Abstract A77: Differential recruitment of myeloid derived immune cells and fibroblasts to the thyroid tumor microenvironment in mouse models of papillary and follicular thyroid cancer

Author(s):  
Lee Ann King ◽  
Sergey Novitsky ◽  
Julio Ricarte-Filho ◽  
Philip Owens ◽  
Aime T. Franco
Human Cell ◽  
2020 ◽  
Vol 33 (4) ◽  
pp. 930-937 ◽  
Author(s):  
Eriko Katsuta ◽  
Omar M. Rashid ◽  
Kazuaki Takabe

2020 ◽  
Vol 21 (17) ◽  
pp. 5950
Author(s):  
Margarita Knyazeva ◽  
Ekaterina Korobkina ◽  
Alexey Karizky ◽  
Maxim Sorokin ◽  
Anton Buzdin ◽  
...  

Over the last few years, incidental thyroid nodules are being diagnosed with increasing frequency with the use of highly sensitive imaging techniques. The ultrasound thyroid gland examination, followed by the fine-needle aspiration cytology is the standard diagnostic approach. However, in cases of the follicular nature of nodules, cytological diagnosis is not enough. Analysis of miRNAs in the biopsy presents a promising approach. Increasing our knowledge of miRNA’s role in follicular carcinogenesis, and development of the appropriate the miRNA analytical technologies are required to implement miRNA-based tests in clinical practice. We used material from follicular thyroid nodes (n.84), grouped in accordance with their invasive properties. The invasion-associated miRNAs expression alterations were assayed. Expression data were confirmed by highly sensitive two-tailed RT-qPCR. Reciprocally dysregulated miRNAs pair concentration ratios were explored as a diagnostic parameter using receiver operation curve (ROC) analysis. A new bioinformatics method (MiRImpact) was applied to evaluate the biological significance of the observed expression alterations. Coupled experimental and computational approaches identified reciprocal dysregulation of miR-146b and miR-451 as important attributes of follicular cell malignant transformation and follicular thyroid cancer progression. Thus, evaluation of combined dysregulation of miRNAs relevant to invasion and metastasis can help to distinguish truly malignant follicular thyroid cancer from indolent follicular adenoma.


2017 ◽  
Vol 35 (15_suppl) ◽  
pp. e14544-e14544 ◽  
Author(s):  
Michal Abraham ◽  
Inbal Mishalian ◽  
Yaniv Harel ◽  
Shiri Klein ◽  
Yaron Pereg ◽  
...  

e14544 Background: Cancer cells affect their micro-environment by recruiting immune cells that support tumor growth, metastasis and inhibition of anti-tumor effector T and NK cell recruitment. In this study, we investigated the role of BL-8040, a CXCR4 antagonist in cancer immunotherapy and its ability to modulate the immunosuppressive tumor micro-environment. Methods: The effect of BL8040 on tumor micro-environment was tested in 3 different cancer mouse models: lung cancer, pancreatic cancer and melanoma. The mobilization of immune cells to the periphery in response to BL8040 was tested, as well as the accumulation of immune cells both within and surrounding the tumor in the pancreatic cancer mouse model. Results: BL8040 was found to be a potent and robust mobilizer of immune cells. Immunophenotyping of the mobilized cells revealed that the mobilization of CD4 and CD8 T lymphocytes, as well as of dendritic cells (DC), was significantly increased in the cancer-bearing mice compared to their naïve counterparts. Importantly, a significant mobilization of effector CD8 T cells and activated CD8 T cells in the cancer-bearing mice was also detected following BL8040 treatment. Concomitantly, in the pancreatic cancer mouse model, treatment with BL8040 increased CD8 T cell accumulation within the tumor and inhibited tumor growth. Conclusions: The immune cell population that is mobilized in response to BL8040 treatment is different in cancer mouse models and naïve mice. The ability of BL8040 to induce mobilization of leukocytes, cytotoxic and activated CD8 T cells and DCs is affected by the presence of a tumor. In our models of pancreatic cancer, mobilization of immune cells from the bone marrow into the circulation and their accumulation within the tumor and tumor microenvironment resulted in inhibition of tumor growth. These results indicate that BL8040 may affect the tumor microenvironment and therefore can potentially synergize with immunomodulatory agents. In vivo pre-clinical studies as well as clinical studies are currently ongoing for testing the combination of BL8040 with immunomodulatory agents in different cancer models.


2020 ◽  
Vol 31 (4) ◽  
pp. 367-376
Author(s):  
Norman G. Nicolson ◽  
Johan O. Paulsson ◽  
C. Christofer Juhlin ◽  
Tobias Carling ◽  
Reju Korah

AbstractWhile minimally invasive follicular thyroid cancer (miFTC) generally has low risk of recurrence or death, encapsulated angioinvasive (eaFTC) or widely invasive (wiFTC) histological subtypes display significantly worse prognosis. Drivers of invasion are incompletely understood. Therefore, tissue samples including miFTC, eaFTC, and wiFTC tumors, as well as histologically normal thyroid adjacent to benign follicular adenomas, were selected from a cohort (n = 21) of thyroid tumor patients, and the gene expression of selected transcription factors was characterized with quantitative PCR. Invasion-relevant spatial expression patterns of selected transcription factors were subsequently characterized with immunohistochemistry. E2F1 was over-expressed in all 3 subtypes (p<0.01). SP1 was differentially expressed in eaFTC and wiFTC compared with normal (p=0.01 and 0.04, respectively). TCF7L2 was significantly upregulated in wiFTC specifically (p<0.05). While these findings were mRNA specific, immunohistochemistry of additional cancer-associated transcription factors revealed differential expression along the tumor invasive front relative to the central tumor, and histone acetylation modulators emerged as putative invasion markers. These findings may have significant implications for the interpretation of bulk gene expression analysis of thyroid tumor samples or for the development of targeted therapeutics for this rare but aggressive thyroid cancer variant.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A43-A43
Author(s):  
Alexa Heaton ◽  
Tiffany Heaster ◽  
Anna Hoefges ◽  
Alexander Rakhmilevich ◽  
Amy Erbe ◽  
...  

BackgroundIntravital multiphoton microscopy (IMM) provides single-cell imaging within intact living systems. IMM of the autofluorescent metabolic co-enzymes NAD(P)H and FAD, or optical metabolic imaging (OMI), provides in vivo label-free imaging of metabolic changes. The metabolism of tumor cells and immune cells is closely associated with cancer progression,1–3 so we aim to study metabolic trends before and after administration of an established, effective, triple-combination immunotherapy within murine melanoma tumors.4 This therapy includes 12 Gy external beam radiation, intratumoral administration of a hu14.18-IL2 immunocytokine (anti-GD2 mAb fused to IL2), and intraperitoneal administration of anti-CTLA-4 leading to in situ vaccination and cure of GD2+ murine tumors.4 Previous work has shown that a T cell response is critical to the efficacy of this therapy,4 5 so we created mCherry-labeled T cell mouse models to study T cell response. Here, IMM was used to image concurrent tumor cell and T cell metabolic trends, T cell infiltration, and tumor microenvironment composition.MethodsWe created mCherry-labeled T cell mouse models through CRISPR/Cas9 knock-in and Cre- lox genetic modifications. We then implanted syngeneic B78 (GD2+) melanoma cells intradermally into the flanks of C57BL/6 mice to induce measurable tumors. Mice were anesthetized, skin flap surgery performed, and tumors imaged at varying time points. IMM was performed using 750–1040 nm to excite NAD(P)H, FAD, and mCherry through a 40X (1.15 NA) objective. Fluorescence lifetime data was collected using time correlated single photon counting electronics. Murine tissues were later harvested and analyzed via flow cytometry and immunohistochemistry to confirm mCherry expression in mouse models and IMM findings.ResultsHere we demonstrate the feasibility of our IMM platform to perform single-cell resolution imaging in vivo. We establish that our genetically engineered mouse models enable clear identification and tracking of mCherry T cell populations. In addition, we show that label-free OMI provides metabolic trends and structural information in vivo (figure 1). Overall, we demonstrate concurrent imaging of intravital tumor cell and T cell populations within the tumor microenvironment.ConclusionsOur preliminary results suggest that the combination of IMM and our mCherry mouse models with OMI allows for concurrent imaging of T cell infiltration and metabolic trends. With continued work, this imaging platform has the potential to provide dynamic, metabolic information on tumor cell and immune cell populations to inform further immunotherapy development.AcknowledgementsThis work is supported by the Morgridge Institute for Research (Interdisciplinary Fellowship awarded to A.R.H.) and the NIH (R01 CA205101 and R35 CA197078). The authors thank the University of Wisconsin Carbone Cancer Center (UWCCC) Support Grant P30 CA014520, the UWCCC Translational Research Initiatives in Pathology laboratory - supported by the UW Department of Pathology and Laboratory Medicine and the Office of The Director NIH (S10OD023526), the UWCCC Flow Cytometry Laboratory, and the Genome Editing and Animal Models Laboratory for core services.Abstract 42 Figure 1Representative intravital multiphoton microscopy images of a B78 syngeneic melanoma growing in a mouse with mCherry-labeled T cells. (A) Fluorescence intensity image of all fluorophores shows mCherry-labeled T cells (red) infiltrating untreated tumor tissue and vasculature as well as metabolic coenzymes NAD(P)H (blue) and FAD (green) expressed by the tumor. (B) Fluorescence intensity image of NAD(P)H alone shows NAD(P)H landscape as well as tumor boundaries and winding vasculature filled with red blood cells. (C) Fluorescence lifetime image shows mCherry-labeled T cell populations and their corresponding mean lifetime (tau m ) values. Fluorescence lifetime values help distinguish mCherry-labeled T cells (typical tau m = 1,400 ps) from nonspecific red autofluorescence in vivo. (D) Fluorescence lifetime image shows NAD(P)H expression and corresponding mean lifetime values which give insight into tumor metabolism and microenvironment.Ethics ApprovalAll animal work was approved by the University of Wisconsin Institutional Animal Care and Use Committees.ReferencesRenner K, Singer K, Koehl GE, Geissler EK, Peter K, Siska PJ, Kreutz M. Metabolic hallmarks of tumor and immune cells in the tumor microenvironment. Front Immunol. 2017, 8 (MAR), 1–11.Mockler MB, Conroy MJ, Lysaght J, Targeting T. Cell Immunometabolism for cancer immunotherapy; understanding the impact of the tumor microenvironment. Front Oncol. 2014, 4 (May), 1–11.Ghesquière B, Wong BW, Kuchnio A, Carmeliet P. Metabolism of stromal and immune cells in health and disease. Nature 2014, 511 (7508), 167–176.Morris ZS, Guy EI, Francis DM, Gressett MM, Werner LR, Carmichael LL, Yang RK, Armstrong EA, Huang S, Navid F, Gillies SD, Korman A, Hank JA, Rakhmilevich AL, Harari PM, Sondel PM. In situ tumor vaccination by combining local radiation and tumor-specific antibody or immunocytokine treatments. Cancer Res 2016;76 (13):3929–3941.Morris ZS, Guy EI, Werner LR, Carlson PM, Heinze CM, Kler JS, Busche SM, Jaquish A, A, Sriramaneni RN, Carmichael LL, Loibner H, Gillies SD, Korman AJ, Erbe AK, Hank J, A, Rakhmilevich AL, Harari PM, Sondel PM. Tumor-specific inhibition of in situ vaccination by distant untreated tumor sites. Cancer Immunol Res 2018;6 (7):825–834.


2018 ◽  
Vol 24 ◽  
pp. 255
Author(s):  
Lakshmi Menon ◽  
Yuanjie Mao ◽  
Sanaz Abedzadeh-Anaraki ◽  
Spyridoula Maraka

2018 ◽  
Author(s):  
Martyna Borowczyk ◽  
Ewelina Szczepanek-Parulska ◽  
Szymon Debicki ◽  
Bartlomiej Budny ◽  
Malgorzata Janicka-Jedynska ◽  
...  

2018 ◽  
Author(s):  
Shazia Hussain ◽  
Carmel Brennan ◽  
Nick Plowman ◽  
Kate Newbold ◽  
William Drake

Sign in / Sign up

Export Citation Format

Share Document