scholarly journals Exosomes: Small EVs with Large Immunomodulatory Effect in Glioblastoma

2021 ◽  
Vol 22 (7) ◽  
pp. 3600
Author(s):  
Laura Benecke ◽  
Mali Coray ◽  
Sandra Umbricht ◽  
Dapi Chiang ◽  
Fabrício Figueiró ◽  
...  

Glioblastomas are among the most aggressive tumors, and with low survival rates. They are characterized by the ability to create a highly immunosuppressive tumor microenvironment. Exosomes, small extracellular vesicles (EVs), mediate intercellular communication in the tumor microenvironment by transporting various biomolecules (RNA, DNA, proteins, and lipids), therefore playing a prominent role in tumor proliferation, differentiation, metastasis, and resistance to chemotherapy or radiation. Exosomes are found in all body fluids and can cross the blood–brain barrier due to their nanoscale size. Recent studies have highlighted the multiple influences of tumor-derived exosomes on immune cells. Owing to their structural and functional properties, exosomes can be an important instrument for gaining a better molecular understanding of tumors. Furthermore, they qualify not only as diagnostic and prognostic markers, but also as tools in therapies specifically targeting aggressive tumor cells, like glioblastomas.

2021 ◽  
Author(s):  
Wyatt M. Becicka ◽  
Peter Bielecki ◽  
Morgan Lorkowski ◽  
Taylor J. Moon ◽  
Yahan Zhang ◽  
...  

The efficacy of immunotherapies is often limited by the immunosuppressive tumor microenvironment, which is populated with dysfunctional innate immune cells. To reprogram the tumor-resident innate immune cells, we developed an...


Cells ◽  
2021 ◽  
Vol 10 (5) ◽  
pp. 1020
Author(s):  
Stefan Grote ◽  
Guillermo Ureña-Bailén ◽  
Kenneth Chun-Ho Chan ◽  
Caroline Baden ◽  
Markus Mezger ◽  
...  

Background: Melanoma is the most lethal of all skin-related cancers with incidences continuously rising. Novel therapeutic approaches are urgently needed, especially for the treatment of metastasizing or therapy-resistant melanoma. CAR-modified immune cells have shown excellent results in treating hematological malignancies and might represent a new treatment strategy for refractory melanoma. However, solid tumors pose some obstacles for cellular immunotherapy, including the identification of tumor-specific target antigens, insufficient homing and infiltration of immune cells as well as immune cell dysfunction in the immunosuppressive tumor microenvironment (TME). Methods: In order to investigate whether CAR NK cell-based immunotherapy can overcome the obstacles posed by the TME in melanoma, we generated CAR NK-92 cells targeting CD276 (B7-H3) which is abundantly expressed in solid tumors, including melanoma, and tested their effectivity in vitro in the presence of low pH, hypoxia and other known factors of the TME influencing anti-tumor responses. Moreover, the CRISPR/Cas9-induced disruption of the inhibitory receptor NKG2A was assessed for its potential enhancement of NK-92-mediated anti-tumor activity. Results: CD276-CAR NK-92 cells induced specific cytolysis of melanoma cell lines while being able to overcome a variety of the immunosuppressive effects normally exerted by the TME. NKG2A knock-out did not further improve CAR NK-92 cell-mediated cytotoxicity. Conclusions: The strong cytotoxic effect of a CD276-specific CAR in combination with an “off-the-shelf” NK-92 cell line not being impaired by some of the most prominent negative factors of the TME make CD276-CAR NK-92 cells a promising cellular product for the treatment of melanoma and beyond.


Cancers ◽  
2021 ◽  
Vol 13 (8) ◽  
pp. 1777
Author(s):  
Ying Zhu ◽  
Sammy Ferri-Borgogno ◽  
Jianting Sheng ◽  
Tsz-Lun Yeung ◽  
Jared K. Burks ◽  
...  

Stromal and immune cells in the tumor microenvironment (TME) have been shown to directly affect high-grade serous ovarian cancer (HGSC) malignant phenotypes, however, how these cells interact to influence HGSC patients’ survival remains largely unknown. To investigate the cell-cell communication in such a complex TME, we developed a SpatioImageOmics (SIO) pipeline that combines imaging mass cytometry (IMC), location-specific transcriptomics, and deep learning to identify the distribution of various stromal, tumor and immune cells as well as their spatial relationship in TME. The SIO pipeline automatically and accurately segments cells and extracts salient cellular features to identify biomarkers, and multiple nearest-neighbor interactions among tumor, immune, and stromal cells that coordinate to influence overall survival rates in HGSC patients. In addition, SIO integrates IMC data with microdissected tumor and stromal transcriptomes from the same patients to identify novel signaling networks, which would lead to the discovery of novel survival rate-modulating mechanisms in HGSC patients.


2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A312-A312
Author(s):  
Shuming Chen ◽  
Tracee McMiller ◽  
Preethi Sankaran ◽  
Kyle Kampta ◽  
Suzanne Topalian

BackgroundWe previously found upregulation of the cyclooxygenase-2/prostaglandin E2 (COX-2/PGE2) pathway in the tumor microenvironment (TME) of cancers that respond poorly to anti-PD-1 therapy.1–2 The potential functional role of this pathway in anti-PD-1 resistance is unknown. We therefore studied modulation of COX-2 expression in cultured human tumor and immune cells, PGE2-mediated effects on myeloid cells and their reversal with prostaglandin (EP) receptor inhibitors.MethodsNineteen tumor lines representing 6 histologies were treated with cytokines reported to induce COX-2 (IL-1B, IL-17A, TNF-a). Peripheral blood monocytes (Monos) were treated with toll-like receptor (TLR) agonists or TME-resident cytokines associated with high PD-L1 expression (IL-1A, IL-10, IL-27, IL-32g, IFN-g).3–4 COX-2 protein was detected by Western blotting and flow cytometry. In some experiments, Monos were pre-incubated with EP2i (PF-04418948) and/or EP4i (ONO-AE3-208), then treated with PGE2 ± TLR4 (LPS) or TLR7 (imiquimod) agonists. IL-6, IL-10, TNF-a, and VEGF secretion were detected by ELISA. Monocytic DCs generated with GM-CSF+IL-4 were matured with CD40L, ± PGE2, then phenotyped.ResultsAmong 19 tumor cell lines, 6 expressed COX-2 constitutively, and 13 were induced to express COX-2 by 1-day exposure to IL-1B, IL-17A, or TNF-a. In Monos, COX-2 was induced by IL-1A and IL-1B, but not IFN-g or IL-27. TLR 1-9 agonists induced COX-2, with TLR2/4/5 agonists being the strongest inducers. COX-2 induction by these factors was non-overlapping with PD-L1 induction in tumor cells and Monos, suggesting non-redundant pathways of immune resistance. PGE2 had context-dependent effects in Monos, depending on the cytokines, TLR agonists, and donors assayed: PGE2 increased VEGF secretion by resting Monos from 4/4 donors tested, but increased IL-6, IL-10 and TNF-a secretion in only 1/4 donors; PGE2 increased imiquimod-induced TNF-a secretion, but decreased LPS-induced TNF-a secretion. EP2 and EP4 inhibitors counteracted PGE2-mediated cytokine modulation, and showed synergistic effects when combined in the context of high dose of PGE2 (500nM). Additionally, PGE2 suppressed the in vitro generation of mature DCs, reducing CD80 and CD83 expression and increasing CD16.ConclusionsUnderstanding and preventing anti-PD-1 treatment resistance is a critical goal. Our results suggest that the COX-2/PGE2 pathway is expressed in tumor and immune cells, and modulates myeloid cell functions in a context-dependent manner. COX-2 expression is non-redundant with PD-L1 expression, providing a rationale to test COX-2 pathway inhibition in conjunction with anti-PD-1. Available drugs targeting this pathway, including IL-1R and IL-1B inhibitors, NSAIDs, and EP2 and EP4 inhibitors, will enable the clinical development of combination treatment regimens.AcknowledgementsWe gratefully acknowledge support from NCI R01-CA142779, Bristol Myers Squibb, and the Johns Hopkins Bloomberg~Kimmel Institute for Cancer Immunotherapy.ReferencesDuffield AS, Ascierto ML, Anders RA, et al. The immunosuppressive tumor microenvironment (TME) in nasopharyngeal carcinoma: implications for immunotherapy. AACR 2018;Abstract 4750.Besharati S, McMiller T, Yarchoan M, et al. The immunosuppressive tumor microenvironment (TME) in epstein-barr virus (EBV)-positive and EBV-negative gastric cancers: implications for immunotherapy. SITC 2018;P541 (abstr).Taube JM, Young GD, McMiller TL, et al. Differential expression of immune-regulatory genes associated with PD-L1 display in melanoma: implications for PD-1 pathway blockade. Clin Cancer Res 2015;21:3969–76.Duffield AS, Ascierto ML, Anders RA, et al. Th17 immune microenvironment in epstein-barr virus negative Hodgkin lymphoma: implications for immunotherapy. Blood Advances 2017;1:1324–34.


Biomolecules ◽  
2021 ◽  
Vol 11 (8) ◽  
pp. 1170
Author(s):  
Lin Jin ◽  
Hong Sun Kim ◽  
Jiaqi Shi

Pancreatic ductal adenocarcinoma (PDAC) is a malignancy with a poor prognosis and low survival rates. PDAC is characterized by a fibroinflammatory tumor microenvironment enriched by abundant fibroblasts and a variety of immune cells, contributing to its aggressiveness. Neutrophils are essential infiltrating immune cells in the PDAC microenvironment. Recent studies have identified several cellular mechanisms by which neutrophils are recruited to tumor lesion and promote tumorigenesis. This review summarizes the current understanding of the interplay between neutrophils, tumor cells, and other components in the PDAC tumor microenvironment. The prognosis and therapeutic implications of neutrophils in PDAC are also discussed.


2021 ◽  
Vol 23 (Supplement_6) ◽  
pp. vi13-vi14
Author(s):  
Quinn Ostrom ◽  
Joanne Xiu ◽  
Giselle lopez ◽  
Ashley Sumrall ◽  
Sonikpreet Aulakh ◽  
...  

Abstract INTRODUCTION IDH mutations are a defining feature of lower-grade glioma and secondary glioblastoma. Approximately 95% of glioma-associated IDH mutations are in codon 132 of IDH1, but a small proportion are in IDH2. IDH mutations produce the oncometabolite 2-hydroxyglutarate, which induces global DNA hypermethylation and is associated with an immunosuppressive tumor microenvironment. IDH1 is localized in the cytosol while IDH2 is found in the mitochondrial matrix, and mutations in these genes may have differing effects on the tumor microenvironment. METHODS Formalin-fixed, paraffin-embedded tissue from 633 IDH-mutant gliomas (615 IDH1-mutant, 18 IDH2-mutant) underwent whole-exome and whole-transcriptome sequencing at Caris Life Sciences (236 grade 2/3 astrocytoma, 158 grade 2/3 oligodendroglioma, 202 IDH-mutant glioblastoma, 37 glioma, NOS). QuantiSEQ was used to infer tumor-infiltrating immune cell populations from RNAseq data, and gene-set enrichment analyses (GSEA) were performed using Wikipathway. RESULTS IDH1-mutant gliomas had higher levels of pro-inflammatory M1 macrophages (P=0.04), modestly higher levels of monocytes (P=0.08), and lower levels of neutrophils (P=0.04) – typically considered immunosuppressive – compared with IDH2-mutant gliomas. No differences were observed in levels of B cells, dendritic cells, NK cells, or T cell subsets (Treg, CD4+, CD8+). IDH2-mutant gliomas were enriched for hallmark oligodendroglioma mutations (TERT promoter, CIC, FUBP1), while IDH1-mutant gliomas were enriched for hallmark astrocytoma mutations (ATRX, TP53). However, associations with tumor-infiltrating immune cells persisted after excluding 1p/19q co-deleted oligodendroglioma from analyses. GSEA revealed upregulation of the microglial TYROBP signaling pathway, the microglial phagocytic pathway, and of Type II Interferon signaling in IDH1-mutant gliomas versus IDH2-mutant gliomas. CONCLUSIONS Although IDH2 mutations are generally thought to function similarly to IDH1 mutations, we observe differences in tumor-infiltrating immune cells across groups. IDH2-mutant gliomas appeared to have a more immunosuppressive tumor microenvironment than their IDH1-mutant counterparts. Early-phase immunotherapy trials should consider covariate-adaptive randomization approaches to equally allocate IDH2-mutant gliomas across treatment arms.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A176-A176
Author(s):  
Xue Yao ◽  
Sandro Matosevic

BackgroundThe effectiveness of natural killer cell-based immunotherapy against solid tumors is limited by the lack of specific antigens and the immunosuppressive tumor microenvironment. To improve the clinical efficacy and specificity of NK cell therapy, we are designing, developing, and characterizing a new generation of multi-specific killer engagers, which consists of a neoantigen-targeting moiety, together with cytokine and chemokine-producing domains.MethodsTargeting a neoantigen-an antigen formed specifically in response to tumor genome mutations-enables substantially enhanced tumor specificity to be achieved. We evaluated the responsiveness of NK cells to Wilms Tumor 1 (WT1) antigen in GBM by synthesizing an antibody that is able to recognize the WT1/HLA complex. Incorporation of cytokine (namely IL-2, IL-15, and IL-21)-essential for the maturation, persistence, and expansion of NK cells in vivo-favors the proliferation and survival of NK cells in the tumor microenvironment, thereby leading to more sustained anti-tumor responses. Additionally, our data have indicated that the chemokine CXCL10 plays an important role in the infiltration of immune cells into GBM, yet the chemokine itself is expressed at low levels in GBM. Incorporation of a CXCL10-producing element into our construct further supports NK cell recruitment and may stimulate the recruitment of other immune cells. NK activation through the tri-specific engager is achieved through NKp46-mediated signaling. We are investigating the ability of the tri-functional engager to support and enhance NK cell-mediated cytotoxicity against GBM in vitro and in patient-derived GBM xenografts in vivo.ResultsWe hypothesize that taking advantage of our multi-functional engager, NK cells will exhibit, at once, superior persistence, infiltration and antitumor activity, simultaneously addressing three of the main limitations to the use of NK cells in immunotherapy of GBM and other solid tumors.ConclusionsN/AAcknowledgementsN/A


2020 ◽  
Vol 28 ◽  
Author(s):  
RamaRao Malla ◽  
Mohammad Amjad Kamal

: The breast tumor microenvironment (TME) promotes drug resistance through an elaborated interaction of TME components mediated by reactive oxygen species (ROS). Despite a massive accumulation of data concerning the targeting the ROS, but little is known about the ROS-responsive nanomedicine for targeting breast TME. This review submits the ROS landscape in breast TME, including ROS biology, ROS mediated carcinogenesis, reprogramming of stromal and immune cells of TME. We also discussed ROS-based precision strategies for imaging TME, including molecular imaging techniques with advanced probes, multiplexed methods, and multi-omic profiling strategies. ROS-responsive nanomedicine also describes various therapies, such as chemo-dynamic, photodynamic, photothermal, sono-dynamic, immune, and gene therapy for BC. We expound ROS-responsive primary delivery systems for chemotherapeutics, phytochemicals, and immunotherapeutics. This review also presents recent updates on nano-theranostics for simultaneous diagnosis and treatment of BCs. We assume that review on this advancing field will be beneficial to the development of ROS-based nanotheranostics for BC.


Author(s):  
Mª Carmen Ocana ◽  
Beatriz Martinez-Poveda ◽  
Ana R. Quesada ◽  
Miguel Angel Medina

Sign in / Sign up

Export Citation Format

Share Document