature plots demonstrating expression of canonical gene products projected onto the UMAP plot in figure 3. Antibody derived tags (ADTs; bottom row) indicate expression level of surface proteins profiled using CITESEQ antibodiesAbstract 321 Figure 5Heatmap of select gene product expression levels in exhausted (cluster 6) CD4+ T-cells across treatment conditionsAbstract 321 Figure 6Simpsons diversity index of the TCR repertoire across treatment arms. Lower values indicate relatively decreased clonalityConclusionsPDL1 immune checkpoint blockade is effective at eradicating residual disease in preclinical models of BCR-ABL+ ALL. ALL elicits a unique CD4+ memory/effector subset characterized by the potential for both chemotactic and cytotoxic functions. Leukemia induces early exhaustion of this subset, which is countered by PDL1 blockade. Efforts to extend these observations to human specimens are underway and will be reported.ReferencesHohtari H, Brück O, Blom S, et al. Immune cell constitution in bone marrow microenvironment predicts outcome in adult ALL. Leukemia 2019;33(7):1570–1582. Blaeschke F, Willier S, Stenger D, et al. Leukemia-induced dysfunctional TIM-3. Leukemia 2020;34(10):2607–2620.Liu L, Chang YJ, Xu LP, et al. T cell exhaustion characterized by compromised MHC class I and II restricted cytotoxic activity associates with acute B lymphoblastic leukemia relapse after allogeneic hematopoietic stem cell transplantation. Clin Immunol 2018;190:32–40.

JCI Insight ◽  
2018 ◽  
Vol 3 (23) ◽  
Author(s):  
Yuji Sato ◽  
Jennifer K. Bolzenius ◽  
Abdallah M. Eteleeb ◽  
Xinming Su ◽  
Christopher A. Maher ◽  
...  

2021 ◽  
Vol 14 (9) ◽  
pp. 101170
Author(s):  
Vera Bauer ◽  
Fatima Ahmetlić ◽  
Nadine Hömberg ◽  
Albert Geishauser ◽  
Martin Röcken ◽  
...  

2020 ◽  
Vol 22 (Supplement_2) ◽  
pp. ii108-ii108
Author(s):  
Jayeeta Ghose ◽  
Baisakhi Raychaudhuri ◽  
Kevin Liu ◽  
William Jiang ◽  
Pooja Gulati ◽  
...  

Abstract BACKGROUND Glioblastoma (GBM) is associated with systemic and intratumoral immunosuppression. Part of this immunosuppression is mediated by myeloid derived suppressor cells (MDSCs). Preclinical evidence shows that ibrutinib, a tyrosine kinase inhibitor FDA approved for use in chronic lymphocytic leukemia and known to be CNS penetrant, can decrease MDSC generation and function. Also, focal radiation therapy (RT) synergizes with anti-PD-1 therapy in mouse GBM models. Thus, we aimed to test the combination of these approaches on immune activation and survival in a preclinical immune-intact GBM mouse model. METHODS C57BL/6 mice intracranially implanted with the murine glioma cell line GL261-Luc2 were divided into 8 groups consisting of treatments with ibrutinib, RT (10 Gy SRS), or anti-PD-1 individually or in each combination (along with a no treatment control group). Immune cell subset changes (flow-cytometry) and animal survival (Kaplan-Meier) were assessed (n=10 mice per group). RESULTS Median survival of the following groups including control (28 days), ibrutinib (27 days), RT (30 days) or anti-PD-1 (32 days) showed no significant differences. However, a significant improvement in median survival was seen in mice given combinations of ibrutinib+RT (35 days), ibrutinib+anti-PD-1 (38 days), and triple therapy with ibrutinib+RT+anti-PD-1 (48 days, p < 0.05) compared to controls or single treatment groups. The reproducible survival benefit of triple combination therapy was abrogated in the setting of CD4+ and CD8+ T cell depletion. Contralateral intracranial tumor re-challenge in long-term surviving mice suggested generation of tumor-specific immune memory responses. The immune profile of the tumor microenvironment (TME) showed increased cytotoxic CD8+ T cells and decreased MDSCs and regulatory T cells in the triple combination therapy mice compared to controls. CONCLUSION The combination of ibrutinib, focal RT, and anti-PD-1 immune checkpoint blockade led to a significant survival benefit compared to controls in a preclinical model of GBM.


Cancer Cell ◽  
2018 ◽  
Vol 34 (4) ◽  
pp. 691 ◽  
Author(s):  
Roberta Zappasodi ◽  
Sadna Budhu ◽  
Matthew D. Hellmann ◽  
Michael A. Postow ◽  
Yasin Senbabaoglu ◽  
...  

2020 ◽  
Vol 8 (Suppl 2) ◽  
pp. A5.1-A5
Author(s):  
A Martinez-Usatorre ◽  
E Kadioglu ◽  
C Cianciaruso ◽  
B Torchia ◽  
J Faget ◽  
...  

BackgroundImmune checkpoint blockade (ICB) with antibodies against PD-1 or PD-L1 may provide therapeutic benefits in patients with non-small cell lung cancer (NSCLC). However, most tumours are resistant and cases of disease hyper-progression have also been reported.Materials and MethodsGenetically engineered mouse models of KrasG12Dp53null NSCLC were treated with cisplatin along with antibodies against angiopoietin-2/VEGFA, PD-1 and CSF1R. Tumour growth was monitored by micro-computed tomography and the tumour vasculature and immune cell infiltrates were assessed by immunofluorescence staining and flow cytometry.ResultsCombined angiopoietin-2/VEGFA blockade by a bispecific antibody (A2V) modulated the vasculature and abated immunosuppressive macrophages while increasing CD8+effector T cells in the tumours, achieving disease stabilization comparable or superior to cisplatin-based chemotherapy. However, these immunological responses were unexpectedly limited by the addition of a PD-1 antibody, which paradoxically enhanced progression of a fraction of the tumours through a mechanism involving regulatory T cells and macrophages. Elimination of tumour-associated macrophages with a CSF1R-blocking antibody induced NSCLC regression in combination with PD-1 blockade and cisplatin.ConclusionsThe immune cell composition of the tumour determines the outcome of PD-1 blockade. In NSCLC, high infiltration of regulatory T cells and immunosuppressive macrophages may account for tumour hyper-progression upon ICB.Disclosure InformationA. Martinez-Usatorre: None. E. Kadioglu: None. C. Cianciaruso: None. B. Torchia: None. J. Faget: None. E. Meylan: None. M. Schmittnaegel: None. I. Keklikoglou: None. M. De Palma: None.


Cancers ◽  
2020 ◽  
Vol 12 (10) ◽  
pp. 2762 ◽  
Author(s):  
Xinrui Zhao ◽  
Chunlin Shao

Radiotherapy (RT) is a conventional method for clinical treatment of local tumors, which can induce tumor-specific immune response and cause the shrinkage of primary tumor and distal metastases via mediating tumor infiltration of CD8+ T cells. Ionizing radiation (IR) induced tumor regression outside the radiation field is termed as abscopal effect. However, due to the mobilization of immunosuppressive signals by IR, the activated CD8+T cells are not sufficient to maintain a long-term positive feedback to make the tumors regress completely. Eventually, the “hot” tumors gradually turn to “cold”. With the advent of emerging immunotherapy, the combination of immune checkpoint blockade (ICB) and local RT has produced welcome changes in stubborn metastases, especially anti-PD-1/PD-L1 and anti-CTLA-4 which have been approved in clinical cancer treatment. However, the detailed mechanism of the abscopal effect induced by combined therapy is still unclear. Therefore, how to formulate a therapeutic schedule to maximize the efficacy should be took into consideration according to specific circumstance. This paper reviewed the recent research progresses in immunomodulatory effects of local radiotherapy on the tumor microenvironment, as well as the unique advantage for abscopal effect when combined with ICB, with a view to exploring the potential application value of radioimmunotherapy in clinic.


2020 ◽  
Vol 3 (1) ◽  
Author(s):  
Asiel A. Benitez ◽  
Sara Khalil-Agüero ◽  
Anjali Nandakumar ◽  
Namita T. Gupta ◽  
Wen Zhang ◽  
...  

AbstractThe endogenous anti-tumor responses are limited in part by the absence of tumor-reactive T cells, an inevitable consequence of thymic central tolerance mechanisms ensuring prevention of autoimmunity. Here we show that tumor rejection induced by immune checkpoint blockade is significantly enhanced in Aire-deficient mice, the epitome of central tolerance breakdown. The observed synergy in tumor rejection extended to different tumor models, was accompanied by increased numbers of activated T cells expressing high levels of Gzma, Gzmb, Perforin, Cxcr3, and increased intratumoural levels of Cxcl9 and Cxcl10 compared to wild-type mice. Consistent with Aire’s central role in T cell repertoire selection, single cell TCR sequencing unveiled expansion of several clones with high tumor reactivity. The data suggest that breakdown in central tolerance synergizes with immune checkpoint blockade in enhancing anti-tumor immunity and may serve as a model to unmask novel anti-tumor therapies including anti-tumor TCRs, normally purged during central tolerance.


Sign in / Sign up

Export Citation Format

Share Document