Abstract 4734: Impact of various immune escape mechanisms on redirected lysis by T cells engaged via EpCAM/CD3 bispecific BiTE® antibody AMG 110 .

Author(s):  
Wibke Deisting ◽  
Tobias Raum ◽  
Peter Kufer ◽  
Patrick A. Baeuerle ◽  
Markus Muenz
Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4252-4252
Author(s):  
Lisa Christiansson ◽  
Camilla Lindqvist ◽  
Thomas H Tötterman ◽  
Bengt Simonsson ◽  
Ulla Olsson-Strömberg ◽  
...  

Abstract Abstract 4252 Cancer patients are known to have an impaired anti-tumor immune response because of various immune escape mechanisms exerted by the tumor cells. These mechanisms involve release of soluble molecules and expression of membrane-bound proteins inhibiting different arms of the anti-tumor immune responses. The immune escape mechanisms seen in cancer patients complicate the use of immunotherapy, such as tumor vaccines and adoptive T cell transfer. The aim of our study was to investigate the presence of IL10, soluble IL2R (sIL2R; CD25) and programmed death receptor ligand 1 (PDL1) in patients with chronic myeloid leukemia (CML) and to study their role as T cell inhibitors. IL10, sIL2R and PDL1 are all known immune inhibitory molecules. IL10 is a secreted molecule that has various suppressive effects on many different immune cells. sIL2R binds IL2 efficiently and may thereby prevent the binding of IL2 to IL2R on T cells, an interaction that is necessary for proliferation and maintenance of tumor-specific T cells. PDL1-expressing tumor cells can bind programmed death receptor 1 (PD1) on T cells. This interaction can induce apoptosis in the T cells. In this study, we used cytometric bead array, ELISA and multicolor flow cytometry to screen CML patients and healthy controls for the presence of IL10, sIL2R and PDL1 in blood. Further, Alamar Blue TM assay and IFNγ detection by flow cytometry were used to investigate T cell proliferation and activation in the presence of the inhibitors. We found that the levels of IL10 and sIL2R were increased in CML patient plasma compared to the levels in healthy control subjects. The level of sIL2R in a subgroup of patients was four-fold higher than the mean level of healthy controls. Patient T cells stimulated with various strong T cell stimuli including CML-specific peptides failed to respond to stimuli as measured by flow cytometric detection of the cytotoxic T cell activation marker IFNγ, indicating that these T cells might be anergic. In vitro studies on T cells from healthy donors showed that both IL10 and sIL2R have the ability to inhibit T cell proliferation. Half of the CML patients had a PDL1 expression on the CD34 cell population raging from 1-36%. PDL1 may, hence, be involved in T cell control in CML. Taken together, our results show that T cells from CML patients fail to respond to stimuli and that these T cells may be controlled by the high levels of IL10, sIL2R and PDL1 seen in the patients. Screening for these inhibitors may aid selection of patients considered for immunotherapy. Disclosures: Simonsson: Novartis: Consultancy, Honoraria, Research Funding, Sponsor; BMS: Consultancy, Honoraria, Research Funding, Sponsor; SCHERING-PLOUGH: Sponsor. Olsson-Strömberg:BMS: Honoraria.


2020 ◽  
Author(s):  
Nasser K. Altorki ◽  
Alain C. Borczuk ◽  
Vivek Mittal ◽  
Olivier Elemento ◽  
Timothy E. McGraw

SummaryThe tumor microenvironment (TME) of lung adenocarcinoma (LUAD) precursor lesions has not been described. We interrogated by multiplex immunofluorescence the TME of preinvasive and invasive Stage 1A LUADs selected by computer tomography (CT) scan-density. Pure non-solid (p-NS) CT density nodules are preinvasive/minimally invasive, whereas solid CT density nodules are frankly invasive cancers. Our data reveal an intensely immune-suppressive immune TME in p-NS tumors characterized by an increase in Treg cells and a decrease in cytotoxic T cells relative to normal lung. The TME of the solid tumor group, more advanced lesions than the p-NS yet still early in disease development, were increasingly more immune-suppressive. Provocatively, there was a further increase in both Treg cells and cytotoxic T cells, establishing a nascent albeit ineffective anti-tumor immune response in transition from preinvasive p-NS to invasive solid tumors. Regulatory T cells play a dominant role throughout progression, while additional immune evasive mechanisms are employed at different stages of disease progression, including T cell exclusion from cancer cell nests early and activation of immune checkpoints later. Our study establishes that different immune-targeted strategies are required to intercept disease progression at these two distinct early points of lung cancer development.Statement of SignificanceUsing multiplexed IF, we compared the cellular composition and activation state of the tumor immune microenvironment between pre/minimally invasive and frankly invasive adenocarcinoma. We found a progressive increase in immunosuppressive mechanisms in association with disease progression suggesting that Interception strategies should be specifically tailored based on underlying immune escape mechanisms


2020 ◽  
Vol 33 (1) ◽  
pp. 39-48
Author(s):  
Tomoaki Muramatsu ◽  
Takuro Noguchi ◽  
Daisuke Sugiyama ◽  
Yoshie Kanada ◽  
Kaori Fujimaki ◽  
...  

Abstract Tumor neoantigens derived from genetic alterations are potential T-cell targets for antitumor immunity. However, tumors develop immune escape mechanisms including loss of preexisting neoantigens and/or impairment of T-cell responses during tumor development and progression. Here, we addressed whether newly emerged immunogenic neoantigens in established tumors enabled hosts to inhibit tumor growth via controlling immune escape mechanisms. Using a doxycycline-driven gene expression system, we generated murine MC38, CT26 (colorectal cancer) and B16 (melanoma) cell lines with inducible expression of model immunogenic neoantigens such as chicken ovalbumin and human NY-ESO-1. A model neoantigen was induced by doxycycline administration in the tumors once tumors became palpable. Tumor growth was significantly inhibited upon induction of the neoantigen and this inhibition was abrogated in nude mice lacking T cells and in mice deprived of CD8+ T cells, indicating the critical role of CD8+ T cells in tumor regression. In addition, PD-1/PD-L1 blockade further augmented the antitumor immune response, resulting in a far stronger inhibition of tumor growth. Accordingly, newly emerged tumor neoantigen-specific CD8+ T cells with enhanced effector functions were significantly increased in mice treated with PD-1/PD-L1 blockade. We propose that a newly emerged neoantigen is sufficient to inhibit tumor growth via preventing immune escape in a T-cell-dependent manner. Our results imply that induction of immunogenic tumor neoantigens is a novel strategy to overcome the resistance to immune checkpoint blockade therapy.


2012 ◽  
Vol 2012 ◽  
pp. 1-6 ◽  
Author(s):  
V. Procházka ◽  
M. Jarošová ◽  
Z. Prouzová ◽  
R. Nedomová ◽  
T. Papajík ◽  
...  

Diffuse large B-cell lymphoma (DLBCL) is the most frequent subtype of non-Hodgkin lymphomas in Western countries. Implementation of immunotherapy using monoclonal antibodies to therapeutic protocols has led to dramatic improvements in overall survival. DLBCL became a model of a successful immunochemotherapy concept. Despite this fact, there is still a proportion of patients who do not respond to or relapse early after treatment. Growing evidence suggests that host antitumor immunity is suppressed by lymphoma cells in many ways. First, host cytotoxic T cells are directly suppressed by interaction with programmed cell death (PD) ligand on lymphoma cell surface and a similar mechanism enhances the activity of suppressive regulatory T cells (Tregs). Second, tumor cells escape host cytotoxic cells due to lower immunogenicity caused by reduced expression of HLA antigens. Both mechanisms have an origin in primary genetic events in lymphomagenesis. Rearrangement of MHC class II transcriptional activator (CIITA) gene and amplification of Janus kinase (JAK2) gene lead to enhanced expression of PD ligands 1 and 2, higher proliferation activity, and lower expression of HLA. This paper summarizes current knowledge about clinically relevant immune escape mechanisms in DLBCL.


2020 ◽  
Vol 8 (Suppl 2) ◽  
pp. A28-A29
Author(s):  
M Thelen ◽  
K Wennhold ◽  
J Lehmann ◽  
E Staib ◽  
MA Garcia Marquez ◽  
...  

BackgroundImmune-checkpoint inhibition (CKI) demonstrated remarkable therapeutic efficacy in several kinds of cancer. However, immune escape mechanisms lead to primary or secondary resistance in the majority of patients. Most predictive biomarkers failed, as the primary target of CKI is not the tumor cell itself, but the crosstalk between immune- and cancer cells. We aimed to characterize the immune evasion landscape in primary tumors across different entities.Materials and MethodsExpression of 32 immune-regulatory molecules on lymphocytes was analyzed in peripheral blood and tumor infiltrating lymphocytes (TILs) of 146 primary tumor patients across 10 different entities using flow cytometry. NanoString was applied to determine RNA expression of the respective ligands and 20 genes associated with antigen presentation. Expression of coinhibitory ligands on tumor cells was assessed by immunohistochemistry. To quantify the immune cell infiltration, digital pathology was used and the Immunoscore was generated for each patient.ResultsWhile an increase of regulatory T cells was a common feature across all entities, we found site-specific differences regarding other lymphocyte subsets and expression of immune-regulatory molecules by TILs and tumor cells. Expression of co-inhibitory molecules on tumor infiltrating T cells accumulated especially in advanced stage cancers whereas immune cell infiltration was mainly associated with enhanced antigen presentation. Co-expression of multiple immune-inhibitory ligands was most frequent in colorectal, lung and ovarian carcinoma. Genes related to antigen presentation were frequently dysregulated in seminoma, liver and lung cancer.ConclusionsImmune evasion is a common feature of cancer and frequently detected co-occurrence of multiple mechanisms probably contributes to resistance against immunotherapy. We describe substantial heterogeneity regarding immune escape mechanisms between patients with the same primary tumor. Individualized immunotherapeutic strategies based on pretherapeutic evaluation of the immune evasion landscape might help to improve response to CKI.Disclosure InformationM. Thelen: None. K. Wennhold: None. J. Lehmann: None. E. Staib: None. M.A. Garcia Marquez: None. P. Lohneis: None. A. Lechner: None. S. Wagener-Ryczek: None. P.S. Plum: None. D. Pfister: None. F. Dörr: None. D. Beutner: None. F. Thangarajah: None. D. Ratiu: None. W. Malter: None. S. Merkelbach-Bruse: None. C.J. Bruns: None. A. Quaas: None. M.S. von Bergwelt-Baildon: B. Research Grant (principal investigator, collaborator or consultant and pending grants as well as grants already received); Significant; Astellas. F. Consultant/Advisory Board; Modest; Bristol-Myers Squibb. H.A. Schlößer: B. Research Grant (principal investigator, collaborator or consultant and pending grants as well as grants already received); Significant; Astra Zeneca.


2019 ◽  
Author(s):  
Carolina Tiraboschi ◽  
Lucas Gentilini ◽  
Felipe M. Jaworski ◽  
Enrique Corapi ◽  
Carla Velazquez ◽  
...  

AbstractProstate cancer (PCa) is a major health problem worldwide. Taxol derivatives–based chemotherapies or immunotherapies are usually proposed depending on the symptomatic status. In the case of immunotherapy, tumors develop robust immune escape mechanisms that abolish any protective response. However, Docetaxel has been shown to enhance the effectiveness of immunotherapy in a variety of cancers, but to date, the mechanism is still unknown. Herein, we showed first that Galectin-3 (Gal-3) expressed by prostate tumor cells is the principal immunological checkpoint responsible of the failure of immunotherapy; and that Docetaxel leads to the inhibition of Gal-3 expression in PCa cells as well as in clinical samples of mCRPC patients promoting a Th1 response. We thus optimized a prostate cancer animal model that undergoes surgical resection of the tumor like prostatectomy to mimic what is usually performed in patients. More importantly, using low and nontoxic doses of taxane prior to immunotherapy, we were able to directly impact the activation and proliferation of CD8+ cytotoxic T cells through reducing the number of CD8+CD122+CD28-T cells and highly control tumor recurrence. Thus, Gal-3 expression by PCa cells is a key inhibitor for the success of immunotherapy, and low doses of Docetaxel with noncytotoxic effect on leukocyte survival should be used prior to vaccination for all PCa patients. This combined treatment sequence right after surgery would promote the preconditioning of the tumor microenvironment, allowing for effective anti-tumor immunotherapy and can be transferred rapidly to clinical therapeutic protocols.


Pathogens ◽  
2021 ◽  
Vol 10 (8) ◽  
pp. 956
Author(s):  
Kirsten Freitag ◽  
Sara Hamdan ◽  
Matthias J. Reddehase ◽  
Rafaela Holtappels

CD8+ T-cell responses to pathogens are directed against infected cells that present pathogen-encoded peptides on MHC class-I molecules. Although natural responses are polyclonal, the spectrum of peptides that qualify for epitopes is remarkably small even for pathogens with high coding capacity. Among those few that are successful at all, a hierarchy exists in the magnitude of the response that they elicit in terms of numbers of CD8+ T cells generated. This led to a classification into immunodominant and non-immunodominant or subordinate epitopes, IDEs and non-IDEs, respectively. IDEs are favored in the design of vaccines and are chosen for CD8+ T-cell immunotherapy. Using murine cytomegalovirus as a model, we provide evidence to conclude that epitope hierarchy reflects competition on the level of antigen recognition. Notably, high-avidity cells specific for non-IDEs were found to expand only when IDEs were deleted. This may be a host’s back-up strategy to avoid viral immune escape through antigenic drift caused by IDE mutations. Importantly, our results are relevant for the design of vaccines based on cytomegaloviruses as vectors to generate high-avidity CD8+ T-cell memory specific for unrelated pathogens or tumors. We propose the deletion of vector-encoded IDEs to avoid the suppression of epitopes of the vaccine target.


2008 ◽  
Vol 14 (12) ◽  
pp. 1390-1395 ◽  
Author(s):  
Angel Varela-Rohena ◽  
Peter E Molloy ◽  
Steven M Dunn ◽  
Yi Li ◽  
Megan M Suhoski ◽  
...  

2018 ◽  
Vol 18 (2) ◽  
pp. 166-181 ◽  
Author(s):  
Antonio Marra ◽  
Cristina R. Ferrone ◽  
Celeste Fusciello ◽  
Giosue Scognamiglio ◽  
Soldano Ferrone ◽  
...  

Melanoma is an aggressive form of skin cancer characterized by poor prognosis and high mortality. The development of targeted agents based on the discovery of driver mutations as well as the implementation of checkpoint inhibitor-based immunotherapy represents a major breakthrough in the treatment of metastatic melanoma. However, in both cases the development of drug resistance and immune escape mechanisms as well as the lack of predictive biomarkers limits their extraordinary clinical efficacy. In this article, we summarize the available therapeutic options for patients with metastatic melanoma, outline the mechanisms implicated in the resistance to both targeted agents and immunotherapy, discuss potential predictive biomarkers and outline future therapeutic approaches under investigation.


Sign in / Sign up

Export Citation Format

Share Document