scholarly journals Tumor-derived exosomes promote carcinogenesis of murine oral squamous cell carcinoma

2019 ◽  
Vol 41 (5) ◽  
pp. 625-633 ◽  
Author(s):  
Beatrice M Razzo ◽  
Nils Ludwig ◽  
Chang-Sook Hong ◽  
Priyanka Sharma ◽  
Kellsye P Fabian ◽  
...  

Abstract Circulating tumor-derived exosomes (TEX) interact with a variety of cells in cancer-bearing hosts, leading to cellular reprogramming which promotes disease progression. To study TEX effects on the development of solid tumors, immunosuppressive exosomes carrying PD-L1 and FasL were isolated from supernatants of murine or human HNSCC cell lines. TEX were delivered (IV) to immunocompetent C57BL/6 mice bearing premalignant oral/esophageal lesions induced by the carcinogen, 4-nitroquinoline 1-oxide (4NQO). Progression of the premalignant oropharyngeal lesions to malignant tumors was monitored. A single TEX injection increased the number of developing tumors (6.2 versus 3.2 in control mice injected with phosphate-buffered saline; P < 0.0002) and overall tumor burden per mouse (P < 0.037). The numbers of CD4+ and CD8+ T lymphocytes infiltrating the developing tumors were coordinately reduced (P < 0.01) in mice injected with SCCVII-derived TEX relative to controls. Notably, TEX isolated from mouse or human tumors had similar effects on tumor development and immune cells. A single IV injection of TEX was sufficient to condition mice harboring premalignant OSCC lesions for accelerated tumor progression in concert with reduced immune cell migration to the tumor.

2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A981-A981
Author(s):  
Tara Muijlwijk ◽  
Naomi Remkes ◽  
Jos Poell ◽  
René Leemans ◽  
Ruud Brakenhoff ◽  
...  

BackgroundHead and neck squamous cell carcinomas (HNSCC) are classified in human papillomavirus (HPV)-positive and HPV-negative tumors. In general, HPV-negative HNSCC are genetically characterized by many chromosomal gains and losses.1 Previously, we and others identified a HPV-negative subgroup with few or absent copy number alterations (CNA-silent), and a more favorable prognosis.2 3 Tumors with low copy number changes have generally been associated with high immune infiltration scores,4 but for CNA-silent versus CNA-high HPV-negative HNSCC such data are lacking.In this study we aim to unravel by functional assays immunological differences between HPV-negative and HPV-positive HNSCC, as well as between CNA-silent and CNA-high HPV-negative HNSCC. We analyzed the immune cell subsets attracted by HNSCC cell lines and by tumor-conditioned supernatants.MethodsEight HNSCC cell lines (3 HPV-positive, 3 HPV-negative CNA-high, 2 HPV-negative CNA-silent) and 24-hour supernatants of thirteen HNSCC biopsies were used to characterize their ability to attract immune cells in a transwell migration system. A chemokine mixture was used as a positive control, while medium alone was used to determine spontaneous migration. Peripheral blood mononuclear cells (PBMCs) of various healthy donors were plated in the upper compartment and after six hours the transwell migration was quantified by flow cytometry.ResultsMost HNSCC cell lines induced migration of monocytes, B cells and CD4+ T-cells up to maximal 12%, whereas CD8+ T-cells and conventional dendritic cells (cDCs) were not attracted, irrespective of the donor. Notably, one HPV-negative CNA-silent cell line induced significantly more migration compared to the negative control and other cell lines. Tumor-conditioned supernatants promoted immune cell migration with no apparent differences between tumor sites or HPV-status. Remarkably, up to 31% of monocytes migrated to these supernatants, 9x more than the chemokine control. Also cDC migration was induced, whereas lymphocytes were not attracted.ConclusionsHNSCC cell lines induced monocyte, B-lymphocyte and CD4+ T-lymphocyte migration, whereas tumor-conditioned supernatants attracted monocytes and cDCs only. No difference in immune cell attraction between HPV-positive and -negative HNSCC was observed. Interestingly, one HPV-negative CNA-silent cell line induced robust immune cell migration. Currently we perform a comprehensive chemokine analysis to explain the observed migration. The noted lack of CD8+ T-cell attraction may explain why current treatments with PD-1 inhibitors are effective in only a minority of HNSCC patients. Our data could provide a means to identify patients who might most likely respond to immune checkpoint blockade and to find clues to improve CD8+ T-cell attraction.ReferencesLeemans CR, Snijders PJF, Brakenhoff RH. The molecular landscape of head and neck cancer. Nat Rev Cancer 2018;18:269–82.Smeets SJ, Brakenhoff RH, Ylstra B, van Wieringen WN, van de Wiel MA, Leemans CR, et al. Genetic classification of oral and oropharyngeal carcinomas identifies subgroups with a different prognosis. Cell Oncol 2009;31:291–300.Cancer Genome Atlas N. Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature 2015;517:576–82.Davoli T, Uno H, Wooten EC, Elledge SJ. Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy. Science 2017;355.Ethics ApprovalWritten informed consent was obtained from all patients from whom fresh tumor biopsies were used for research, as part of the HNcol protocol at the Department of Otolaryngology|Head and Neck Surgery of Amsterdam UMC (VUmc) as approved by the Institutional Review Board (2008.071|A2016.035). Buffy coats, with written consent from the donors, were purchased from the Dutch blood bank (Sanquin) and used to isolate PBMC.


2020 ◽  
Vol 21 (23) ◽  
pp. 8929
Author(s):  
Melanie Kienzl ◽  
Julia Kargl ◽  
Rudolf Schicho

Leukocytes are part of the tumor microenvironment (TME) and are critical determinants of tumor progression. Because of the immunoregulatory properties of cannabinoids, the endocannabinoid system (ECS) may have an important role in shaping the TME. Members of the ECS, an entity that consists of cannabinoid receptors, endocannabinoids and their synthesizing/degrading enzymes, have been associated with both tumor growth and rejection. Immune cells express cannabinoid receptors and produce endocannabinoids, thereby forming an “immune endocannabinoid system”. Although in vitro effects of exogenous cannabinoids on immune cells are well described, the role of the ECS in the TME, and hence in tumor development and immunotherapy, is still elusive. This review/opinion discusses the possibility that the “immune endocannabinoid system” can fundamentally influence tumor progression. The widespread influence of cannabinoids on immune cell functions makes the members of the ECS an interesting target that could support immunotherapy.


Cancers ◽  
2020 ◽  
Vol 12 (3) ◽  
pp. 725
Author(s):  
Yoshihiro Morita ◽  
Macall Leslie ◽  
Hiroyasu Kameyama ◽  
Ganesh L. R. Lokesh ◽  
Norihisa Ichimura ◽  
...  

Chemotherapy is a mainstay of treatment for solid tumors. However, little is known about how therapy-induced immune cell infiltration may affect therapy response. We found substantial CD45+ immune cell density adjacent to E-selectin expressing inflamed vessels in doxorubicin (DOX)-treated residual human breast tumors. While CD45 level was significantly elevated in DOX-treated wildtype mice, it remained unchanged in DOX-treated tumors from E-selectin null mice. Similarly, intravenous administration of anti-E-selectin aptamer (ESTA) resulted in a significant reduction in CD45+ immune cell density in DOX-treated residual tumors, which coincided with a delay in tumor growth and lung metastasis in MMTV-pyMT mice. Additionally, both tumor infiltrating T-lymphocytes and tumor associated-macrophages were skewed towards TH2 in DOX-treated residual breast tumors; however, ESTA suppressed these changes. This study suggests that DOX treatment instigates de novo intratumoral infiltration of immune cells through E-selectin, and functional blockade of E-selectin may reduce residual tumor burden as well as metastasis through suppression of TH2 shift.


Author(s):  
Karan Kohli ◽  
Venu G. Pillarisetty ◽  
Teresa S. Kim

AbstractImmune cell infiltration into solid tumors, their movement within the tumor microenvironment (TME), and interaction with other immune cells are controlled by their directed migration towards gradients of chemokines. Dysregulated chemokine signaling in TME favors the growth of tumors, exclusion of effector immune cells, and abundance of immunosuppressive cells. Key chemokines directing the migration of immune cells into tumor tissue have been identified. In this review, we discuss well-studied chemokine receptors that regulate migration of effector and immunosuppressive immune cells in the context of cancer immunology. We discuss preclinical models that have described the role of respective chemokine receptors in immune cell migration into TME and review preclinical and clinical studies that target chemokine signaling as standalone or combination therapies.


Sports ◽  
2021 ◽  
Vol 9 (4) ◽  
pp. 46
Author(s):  
Polyxeni Spiliopoulou ◽  
Maria Gavriatopoulou ◽  
Efstathios Kastritis ◽  
Meletios Athanasios Dimopoulos ◽  
Gerasimos Terzis

Immunity in the tumor microenvironment plays a central role in tumor development. Cytotoxic immune cells act against tumors, while tumors manage to trigger immunosuppressive mechanisms for defense. One bout of physical exercise acutely regulates the immune system inducing short-term redistribution of immune cells among body organs. Repeated acute immune cell mobilization with continuing exercise training results in long-term adaptations. These long-term exercise-induced changes in the immune system arise both in healthy and in diseased populations, including cancer patients. Recent preclinical studies indicate that physical exercise may have a positive impact on intra-tumoral immune cell processes, resulting in tumor suppression. This short narrative review describes the effect of physical exercise on tumor growth as detected via changes in tumor immunity. Research evidence shows that exercise may improve tumor-suppressive functions and may reduce tumor-progressive responses and mechanisms of immune cells, controlling tumor development. Specifically, it seems that exercise in rodents triggers shifts in tumor infiltration of macrophages, neutrophils, natural killer cells, cytotoxic and regulatory T lymphocytes, resulting in tumor suppression. These recent promising data suggest that physical exercise could be combined with anticancer immunotherapies, although exercise parameters like intensity, duration, and frequency need to be evaluated in more detail. More research is needed to investigate the effect of exercise in other immune cell subtypes and their possible connection with tumor growth, whilst information from human tumors is also required.


2020 ◽  
Vol 0 (0) ◽  
Author(s):  
Anna Helmin-Basa ◽  
Lidia Gackowska ◽  
Sara Balcerowska ◽  
Marcelina Ornawka ◽  
Natalia Naruszewicz ◽  
...  

AbstractInnate immune cells such as natural killer (NK) cells, macrophages and dendritic cells (DCs) are involved in the surveillance and clearance of tumor. Intensive research has exposed the mechanisms of recognition and elimination of tumor cells by these immune cells as well as how cancers evade immune response. Hence, harnessing the immune cells has proven to be an effective therapy in treating a variety of cancers. Strategies aimed to harness and augment effector function of these cells for cancer therapy have been the subject of intense researches over the decades. Different immunotherapeutic possibilities are currently being investigated for anti-tumor activity. Pharmacological agents known to influence immune cell migration and function include therapeutic antibodies, modified antibody molecules, toll-like receptor agonists, nucleic acids, chemokine inhibitors, fusion proteins, immunomodulatory drugs, vaccines, adoptive cell transfer and oncolytic virus–based therapy. In this review, we will focus on the preclinical and clinical applications of NK cell, macrophage and DC immunotherapy in cancer treatment.


Author(s):  
Jason I Griffiths ◽  
Pierre Wallet ◽  
Lance T. Pflieger ◽  
David Stenehjem ◽  
Xuan Liu ◽  
...  

AbstractThe extent that immune cell phenotypes in the peripheral blood reflect within-tumor immune activity prior to and early in cancer therapy is unclear. To address this question, we studied the population dynamics of tumor and immune cells, and immune phenotypic changes, using clinical tumor and immune cell measurements and single cell genomic analyses. These samples were serially obtained from a cohort of advanced gastrointestinal cancer patients enrolled on a trial with chemotherapy and immunotherapy. Using an ecological population model, fitted to clinical tumor burden and immune cell abundance data from each patient, we find evidence of a strong tumor-circulating immune cell interaction in responder patients, but not those patients that progress on treatment. Upon initiation of therapy, immune cell abundance increased rapidly in responsive patients, and once the peak level is reached, tumor burden decreases, similar to models of predator-prey interactions; these dynamic patterns were absent in non-responder patients. To interrogate phenotype dynamics of circulating immune cells, we performed single cell RNA sequencing at serial time points during treatment. These data show that peripheral immune cell phenotypes were linked to the increased strength of patients’ tumor-immune cell interaction, including increased cytotoxic differentiation and strong activation of interferon signaling in peripheral T-cells in responder patients. Joint modeling of clinical and genomic data highlights the interactions between tumor and immune cell populations and reveals how variation in patient responsiveness can be explained by differences in peripheral immune cell signaling and differentiation soon after the initiation of immunotherapy.One sentence summaryPeripheral immune cell differentiation and signaling, upon initiation of immunotherapy, reflects tumor attacking ability and patient response.Significance statementThe evolution of peripheral immune cell abundance and signaling over time, as well as how these immune cells interact with the tumor, may impact a cancer patient’s response to therapy. By developing an ecological population model, we provide evidence of a dynamic predator-prey like relationship between circulating immune cell abundance and tumor size in patients that respond to immunotherapy. This relationship is not found either in patients that are non-responsive to immunotherapy or during chemotherapy. Single cell RNA-sequencing (scRNAseq) of serial peripheral blood samples from patients show that the strength of tumor-immune cell interactions is reflected in T-cells interferon activation and differentiation early in treatment. Thus, circulating immune cell dynamics reflect a tumor’s response to immunotherapy.


2020 ◽  
Vol 117 (27) ◽  
pp. 16072-16082 ◽  
Author(s):  
Jason I. Griffiths ◽  
Pierre Wallet ◽  
Lance T. Pflieger ◽  
David Stenehjem ◽  
Xuan Liu ◽  
...  

The extent to which immune cell phenotypes in the peripheral blood reflect within-tumor immune activity prior to and early in cancer therapy is unclear. To address this question, we studied the population dynamics of tumor and immune cells, and immune phenotypic changes, using clinical tumor and immune cell measurements and single-cell genomic analyses. These samples were serially obtained from a cohort of advanced gastrointestinal cancer patients enrolled in a trial with chemotherapy and immunotherapy. Using an ecological population model, fitted to clinical tumor burden and immune cell abundance data from each patient, we find evidence of a strong tumor-circulating immune cell interaction in responder patients but not in those patients that progress on treatment. Upon initiation of therapy, immune cell abundance increased rapidly in responsive patients, and once the peak level is reached tumor burden decreases, similar to models of predator–prey interactions; these dynamic patterns were absent in nonresponder patients. To interrogate phenotype dynamics of circulating immune cells, we performed single-cell RNA sequencing at serial time points during treatment. These data show that peripheral immune cell phenotypes were linked to the increased strength of patients’ tumor–immune cell interaction, including increased cytotoxic differentiation and strong activation of interferon signaling in peripheral T cells in responder patients. Joint modeling of clinical and genomic data highlights the interactions between tumor and immune cell populations and reveals how variation in patient responsiveness can be explained by differences in peripheral immune cell signaling and differentiation soon after the initiation of immunotherapy.


2020 ◽  
Vol 21 (12) ◽  
pp. 4346 ◽  
Author(s):  
Anthony M. Battram ◽  
Mireia Bachiller ◽  
Beatriz Martín-Antonio

Cellular senescence was first described as a physiological tumor cell suppressor mechanism that leads to cell growth arrest with production of the senescence-associated secretory phenotype known as SASP. The main role of SASP in physiological conditions is to attract immune cells to clear senescent cells avoiding tumor development. However, senescence can be damage-associated and, depending on the nature of these stimuli, additional types of senescence have been described. In the context of cancer, damage-associated senescence has been described as a consequence of chemotherapy treatments that were initially thought of as a tumor suppressor mechanism. However, in certain contexts, senescence after chemotherapy can promote cancer progression, especially when immune cells become senescent and cannot clear senescent tumor cells. Moreover, aging itself leads to continuous inflammaging and immunosenescence which are responsible for rewiring immune cells to become defective in their functionality. Here, we define different types of senescence, pathways that activate them, and functions of SASP in these events. Additionally, we describe the role of senescence in cancer and its treatments, including how aging and chemotherapy contribute to senescence in tumor cells, before focusing on immune cell senescence and its role in cancer. Finally, we discuss potential therapeutic interventions to reverse cell senescence.


2021 ◽  
Vol 12 ◽  
Author(s):  
Olga Zimmermannova ◽  
Inês Caiado ◽  
Alexandra G. Ferreira ◽  
Carlos-Filipe Pereira

Advances in understanding how cancer cells interact with the immune system allowed the development of immunotherapeutic strategies, harnessing patients’ immune system to fight cancer. Dendritic cell-based vaccines are being explored to reactivate anti-tumor adaptive immunity. Immune checkpoint inhibitors and chimeric antigen receptor T-cells (CAR T) were however the main approaches that catapulted the therapeutic success of immunotherapy. Despite their success across a broad range of human cancers, many challenges remain for basic understanding and clinical progress as only a minority of patients benefit from immunotherapy. In addition, cellular immunotherapies face important limitations imposed by the availability and quality of immune cells isolated from donors. Cell fate reprogramming is offering interesting alternatives to meet these challenges. Induced pluripotent stem cell (iPSC) technology not only enables studying immune cell specification but also serves as a platform for the differentiation of a myriad of clinically useful immune cells including T-cells, NK cells, or monocytes at scale. Moreover, the utilization of iPSCs allows introduction of genetic modifications and generation of T/NK cells with enhanced anti-tumor properties. Immune cells, such as macrophages and dendritic cells, can also be generated by direct cellular reprogramming employing lineage-specific master regulators bypassing the pluripotent stage. Thus, the cellular reprogramming toolbox is now providing the means to address the potential of patient-tailored immune cell types for cancer immunotherapy. In parallel, development of viral vectors for gene delivery has opened the door for in vivo reprogramming in regenerative medicine, an elegant strategy circumventing the current limitations of in vitro cell manipulation. An analogous paradigm has been recently developed in cancer immunotherapy by the generation of CAR T-cells in vivo. These new ideas on endogenous reprogramming, cross-fertilized from the fields of regenerative medicine and gene therapy, are opening exciting avenues for direct modulation of immune or tumor cells in situ, widening our strategies to remove cancer immunotherapy roadblocks. Here, we review current strategies for cancer immunotherapy, summarize technologies for generation of immune cells by cell fate reprogramming as well as highlight the future potential of inducing these unique cell identities in vivo, providing new and exciting tools for the fast-paced field of cancer immunotherapy.


Sign in / Sign up

Export Citation Format

Share Document