scholarly journals Motility Dynamics of T Cells in Tumor-Draining Lymph Nodes: A Rational Indicator of Antitumor Response and Immune Checkpoint Blockade

Cancers ◽  
2021 ◽  
Vol 13 (18) ◽  
pp. 4616
Author(s):  
Yasuhiro Kanda ◽  
Taku Okazaki ◽  
Tomoya Katakai

The migration status of T cells within the densely packed tissue environment of lymph nodes reflects the ongoing activation state of adaptive immune responses. Upon encountering antigen-presenting dendritic cells, actively migrating T cells that are specific to cognate antigens slow down and are eventually arrested on dendritic cells to form immunological synapses. This dynamic transition of T cell motility is a fundamental strategy for the efficient scanning of antigens, followed by obtaining the adequate activation signals. After receiving antigenic stimuli, T cells begin to proliferate, and the expression of immunoregulatory receptors (such as CTLA-4 and PD-1) is induced on their surface. Recent findings have revealed that these ‘immune checkpoint’ molecules control the activation as well as motility of T cells in various situations. Therefore, the outcome of tumor immunotherapy using checkpoint inhibitors is assumed to be closely related to the alteration of T cell motility, particularly in tumor-draining lymph nodes (TDLNs). In this review, we discuss the migration dynamics of T cells during their activation in TDLNs, and the roles of checkpoint molecules in T cell motility, to provide some insight into the effect of tumor immunotherapy via checkpoint blockade, in terms of T cell dynamics and the importance of TDLNs.

2019 ◽  
Author(s):  
Xianda Zhao ◽  
Beminet Kassaye ◽  
Dechen Wangmo ◽  
Emil Lou ◽  
Subbaya Subramanian

SUMMARYImmunotherapies are used as adjuvant therapies for cancers. However, knowledge of how traditional cancer treatments affect immunotherapies is limited. Using mouse models, we demonstrate that tumor-draining lymph nodes (TdLNs) are critical for tumor antigen-specific T-cell response. However, removing TdLNs concurrently with established primary tumors did not affect the immune checkpoint blockade (ICB) response on localized secondary tumor due to immunotolerance in TdLNs and distribution of antigen-specific T cells in peripheral lymphatic organs. Notably, treatment response improved with sequential administration of 5-fluorouracil (5-FU) and ICB compared to concurrent administration of ICB with 5-FU. Immune profiling revealed that using 5-FU as induction treatment increased tumor visibility to immune cells, decreased immunosuppressive cells in the tumor microenvironment, and limited chemotherapy-induced T-cell depletion. We show that the effect of traditional cytotoxic treatment, not TdLNs, influences immunotherapy response in localized secondary tumors. We postulate essential considerations for successful immunotherapy strategies in clinical conditions.Graphic abstractThe effects of tumor-draining lymph nodes (TdLNs) resection and a combination of cytotoxic chemotherapy on immune checkpoint blockade therapies are evaluated in this study. TdLNs resection was adverse in eliciting an antitumor immune response in early-stage tumors, but not in late-stage tumors. Further, sequential treatments with cytotoxic chemotherapy and immunotherapy showed better tumor control compared to concurrent combinatorial treatments.


Neurosurgery ◽  
2019 ◽  
Vol 66 (Supplement_1) ◽  
Author(s):  
Connor Liu ◽  
Maximilian Schaettler ◽  
Jay Bowman-Kirigin ◽  
Diane Bender ◽  
Dale K Kobayashi ◽  
...  

Abstract INTRODUCTION The treatment of glioblastoma remains a challenge for modern therapy. Checkpoint blockade inhibitors (CBI), designed to block inhibitory T-cell signaling, represent attractive potential therapeutic interventions. However, glioblastoma response rates to CBI remain low. To this end, preclinical models are critical to studying multi-modal immune interventions to overcome CBI resistance. Therefore, we set out to identify endogenous neoantigens in a CBI resistant murine glioblastoma and assess the efficacy of neoantigen vaccination in combination with CBI treatment. METHODS Whole exome DNA and RNA sequencing was used to identify expressed, missense mutations in the C57BL/6 derived murine glioblastoma, CT2A. The pVAC-seq software suite was used to identify candidate neoantigens predicted to bind H2-Kb and H2-Db molecules. CD8 + T cells isolated from CT2A tumor-infiltrating lymphocytes (TIL) were screened for neoantigen reactivity by IFN-gamma enzyme linked immunospot assays. Survival analysis was performed on intracranial tumor bearing mice treated with neoantigen vaccination, anti-PD-L1, or combination therapy. RESULTS In silico analysis identified 649 CT2A-derived candidate neoantigens predicted to bind H2-Kb or H2-Db molecules. Of the 40 top-ranking neoantigen candidates, 16 elicited CD8 + TIL responses in mice vaccinated with cognate peptides. Assessing for endogenous reactivity, we identified neoantigen specific CD8 + T cell responses in the intracranial TIL and draining lymph nodes to two H2-Kb restricted, Epb4 (H471L) and Pomgnt1 (R497L), and one H2-Db restricted neoantigen, Plin2 (G332R). Survival studies showed that therapeutic neoantigen vaccination with Epb4, Pomgnt1, and Plin2, in combination with anti-PD-L1 treatment was superior to anti-PD-L1 alone. CONCLUSION We identified endogenous neoantigen specific CD8 + T cells within brain tumors and draining lymph nodes of a CBI resistant murine glioblastoma. Furthermore, we find that neoantigen vaccination significantly augments survival benefit in combination with anti-PD-L1 treatment. These observations provide important preclinical correlates for glioblastoma immunotherapy trials and support further investigation into the effects of multi-modal immunotherapeutic interventions on anti-glioma immunity.


2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A593-A593
Author(s):  
Ang Cui ◽  
Kelly Moynihan ◽  
Shuqiang Li ◽  
Chensu Wang ◽  
Jackson Southard ◽  
...  

BackgroundA major goal in cancer immunology is to rationally design combination therapies that lead to a higher response rate, especially for poorly immunogenic tumors that do not respond to immune checkpoint blockade therapy alone. We previously developed a combination therapeutic strategy, termed AIPV, consisting of a tumor-targeting antibody, a recombinant interleukin-2 with an extended half-life, an anti-PD-1 antibody, and a T cell vaccine [1]. The full AIPV therapy can eradicate large, aggressive, poorly immunogenic tumors in multiple mouse tumor models. However, the exact cellular and molecular pathways involved in such an effective response remain poorly understood.MethodsIn this study, we used single-cell RNA-sequencing to define the detailed cellular and molecular changes in tumors and tumor-draining lymph nodes following the full AIPV therapy or a less effective sub-combination therapy in mice with poorly immunogenic B16F10 tumors.ResultsUsing our approach, we were able to uncover T cells, NK cells, neutrophils, macrophages/monocytes, classical dendritic cells, and plasmacytoid dendritic cells in tumors. We observed profound remodeling of every immune cell type following the effective therapeutic treatment. In particular, we found that classical dendritic cells take up tumor antigens, become activated, and migrate to draining lymph nodes following the AIPV therapy, but not following the less effective IPV therapy. We characterized the transcriptomic changes of these dendritic cells and found that they over-express molecules involved in antigen uptake.ConclusionsOur study comprehensively characterized a system that can overcome resistance to immune checkpoint blockade therapy, paving a cellular and molecular roadmap for immune-based therapeutic strategies that offer clinical benefits for poorly immunogenic tumors.ReferencesMoynihan KD, Opel CF, Szeto GL, Tzeng A, Zhu EF, Engreitz JM, et al. Eradication of large established tumors in mice by combination immunotherapy that engages innate and adaptive immune responses. Nat Med. 2016;22: 1402–1410.Ethics ApprovalAll mouse experiments were reviewed and approved by the Koch Institute and Broad Institute Animal Care and Use Committee (IACUC) (ID 0222-08-18).


Blood ◽  
2006 ◽  
Vol 108 (7) ◽  
pp. 2257-2264 ◽  
Author(s):  
Sophie Laffont ◽  
Jérôme D. Coudert ◽  
Lucile Garidou ◽  
Laurent Delpy ◽  
Aurélie Wiedemann ◽  
...  

Abstract Accumulating evidence indicates that, in absence of CD8+ T-cell activation, CD4+ T-cell–mediated allograft rejection is associated with a dominant Th2-cell response and eosinophil infiltrates. In this study, we analyzed the mechanisms by which CD8+ T cells regulate alloreactive CD4+ T-cell priming and differentiation into interleukin 4 (IL-4)–producing cells. We showed that interferon γ (IFN-γ) production by CD8+ T cells was dispensable for the inhibition of Th2-cell development, as well as tissue eosinophilia and type 2 cytokine production in the rejected grafts. Since we noticed that CD8+ T cells not only suppressed Th2 differentiation, but also down-modulated the overall priming of alloreactive CD4+ T cells, we evaluated whether CD8+ T cells act by limiting the accumulation of donor-derived dendritic cells (DCs) in lymph nodes. We found that indeed, alloreactive CD8+ T cells rapidly eliminated allogeneic DCs from T-cell areas of draining lymph nodes, through a perforin-dependent mechanism. Thus, our data demonstrate that cytotoxic T lymphocyte (CTL)–mediated clearance of allogeneic DCs is a negative feedback mechanism that limits the duration of alloantigen presentation in draining lymph nodes, thereby modulating the amplitude and polarization of the primary alloreactive CD4+ T-cell responses.


2020 ◽  
Vol 11 (1) ◽  
Author(s):  
Qi Peng ◽  
Xiangyan Qiu ◽  
Zihan Zhang ◽  
Silin Zhang ◽  
Yuanyuan Zhang ◽  
...  

Abstract Immune checkpoint blockade therapies have shown clinical promise in a variety of cancers, but how tumor-infiltrating T cells are activated remains unclear. In this study, we explore the functions of PD-L1 on dendritic cells (DCs), which highly express PD-L1. We observe that PD-L1 on DC plays a critical role in limiting T cell responses. Type 1 conventional DCs are essential for PD-L1 blockade and they upregulate PD-L1 upon antigen uptake. Upregulation of PD-L1 on DC is mediated by type II interferon. While DCs are the major antigen presenting cells for cross-presenting tumor antigens to T cells, subsequent PD-L1 upregulation protects them from killing by cytotoxic T lymphocytes, yet dampens the antitumor responses. Blocking PD-L1 in established tumors promotes re-activation of tumor-infiltrating T cells for tumor control. Our study identifies a critical and dynamic role of PD-L1 on DC, which needs to be harnessed for better invigoration of antitumor immune responses.


1999 ◽  
Vol 189 (1) ◽  
pp. 169-178 ◽  
Author(s):  
Omid Akbari ◽  
Naveed Panjwani ◽  
Sylvie Garcia ◽  
Ricardo Tascon ◽  
Doug Lowrie ◽  
...  

The mechanisms underlying initiation and maintenance of CD4 T cell responses after DNA vaccination were studied using a construct coding for nonsecreted fifth component of complement (C5) protein, thus restricting the availability of antigen. The only cell types to express C5 were keratinocytes at the site of DNA application and a small number of dendritic cells present in the draining lymph nodes. Antigen expression persisted for up to 12 wk in keratinocytes, but dendritic cells did not express C5 beyond 2 wk after vaccination. Cross-priming of dendritic cells by C5 expressed in keratinocytes did not occur unless keratinocyte death was induced by irradiation in vitro. CD4 T cells were activated in the draining lymph nodes only and subsequently migrated to the spleen, where memory T cells persisted for longer than 40 wk despite the absence of a source of persistent antigen. While DNA vaccination resulted in transfection of a small proportion of dendritic cells only, it led to general activation of all dendritic cells, thus providing optimal conditions for effective T cell activation and maintenance of memory.


Cancers ◽  
2021 ◽  
Vol 13 (19) ◽  
pp. 5021
Author(s):  
Dechen Wangmo ◽  
Prem K. Premsrirut ◽  
Ce Yuan ◽  
William S. Morris ◽  
Xianda Zhao ◽  
...  

Colorectal cancer (CRC) is one of the most common malignancies in both morbidity and mortality. Immune checkpoint blockade (ICB) treatments have been successful in a portion of mismatch repair-deficient (dMMR) CRC patients but have failed in mismatch repair-proficient (pMMR) CRC patients. Atypical Chemokine Receptor 4 (ACKR4) is implicated in regulating dendritic cell (DC) migration. However, the roles of ACKR4 in CRC development and anti-tumor immunoregulation are not known. By analyzing human CRC tissues, transgenic animals, and genetically modified CRC cells lines, our study revealed an important function of ACKR4 in maintaining CRC immune response. Loss of ACKR4 in CRC is associated with poor immune infiltration in the tumor microenvironment. More importantly, loss of ACKR4 in CRC tumor cells, rather than stromal cells, restrains the DC migration and antigen presentation to the tumor-draining lymph nodes (TdLNs). Moreover, tumors with ACKR4 knockdown become less sensitive to immune checkpoint blockade. Finally, we identified that microRNA miR-552 negatively regulates ACKR4 expression in human CRC. Taken together, our studies identified a novel and crucial mechanism for the maintenance of the DC-mediated T-cell priming in the TdLNs. These new findings demonstrate a novel mechanism leading to immunosuppression and ICB treatment resistance in CRC.


2021 ◽  
Vol 9 (1) ◽  
pp. e001460 ◽  
Author(s):  
Xiuting Liu ◽  
Graham D Hogg ◽  
David G DeNardo

The clinical success of immune checkpoint inhibitors has highlighted the central role of the immune system in cancer control. Immune checkpoint inhibitors can reinvigorate anti-cancer immunity and are now the standard of care in a number of malignancies. However, research on immune checkpoint blockade has largely been framed with the central dogma that checkpoint therapies intrinsically target the T cell, triggering the tumoricidal potential of the adaptive immune system. Although T cells undoubtedly remain a critical piece of the story, mounting evidence, reviewed herein, indicates that much of the efficacy of checkpoint therapies may be attributable to the innate immune system. Emerging research suggests that T cell-directed checkpoint antibodies such as anti-programmed cell death protein-1 (PD-1) or programmed death-ligand-1 (PD-L1) can impact innate immunity by both direct and indirect pathways, which may ultimately shape clinical efficacy. However, the mechanisms and impacts of these activities have yet to be fully elucidated, and checkpoint therapies have potentially beneficial and detrimental effects on innate antitumor immunity. Further research into the role of innate subsets during checkpoint blockade may be critical for developing combination therapies to help overcome checkpoint resistance. The potential of checkpoint therapies to amplify innate antitumor immunity represents a promising new field that can be translated into innovative immunotherapies for patients fighting refractory malignancies.


2012 ◽  
Vol 278 (1-2) ◽  
pp. 158-165 ◽  
Author(s):  
Tamás Kobezda ◽  
Sheida Ghassemi-Nejad ◽  
Tibor T. Glant ◽  
Katalin Mikecz

Sign in / Sign up

Export Citation Format

Share Document