80 results on '"Konrad Gabrusiewicz"'
Search Results
2. Glioblastoma stem cell-derived exosomes induce M2 macrophages and PD-L1 expression on human monocytes
- Author
-
Konrad Gabrusiewicz, Xu Li, Jun Wei, Yuuri Hashimoto, Anantha L. Marisetty, Martina Ott, Fei Wang, David Hawke, John Yu, Luke M. Healy, Anwar Hossain, Johnny C. Akers, Sourindra N. Maiti, Shinji Yamashita, Yuzaburo Shimizu, Kenneth Dunner, M. Anna Zal, Jared K. Burks, Joy Gumin, Felix Nwajei, Aras Rezavanian, Shouhao Zhou, Ganesh Rao, Raymond Sawaya, Gregory N. Fuller, Jason T. Huse, Jack P. Antel, Shulin Li, Laurence Cooper, Erik P. Sulman, Clark Chen, Changiz Geula, Raghu Kalluri, Tomasz Zal, and Amy B. Heimberger
- Subjects
cancer stem cells ,exosome ,glioblastoma ,immune cells ,stat3 ,pd-l1 ,Immunologic diseases. Allergy ,RC581-607 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Exosomes can mediate a dynamic method of communication between malignancies, including those sequestered in the central nervous system and the immune system. We sought to determine whether exosomes from glioblastoma (GBM)-derived stem cells (GSCs) can induce immunosuppression. We report that GSC-derived exosomes (GDEs) have a predilection for monocytes, the precursor to macrophages. The GDEs traverse the monocyte cytoplasm, cause a reorganization of the actin cytoskeleton, and skew monocytes toward the immune suppresive M2 phenotype, including programmed death-ligand 1 (PD-L1) expression. Mass spectrometry analysis demonstrated that the GDEs contain a variety of components, including members of the signal transducer and activator of transcription 3 (STAT3) pathway that functionally mediate this immune suppressive switch. Western blot analysis revealed that upregulation of PD-L1 in GSC exosome-treated monocytes and GBM-patient-infiltrating CD14+ cells predominantly correlates with increased phosphorylation of STAT3, and in some cases, with phosphorylated p70S6 kinase and Erk1/2. Cumulatively, these data indicate that GDEs are secreted GBM-released factors that are potent modulators of the GBM-associated immunosuppressive microenvironment.
- Published
- 2018
- Full Text
- View/download PDF
3. Macrophage Ablation Reduces M2-Like Populations and Jeopardizes Tumor Growth in a MAFIA-Based Glioma Model
- Author
-
Konrad Gabrusiewicz, Mohammad B. Hossain, Nahir Cortes-Santiago, Xuejun Fan, Bozena Kaminska, Frank C. Marini, Juan Fueyo, and Candelaria Gomez-Manzano
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Monocytes/macrophages are an influential component of the glioma microenvironment. However, understanding their diversity and plasticity constitute one of the most challenging areas of research due to the paucity of models to study these cells' inherent complexity. Herein, we analyzed the role of monocytes/macrophages in glioma growth by using a transgenic model that allows for conditional ablation of this cell population. We modeled glioma using intracranial GL261-bearing CSF-1R–GFP+ macrophage Fas-induced apoptosis (MAFIA) transgenic mice. Conditional macrophage ablation was achieved by exposure to the dimerizer AP20187. Double immunofluorescence was used to characterize M1- and M2-like monocytes/macrophages during tumor growth and after conditional ablation. During glioma growth, the monocyte/macrophage population consisted predominantly of M2 macrophages. Conditional temporal depletion of macrophages reduced the number of GFP+ cells, targeting mainly the repopulation of M2-polarized cells, and altered the appearance of M1-like monocytes/macrophages, which suggested a shift in the M1/M2 macrophage balance. Of interest, compared with control-treated mice, macrophage-depleted mice had a lower tumor mitotic index, microvascular density, and reduced tumor growth. These results demonstrated the possibility of studying in vivo the role and phenotype of macrophages in gliomas and suggested that transitory depletion of CSF-1R+ population influences the reconstitutive phenotypic pool of these cells, ultimately suppressing tumor growth. The MAFIA model provides a much needed advance in defining the role of macrophages in gliomas.
- Published
- 2015
- Full Text
- View/download PDF
4. Author Correction: FGL2 promotes tumor progression in the CNS by suppressing CD103+ dendritic cell differentiation
- Author
-
Jun Yan, Qingnan Zhao, Konrad Gabrusiewicz, Ling-Yuan Kong, Xueqing Xia, Jian Wang, Martina Ott, Jingda Xu, R. Eric Davis, Longfei Huo, Ganesh Rao, Shao-Cong Sun, Stephanie S. Watowich, Amy B. Heimberger, and Shulin Li
- Subjects
Science - Abstract
The original version of this Article contained errors in the author affiliations. Qingnan Zhao, Xueqing Xia, Longfei Huo and Shulin Li were incorrectly associated with Beijing Institute for Brain Disorders, 100069, Beijing, China.This has now been corrected in both the PDF and HTML versions of the Article.
- Published
- 2019
- Full Text
- View/download PDF
5. The Controversial Role of Microglia in Malignant Gliomas
- Author
-
Jun Wei, Konrad Gabrusiewicz, and Amy Heimberger
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Abstract
Malignant gliomas contain stroma and a variety of immune cells including abundant activated microglia/macrophages. Mounting evidence indicates that the glioma microenvironment converts the glioma-associated microglia/macrophages (GAMs) into glioma-supportive, immunosuppressive cells; however, GAMs can retain intrinsic anti-tumor properties. Here, we review and discuss this duality and the potential therapeutic strategies that may inhibit their glioma-supportive and propagating functions.
- Published
- 2013
- Full Text
- View/download PDF
6. Characteristics of the alternative phenotype of microglia/macrophages and its modulation in experimental gliomas.
- Author
-
Konrad Gabrusiewicz, Aleksandra Ellert-Miklaszewska, Maciej Lipko, Malgorzata Sielska, Marta Frankowska, and Bozena Kaminska
- Subjects
Medicine ,Science - Abstract
Microglia (brain resident macrophages) accumulate in malignant gliomas and instead of initiating the anti-tumor response, they switch to a pro-invasive phenotype, support tumor growth, invasion, angiogenesis and immunosuppression by release of cytokines/chemokines and extracellular matrix proteases. Using immunofluorescence and flow cytometry, we demonstrate an early accumulation of activated microglia followed by accumulation of macrophages in experimental murine EGFP-GL261 gliomas. Those cells acquire the alternative phenotype, as evidenced by evaluation of the production of ten pro/anti-inflammatory cytokines and expression profiling of 28 genes in magnetically-sorted CD11b(+) cells from tumor tissues. Furthermore, we show that infiltration of implanted gliomas by amoeboid, Iba1-positive cells can be reduced by a systematically injected cyclosporine A (CsA) two or eight days after cell inoculation. The up-regulated levels of IL-10 and GM-CSF, increased expression of genes characteristic for the alternative and pro-invasive phenotype (arg-1, mt1-mmp, cxcl14) in glioma-derived CD11b(+) cells as well as enhanced angiogenesis and tumor growth were reduced in CsA-treated mice. Our findings define for the first time kinetics and biochemical characteristics of glioma-infiltrating microglia/macrophages. Inhibition of the alternative activation of tumor-infiltrating macrophages significantly reduced tumor growth. Thus, blockade of microglia/macrophage infiltration and their pro-invasive functions could be a novel therapeutic strategy in malignant gliomas.
- Published
- 2011
- Full Text
- View/download PDF
7. Cytotoxic T lymphocytes targeting a conserved SARS-CoV-2 spike epitope are efficient serial killers
- Author
-
Mohsen Fathi, Daniel D. Meyer, Lindsey Charley, Konrad Gabrusiewicz, and Navin Varadarajan
- Subjects
Epitopes ,COVID-19 Vaccines ,SARS-CoV-2 ,Spike Glycoprotein, Coronavirus ,COVID-19 ,Humans ,Antibodies, Viral ,Article ,General Biochemistry, Genetics and Molecular Biology ,Biotechnology ,T-Lymphocytes, Cytotoxic - Abstract
Understanding the cellular immune response to infections, cancers and vaccines lags behind the investigation of humoral responses. While neutralizing antibody responses wane over time, the ability of T cells to recognize viruses including SARS-CoV-2 is instrumental to providing long-term immunity. Although T-cell receptor (TCR) repertoire screening can provide insights into the skewing of a T-cell response elicited upon vaccination or infection, they unfortunately provide no assessment into the functional capacity of T cells or their ability to eliminate virally infected targets. We have used time-lapse imaging microscopy in nanowell grids (TIMING) to integrate the migration of individual T cells with analysis of effector functions including cytokine secretion and cytotoxicity. Machine learning is then applied to study thousands of videos of dynamic interactions as T cells with specificity for SARS-CoV-2 eliminate targets bearing spike protein as a surrogate for viral infection. Our data provide the first direct evidence that cytotoxic T lymphocytes from a convalescent patient targeting an epitope conserved across all known variants of concern (VoC) are serial killers capable of eliminating multiple infected targets. These data have implications for development of vaccines to provide broad and sustained cellular immunity and for the recovery and monitoring of individuals who have been exposed to SARS-CoV-2.Multidisciplinary abstractWe present an imaging platform that uses artificial intelligence (AI) to track thousands of individual cell-cell interactions within nanowell arrays. We apply this platform to quantify how the T cell component of adaptive immunity responds to infections. Our results show that T cells specific for a conserved epitope within the SARS-CoV-2 spike protein are serial killers that can rapidly eliminate virally infected targets. The ability to map the functional capacity of T cells and their ability to kill infected cells provides fundamental insights into the immunology of vaccines and recovery from infections.Graphical Abstract
- Published
- 2022
8. Single-Cell Cloning of Breast Cancer Cells Secreting Specific Subsets of Extracellular Vesicles
- Author
-
Navin Varadarajan, Robiya Joseph, Sendurai A. Mani, Jay R Adolacion, Xingyue An, Konrad Gabrusiewicz, Mohsen Fathi, and Melisa Martinez-Paniagua
- Subjects
Cancer Research ,Cell division ,Cell ,exosomes ,macrophage ,Article ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Single-cell analysis ,medicine ,single-cell analysis ,metastasis ,Secretion ,RC254-282 ,030304 developmental biology ,0303 health sciences ,Chemistry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Microvesicles ,3. Good health ,Cell biology ,medicine.anatomical_structure ,Oncology ,Cell culture ,030220 oncology & carcinogenesis ,Cancer cell - Abstract
Simple Summary Extracellular vesicles (EVs) are a pivotal mechanism for long-distance intercellular communication and facilitate the stable transport of biological information. Conventional methods for profiling EVs are focused on the biological cargo obtained from large populations of cells and cannot map the secretion of specific subsets of EVs onto their cell of origin. We developed a high-throughput single-cell cloning method that can identify the kinetics of secretion of specific subsets of EVs. With the aid of this methodology, we illustrate that secretion of specific subsets of EVs can be an inheritable property of cancer cells. Our single-cell methodology enables the direct integration of EV secretion with multiple cellular functions and can enable new insights into cell and disease biology. Abstract Extracellular vesicles (EVs) mediate communication in health and disease. Conventional assays are limited in profiling EVs secreted from large populations of cells and cannot map EV secretion onto individual cells and their functional profiles. We developed a high-throughput single-cell technique that enabled the mapping of dynamics of EV secretion. By utilizing breast cancer cell lines, we established that EV secretion is heterogeneous at the single-cell level and that non-metastatic cancer cells can secrete specific subsets of EVs. Single-cell RNA sequencing confirmed that pathways related to EV secretion were enriched in the non-metastatic cells compared with metastatic cells. We established isogenic clonal cell lines from non-metastatic cells with differing propensities for CD81+CD63+EV secretion and showed for the first time that specificity in EV secretion is an inheritable property preserved during cell division. Combined in vitro and animal studies with these cell lines suggested that CD81+CD63+EV secretion can impede tumor formation. In human non-metastatic breast tumors, tumors enriched in signatures of CD81+CD63+EV have a better prognosis, higher immune cytolytic activity, and enrichment of pro-inflammatory macrophages compared with tumors with low CD81+CD63+EVs signatures. Our single-cell methodology enables the direct integration of EV secretion with multiple cellular functions and enables new insights into cell/disease biology.
- Published
- 2021
9. Targeting the αv integrin/TGF-β axis improves natural killer cell function against glioblastoma stem cells
- Author
-
Natalie W. Fowlkes, Jian Hu, Yuefan Huang, Ye Li, Katayoun Rezvani, Luis Muniz-Feliciano, Amy B. Heimberger, May Daher, Jinzhuang Dou, Nadima Uprety, Elizabeth J. Shpall, Jun Jun Lu, Enli Liu, Gonca Ozcan, Nobuhiko Imahashi, Qi Miao, Ken Chen, Ana K. Nunez-Cortes, Mayela Mendt, Cynthia Kassab, Daniel Zamler, Li Li, Konrad Gabrusiewicz, Joy Gumin, Jun Yu, Yifei Shen, Elif Gokdemir, Corry M. Jones, Fang Wang, Sufang Li, Mecit Kaplan, Vakul Mohanty, Stephan Mielke, Richard E. Champlin, Mustafa H Bdiwi, Rafet Basar, Mayra Shanley, Matthias Eyrich, Giulio Draetta, Pinaki P. Banerjee, Dihua Yu, Sunil Acharya, David Marin, Emily Ensley, Lucila Nassif Kerbauy, Abdullah Alsuliman, Jun Wei, Frederick F. Lang, Hila Shaim, and April L. Gilbert
- Subjects
Male ,0301 basic medicine ,Integrins ,endocrine system ,medicine.medical_treatment ,Integrin ,Cell ,Biology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Cancer immunotherapy ,Transforming Growth Factor beta ,medicine ,Animals ,Humans ,Mass cytometry ,Receptor, Transforming Growth Factor-beta Type II ,General Medicine ,In vitro ,Neoplasm Proteins ,Killer Cells, Natural ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Neoplastic Stem Cells ,Cancer research ,biology.protein ,Heterografts ,Female ,Stem cell ,Glioblastoma ,Neoplasm Transplantation ,Research Article ,Transforming growth factor - Abstract
Glioblastoma multiforme (GBM), the most aggressive brain cancer, recurs because glioblastoma stem cells (GSCs) are resistant to all standard therapies. We showed that GSCs, but not normal astrocytes, are sensitive to lysis by healthy allogeneic natural killer (NK) cells in vitro. Mass cytometry and single-cell RNA sequencing of primary tumor samples revealed that GBM tumor–infiltrating NK cells acquired an altered phenotype associated with impaired lytic function relative to matched peripheral blood NK cells from patients with GBM or healthy donors. We attributed this immune evasion tactic to direct cell-to-cell contact between GSCs and NK cells via αv integrin–mediated TGF-β activation. Treatment of GSC-engrafted mice with allogeneic NK cells in combination with inhibitors of integrin or TGF-β signaling or with TGFBR2 gene–edited allogeneic NK cells prevented GSC-induced NK cell dysfunction and tumor growth. These findings reveal an important mechanism of NK cell immune evasion by GSCs and suggest the αv integrin/TGF-β axis as a potentially useful therapeutic target in GBM.
- Published
- 2021
10. FGL2 promotes tumor progression in the CNS by suppressing CD103+ dendritic cell differentiation
- Author
-
Jian Wang, Ling Yuan Kong, Jun Yan, Jingda Xu, Amy B. Heimberger, R. Eric Davis, Longfei Huo, Shao Cong Sun, Martina Ott, Shulin Li, Ganesh Rao, Stephanie S. Watowich, Xueqing Xia, Qingnan Zhao, and Konrad Gabrusiewicz
- Subjects
CD4-Positive T-Lymphocytes ,0301 basic medicine ,Cellular differentiation ,General Physics and Astronomy ,02 engineering and technology ,Dendritic cell differentiation ,CD8-Positive T-Lymphocytes ,p38 Mitogen-Activated Protein Kinases ,Mice ,lcsh:Science ,Multidisciplinary ,Brain Neoplasms ,NF-kappa B ,Cell Differentiation ,021001 nanoscience & nanotechnology ,FGL2 ,Tumor Burden ,Gene Expression Regulation, Neoplastic ,STAT Transcription Factors ,Disease Progression ,Neoplastic Stem Cells ,Heterografts ,Stem cell ,0210 nano-technology ,Integrin alpha Chains ,Neuroglia ,Science ,Brain tumor ,Mice, Transgenic ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Antigens, CD ,Cell Line, Tumor ,Glioma ,medicine ,Animals ,Humans ,Author Correction ,Fibrinogen ,Dendritic Cells ,General Chemistry ,medicine.disease ,Survival Analysis ,nervous system diseases ,030104 developmental biology ,Cell culture ,Tumor progression ,Cancer research ,lcsh:Q ,Glioblastoma - Abstract
Few studies implicate immunoregulatory gene expression in tumor cells in arbitrating brain tumor progression. Here we show that fibrinogen-like protein 2 (FGL2) is highly expressed in glioma stem cells and primary glioblastoma (GBM) cells. FGL2 knockout in tumor cells did not affect tumor-cell proliferation in vitro or tumor progression in immunodeficient mice but completely impaired GBM progression in immune-competent mice. This impairment was reversed in mice with a defect in dendritic cells (DCs) or CD103+ DC differentiation in the brain and in tumor-draining lymph nodes. The presence of FGL2 in tumor cells inhibited granulocyte-macrophage colony-stimulating factor (GM-CSF)-induced CD103+ DC differentiation by suppressing NF-κB, STAT1/5, and p38 activation. These findings are relevant to GBM patients because a low level of FGL2 expression with concurrent high GM-CSF expression is associated with higher CD8B expression and longer survival. These data provide a rationale for therapeutic inhibition of FGL2 in brain tumors., Fibrinogen-like protein 2 (FGL2) mediates immune suppression in glioblastoma (GBM). Here, the authors show that FGL-2 expressed by GBM cancer cells acts by suppressing the differentiation of CD103+ DC cells required to activate the anti-tumor CD8+ T cell response via blocking GM-CSF signalling at NFKB, STAT1/5 and p38 level.
- Published
- 2019
11. Re: Human Chimeric Antigen Receptor Macrophages for Cancer Immunotherapy
- Author
-
Benjamin A. Garcia, Saar Gill, Dylan M. Marchione, Konrad Gabrusiewicz, Kimberly Veliz, Xueqing Maggie Lu, Nicholas R. Anderson, Olga Shestova, Miriam Y. Kim, Feng Shen, Stephen R. Wallace, Michael Klichinsky, Xinhe Shan, Carl H. June, Roddy S. O’Connor, Kristin Blouch, Maksim Shestov, Marco Ruella, Yumi Yashiro-Ohtani, Miroslaw Kozlowski, Martha Zeeman, Katherine D. Cummins, Maggie Schmierer, Andrew Best, Nicholas E. Petty, and Saad S. Kenderian
- Subjects
Lung Neoplasms ,Cell Survival ,T cell ,Urology ,medicine.medical_treatment ,Antigen presentation ,Biomedical Engineering ,Bioengineering ,Biology ,Applied Microbiology and Biotechnology ,Immunotherapy, Adoptive ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Antigen ,Cancer immunotherapy ,Cell Line, Tumor ,Neoplasms ,medicine ,Macrophage ,Animals ,Humans ,030304 developmental biology ,0303 health sciences ,Tumor microenvironment ,Microscopy, Video ,business.industry ,Macrophages ,Immunotherapy ,Neoplasms, Experimental ,Chimeric antigen receptor ,medicine.anatomical_structure ,Humanized mouse ,Cancer research ,Molecular Medicine ,business ,030217 neurology & neurosurgery ,Biotechnology - Abstract
Chimeric antigen receptor (CAR) T cell therapy has shown promise in hematologic malignancies, but its application to solid tumors has been challenging1–4. Given the unique effector functions of macrophages and their capacity to penetrate tumors5, we genetically engineered human macrophages with CARs to direct their phagocytic activity against tumors. We found that a chimeric adenoviral vector overcame the inherent resistance of primary human macrophages to genetic manipulation and imparted a sustained pro-inflammatory (M1) phenotype. CAR macrophages (CAR-Ms) demonstrated antigen-specific phagocytosis and tumor clearance in vitro. In two solid tumor xenograft mouse models, a single infusion of human CAR-Ms decreased tumor burden and prolonged overall survival. Characterization of CAR-M activity showed that CAR-Ms expressed pro-inflammatory cytokines and chemokines, converted bystander M2 macrophages to M1, upregulated antigen presentation machinery, recruited and presented antigen to T cells and resisted the effects of immunosuppressive cytokines. In humanized mouse models, CAR-Ms were further shown to induce a pro-inflammatory tumor microenvironment and boost anti-tumor T cell activity. Primary macrophages engineered to express chimeric antigen receptors have anti-tumor activity in humanized mice.
- Published
- 2021
12. Functional single-cell profiling identifies that exosomes are associated with increased immune cell infiltration in non-metastatic breast cancer
- Author
-
Robiya Joseph, Jay R Adolacion, Konrad Gabrusiewicz, Melisa Martinez-Paniagua, Mohsen Fathi, Xingyue An, Sendurai A. Mani, and Navin Varadarajan
- Subjects
Immune system ,medicine.anatomical_structure ,Cell culture ,Cancer cell ,Cell ,medicine ,Cancer research ,Secretion ,Biology ,Carcinogenesis ,medicine.disease_cause ,Exosome ,Microvesicles - Abstract
Exosomes mediate intercellular communication in health and disease. Conventional assays are limited in profiling exosomes secreted from large populations of cells and are unsuitable for studying the functional consequences of individual cells exhibiting varying propensity for exosome secretion. In cancer, since exosomes can support the development of the pre-metastatic niche, cells with varying abilities to secrete exosomes can directly impact tumorigenesis. Here, we developed a high throughput single-cell technique that enabled the mapping of exosome secretion dynamics. By utilizing clinically relevant models of breast cancer, we established that non-metastatic cancer cells secrete more exosomes than metastatic cancer cells. Single-cell RNA-sequencing confirmed that pathways related to exosome secretion were enriched in the non-metastatic cells compared to the metastatic cells. We established isogenic clonal cell lines from non-metastatic cells with differing propensities for exosome secretion and showed that exosome secretion is an inheritable property preserved during cell division. Combined in vitro and in vivo studies with these cell lines suggested that exosome secretion can impede tumor formation. In human non-metastatic breast tumors, tumors with higher secretion of exosomes have a better prognosis, higher immune cytolytic activity, and enrichment of pro-inflammatory macrophages compared to tumors with lower secretion of exosomes. Our single-cell methodology can become an essential tool that enables the direct integration of exosome secretion with multiple cellular functions.
- Published
- 2020
13. Inhibition of the αv integrin-TGF-β axis improves natural killer cell function against glioblastoma stem cells
- Author
-
Joy Gumin, Mayela Mendt, Jun Yu, Matthias Eyrich, May Daher, Jinzhuang Dou, David Marin, Mecit Kaplan, Sufang Li, Jian Hu, Ana Karen Nunez Cortes, Stephan Mielke, Gonca Ozcan, Nobuhiko Imahashi, Richard E. Champlin, Pinaki P. Banerjee, Junjun Lu, Ken Chen, Elif Gokdemir, Katayoun Rezvani, Amy B. Heimberger, Cynthia Kassab, Enli Liu, Yifei Shen, Dihua Yu, Rafet Basar, Sunil Acharya, Luis Muniz-Feliciano, Giulio Draetta, Elizabeth J. Shpall, Fang Wang, Vakul Mohanty, Li Li, Nadima Uprety, Konrad Gabrusiewicz, Mustafa H Bdiwi, Natalie W. Fowlkes, Qi Miao, Lucila Nassif Kerbauy, Abdullah Alsuliman, Jun Wei, Daniel Zamler, Emily Ensley, Frederick F. Lang, Mayra Hernandez Sanabria, Hila Shaim, and April L. Gilbert
- Subjects
endocrine system ,0303 health sciences ,Cell ,Integrin ,Biology ,In vitro ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Immune system ,Single-cell analysis ,030220 oncology & carcinogenesis ,medicine ,biology.protein ,Cancer research ,Mass cytometry ,Stem cell ,Receptor ,030304 developmental biology - Abstract
Glioblastoma, the most aggressive brain cancer, often recurs because glioblastoma stem cells (GSCs) are resistant to all standard therapies. Here, we show that patient-derived GSCs, but not normal astrocytes, are highly sensitive to lysis by healthy allogeneic natural killer (NK) cellsin vitro. In contrast, single cell analysis of autologous, tissue infiltrating NK cells isolated from surgical samples of high-grade glioblastoma patient tumors using mass cytometry and single cell RNA sequencing revealed an abnormal phenotype associated with impaired lytic function compared with peripheral blood NK cells from GBM patients or healthy donors. This immunosuppression was attributed to an integrin-TGF-β mechanism, activated by direct cell-cell contact between GSCs and NK cells. Treatment of GSC-engrafted mice with allogeneic NK cells in combination with inhibitors of integrin or TGF-β signaling, or withTGF-β receptor 2gene-edited NK cells prevented GSC-induced NK cell dysfunction and tumor growth. Collectively, our findings reveal a novel mechanism of NK cell immune evasion by GSCs and implicate the integrin-TGF-β axis as a useful therapeutic target to eliminate GSCs in this devastating tumor.
- Published
- 2020
14. Osteopontin mediates glioblastoma-associated macrophage infiltration and is a potential therapeutic target
- Author
-
Nannan Kang, Y. Alan Wang, Shouhao Zhou, Zacharia A. Grami, Ling-Yuan Kong, Brett Schrand, Gregory N. Fuller, Yuuri Hashimoto, Amy B. Heimberger, Eli Gilboa, Konrad Gabrusiewicz, Xiaoyang Ling, Jason T. Huse, Anantha Marisetty, Shulin Li, Jun Wei, Hillary G. Caruso, Martina Ott, Roel G.W. Verhaak, and Xingxu Huang
- Subjects
Male ,0301 basic medicine ,Chemokine ,Angiogenesis ,T cell ,Mesenchymal Glioblastoma ,CD8-Positive T-Lymphocytes ,Mice ,03 medical and health sciences ,stomatognathic system ,Glioma ,medicine ,Animals ,Humans ,Receptors, Vitronectin ,Osteopontin ,Mice, Knockout ,Immunity, Cellular ,Innate immune system ,biology ,Brain Neoplasms ,Macrophages ,General Medicine ,Aptamers, Nucleotide ,medicine.disease ,M2 Macrophage ,Immunity, Innate ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,medicine.anatomical_structure ,Gene Knockdown Techniques ,Cancer research ,biology.protein ,Female ,Glioblastoma ,Research Article - Abstract
Glioblastoma is highly enriched with macrophages, and osteopontin (OPN) expression levels correlate with glioma grade and the degree of macrophage infiltration; thus, we studied whether OPN plays a crucial role in immune modulation. Quantitative PCR, immunoblotting, and ELISA were used to determine OPN expression. Knockdown of OPN was achieved using complementary siRNA, shRNA, and CRISPR/Cas9 techniques, followed by a series of in vitro functional migration and immunological assays. OPN gene–deficient mice were used to examine the roles of non-tumor-derived OPN on survival of mice harboring intracranial gliomas. Patients with mesenchymal glioblastoma multiforme (GBM) show high OPN expression, a negative survival prognosticator. OPN is a potent chemokine for macrophages, and its blockade significantly impaired the ability of glioma cells to recruit macrophages. Integrin α(v)β(5) (ITGαvβ5) is highly expressed on glioblastoma-infiltrating macrophages and constitutes a major OPN receptor. OPN maintains the M2 macrophage gene signature and phenotype. Both tumor-derived and host-derived OPN were critical for glioma development. OPN deficiency in either innate immune or glioma cells resulted in a marked reduction in M2 macrophages and elevated T cell effector activity infiltrating the glioma. Furthermore, OPN deficiency in the glioma cells sensitized them to direct CD8(+) T cell cytotoxicity. Systemic administration in mice of 4-1BB–OPN bispecific aptamers was efficacious, increasing median survival time by 68% (P < 0.05). OPN is thus an important chemokine for recruiting macrophages to glioblastoma, mediates crosstalk between tumor cells and the innate immune system, and has the potential to be exploited as a therapeutic target.
- Published
- 2018
15. Germline polymorphisms in myeloid-associated genes are not associated with survival in glioma patients
- Author
-
Melissa L. Bondy, George A. Calin, Cristina Ivan, Spiridon Tsavachidis, Margaret Wrensch, Jun Wei, John K. Wiencke, Helen M. Hansen, Terri Rice, Konrad Gabrusiewicz, Renke Zhou, Georgina Armstrong, Daniel I. Jacobs, Paige M. Bracci, Yanhong Liu, Annette M. Molinaro, and Amy B. Heimberger
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Myeloid ,Adolescent ,Genotype ,Single-nucleotide polymorphism ,Polymorphism, Single Nucleotide ,Article ,Germline ,Young Adult ,03 medical and health sciences ,Glioma ,Internal medicine ,medicine ,Humans ,SNP ,Genetic Predisposition to Disease ,Myeloid Cells ,neoplasms ,Aged ,Microglia ,Brain Neoplasms ,Proportional hazards model ,business.industry ,Cancer ,Middle Aged ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Neurology ,Immunology ,Female ,Neurology (clinical) ,Glioblastoma ,business - Abstract
Immune cells of myeloid origin, including microglia, macrophages, and myeloid-derived suppressor cells adopt immunosuppressive phenotypes that support gliomagenesis. Here, we tested an a priori hypothesis that single nucleotide polymorphisms (SNPs) in genes related to glioma-associated myeloid cell regulation and function are also associated with patient survival after glioma diagnosis. Subjects for this study were 992 glioma patients treated at The University of Texas MD Anderson Cancer Center in Houston, Texas between 1992 and 2008. Haplotype-tagging SNPs in 91 myeloid-associated genes were analyzed for association with survival by Cox regression. Individual SNP- and gene-based tests were performed separately in glioblastoma (WHO grade IV, n = 511) and lower-grade glioma (WHO grade II–III, n = 481) groups. After adjustment for multiple testing, no myeloid-associated gene variants were significantly associated with survival in glioblastoma. Two SNPs, rs147960238 in CD163 (p = 2.2 × 10−5) and rs17138945 in MET (p = 5.6 × 10−5) were significantly associated with survival of patients with lower-grade glioma. However, these associations were not confirmed in an independent analysis of 563 lower-grade glioma cases from the University of California at San Francisco Adult Glioma Study (p = 0.65 and p = 0.41, respectively). The results of this study do not support a role for inherited polymorphisms in myeloid-associated genes in affecting survival of patients diagnosed with glioblastoma or lower-grade glioma.
- Published
- 2017
16. Soluble Tie2 overrides the heightened invasion induced by anti-angiogenesis therapies in gliomas
- Author
-
Marta M. Alonso, Nahir Cortes-Santiago, Frederick Lang, Joy Gumin, Mohammad B. Hossain, Juan Fueyo, W. K. A. Yung, Candelaria Gomez-Manzano, Frank C. Marini, Konrad Gabrusiewicz, and Xuejun Fan
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Bevacizumab ,medicine.medical_treatment ,Mice, Nude ,Angiogenesis Inhibitors ,angiopoietin 2 ,Monocytes ,Angiopoietin-2 ,Mice ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Cell Line, Tumor ,Glioma ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Receptor ,Tumor microenvironment ,biology ,Brain Neoplasms ,business.industry ,Growth factor ,invasion ,medicine.disease ,Receptor, TIE-2 ,Angiopoietin receptor ,Tie2-expressing monocytes ,Chemotaxis, Leukocyte ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,cardiovascular system ,biology.protein ,Immunohistochemistry ,anti-angiogenesis ,Neoplasm Recurrence, Local ,business ,Research Paper ,medicine.drug - Abstract
Glioblastoma recurrence after treatment with the anti-vascular endothelial growth factor (VEGF) agent bevacizumab is characterized by a highly infiltrative and malignant behavior that renders surgical excision and chemotherapy ineffective. Our group has previously reported that Tie2-expressing monocytes (TEMs) are aberrantly present at the tumor/normal brain interface after anti-VEGF therapies and their significant role in the invasive outgrowth of these tumors. Here, we aimed to further understand the mechanisms leading to this pro-invasive tumor microenvironment. Examination of a U87MG xenogeneic glioma model and a GL261 murine syngeneic model showed increased tumor expression of angiopoietin 2 (Ang2), a natural ligand of Tie2, after anti-angiogenesis therapies targeting VEGF or VEGF receptor (VEGFR), as assessed by immunohistochemical analysis, immunofluorescence analysis, and enzyme-linked immunosorbent assays of tumor lysates. Migration and gelatinolytic assays showed that Ang2 acts as both a chemoattractant of TEMs and an enhancing signal for their tumor-remodeling properties. Accordingly, in vivo transduction of Ang2 into intracranial gliomas increased recruitment of TEMs into the tumor. To reduce invasive tumor outgrowth after anti-angiogenesis therapy, we targeted the Ang-Tie2 axis using a Tie2 decoy receptor. Using syngeneic models, we observed that overexpression of soluble Tie2 within the tumor prevented the recruitment of TEMs to the tumor and the development of invasion after anti-angiogenesis treatment. Taken together, these data indicate an active role for the Ang2-Tie2 pathway in invasive glioma recurrence after anti-angiogenesis treatment and provide a rationale for testing the combined targeting of VEGF and Ang-Tie2 pathways in patients with glioblastoma.
- Published
- 2016
17. MiR-138 exerts anti-glioma efficacy by targeting immune checkpoints
- Author
-
Yuuri Hashimoto, Jun Wei, Neal Huang, Willem W. Overwijk, Xiaoyang Ling, Mark R. Gilbert, Ling Yuan Kong, George A. Calin, Shuo Xu, Greg Fuller, Edjah K. Nduom, Konrad Gabrusiewicz, Cristina Ivan, Wei Qiao, Amy B. Heimberger, and Shouhao Zhou
- Subjects
0301 basic medicine ,Cancer Research ,Cell cycle checkpoint ,biology ,FOXP3 ,chemical and pharmacologic phenomena ,Immune checkpoint ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,Oncology ,CTLA-4 ,030220 oncology & carcinogenesis ,Immunology ,biology.protein ,Cancer research ,Cytotoxic T cell ,Neurology (clinical) ,Antibody ,CD8 - Abstract
BACKGROUND Antibody therapeutic targeting of the immune checkpoints cytotoxic T-lymphocyte-associated molecule 4 (CTLA-4) and programmed cell death 1 (PD-1) has demonstrated marked tumor regression in clinical trials. MicroRNAs (miRNAs) can modulate multiple gene transcripts including possibly more than one immune checkpoint and could be exploited as immune therapeutics. METHODS Using online miRNA targeting prediction algorithms, we searched for miRNAs that were predicted to target both PD-1 and CTLA-4. MiR-138 emerged as a leading candidate. The effects of miR-138 on CTLA-4 and PD-1 expression and function in T cells were determined and the therapeutic effect of intravenous administration of miR-138 was investigated in both immune-competent and -incompetent murine models of GL261 glioma. RESULTS Target binding algorithms predicted that miR-138 could bind the 3' untranslated regions of CTLA-4 and PD-1, which was confirmed with luciferase expression assays. Transfection of human CD4+ T cells with miR-138 suppressed expression of CTLA-4, PD-1, and Forkhead box protein 3 (FoxP3) in transfected human CD4+ T cells. In vivo miR-138 treatment of GL261 gliomas in immune-competent mice demonstrated marked tumor regression, a 43% increase in median survival time (P = .011), and an associated decrease in intratumoral FoxP3+ regulatory T cells, CTLA-4, and PD-1 expression. This treatment effect was lost in nude immune-incompetent mice and with depletion of CD4+ or CD8+ T cells, and miR-138 had no suppressive effect on glioma cells when treated directly at physiological in vivo doses. CONCLUSIONS MiR-138 exerts anti-glioma efficacy by targeting immune checkpoints which may have rapid translational potential as a novel immunotherapeutic agent.
- Published
- 2015
18. Abstract 3242: CT-0508 is an anti-HER2 chimeric antigen receptor (CAR) macrophage with targeted anti-tumor activity that promotes a pro-inflammatory solid tumor microenvironment
- Author
-
Sotheavy Chhum, Michael Klichinsky, Nicholas E. Petty, Xueqing Lu, Konrad Gabrusiewicz, Xinhe Shan, Maggie Schmierer, Olga Shestova, Andrew Best, Feng Shen, Yumi Ohtani, Nicholas R. Anderson, Martha Zeeman, and Saar Gill
- Subjects
Cancer Research ,Tumor microenvironment ,Adoptive cell transfer ,T cell ,Antigen presentation ,Biology ,Chimeric antigen receptor ,medicine.anatomical_structure ,Oncology ,Antigen ,medicine ,Cancer research ,Cytotoxic T cell ,Macrophage - Abstract
Despite recent advances in T cell immunotherapy for the treatment of human cancer, metastatic solid tumors remain an intractable challenge. Macrophages are often the most abundant immune cell in the tumor microenvironment (TME) where, as immunosuppressive tumor associated macrophages (TAMs), they participate in disease progression. Currently, most macrophage based immunotherapeutic approaches are focused on the depletion, repolarization, or phagocytic disinhibition of TAMs. We have developed a new paradigm based on the adoptive transfer of genetically engineered CAR macrophages (CAR-M) for the treatment of human cancer. CAR-M can be efficiently produced using the chimeric adenoviral vector Ad5f35. We have previously shown that the primary mechanism of action of CAR-M is antigen dependent phagocytosis, and that a single dose of primary human anti-HER2 CAR-M leads to significantly improved overall survival in multiple solid tumor xenograft models. Given that Ad5f35-transduced anti-HER2 CAR-M (CT-0508) adopt a unique pro-inflammatory M1-like phenotype, we hypothesized that CT-0508 may have the capacity to reprogram the TME toward an activated state. Functional evaluation and transcriptome-wide characterization revealed that CT-0508 maintain a pro-inflammatory phenotype despite challenge with immunosuppressive environments in vitro. By engrafting immunodeficient mice with human hematopoietic cells and human cancer cells we established a novel xenografted human TME model. We demonstrate with single cell resolution that CT-0508 maintain their M1 phenotype within the human TME. Additionally, CT-0508 augmented the human TME by inducing a pro-inflammatory signature in surrounding immune cells, characterized by induction of MHC-II and TNFα. To further investigate the potential of CT-0508 for TME activation, we modeled the interaction of CT-0508 with primary human M2 macrophages, dendritic cells, and T cells in vitro. CT-0508 repolarized bystander M2 macrophages toward a pro-inflammatory phenotype, induced activation and maturation markers on immature dendritic cells, and recruited resting as well as activated T cells in chemotaxis assays. CT-0508 demonstrated enhanced antigen presentation when compared to control human macrophages and cross-presented tumor derived intracellular antigens to CD8 T cells after tumor phagocytosis. Our results show that in addition to direct anti-tumor activity, the anti-HER2 CAR macrophage cell product CT-0508 is capable of promoting a pro-inflammatory tumor microenvironment and has the potential to induce epitope spreading via T cell recruitment and antigen presentation. Citation Format: Michael Klichinsky, Konrad Gabrusiewicz, Nicholas Anderson, Maggie Schmierer, Andrew Best, Martha Zeeman, Sotheavy Chhum, Yumi Ohtani, Olga Shestova, Xueqing Lu, Nicholas Petty, Xinhe Shan, Feng Shen, Saar Gill. CT-0508 is an anti-HER2 chimeric antigen receptor (CAR) macrophage with targeted anti-tumor activity that promotes a pro-inflammatory solid tumor microenvironment [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 3242.
- Published
- 2020
19. Abstract 2193: Small molecule inducible MyD88/CD40 (iMC) in CAR-T cells can repolarize M2 macrophage to an anti-tumor M1 phenotype
- Author
-
J. Henri Bayle, Aaron E. Foster, Konrad Gabrusiewicz, Xiao-Hong Wang, and David M. Spencer
- Subjects
Antitumor activity ,Cancer Research ,CD40 ,Oncology ,biology ,Chemistry ,biology.protein ,Cancer research ,Car t cells ,M2 Macrophage ,Phenotype ,Small molecule - Abstract
Background: Effective chimeric antigen receptor (CAR) T cell therapy against solid tumors must overcome a hostile, tumor microenvironment that includes tumor-associated macrophages (TAM). Pancreatic adenocarcinomas (PDAC) are commonly infiltrated with TAMs polarized to a tumor-promoting M2 phenotype rather than a T cell-stimulatory and tumor-inhibitory M1 phenotype. The GoCAR platform combines an inducible MyD88/CD40 (iMC) costimulation protein with a 1st generation CAR. Our previously published results demonstrated that iMC costimulation, activated by the small molecule dimerizer, rimiducid (Rim), enhanced CAR-T proliferation and anti-tumor efficacy. Here, we examined the extrinsic effects of iMC signaling on CAR-T cell immune-activating ligands, cytokine production and their ability to polarize M2 macrophage to an anti-tumor phenotype. Methods: Macrophages were prepared from peripheral blood monocytes of four or more random blood donors from the Gulf Coast Regional Blood Center (Houston, TX) and differentiated in vitro to an M2 phenotype with TGF-β and IL-10, or an M1 phenotype (as a positive control). GoCAR-T cells targeting prostate stem cell antigen (PSCA) were prepared by retroviral transduction from the autologous donors. To test the effects of iMC activation on macrophage polarization, contact-dependent or independent (i.e., separation of cell populations with transwell inserts) coculture assays were performed with and without activation of iMC with 1 nM Rim and/or surface-bound PSCA. Anti-tumor cytotoxicity was measured by coculture with PSCA+ Panc1-GFP cells. Results: CAR activation by PSCA antigen recognition or iMC activation with Rim decreased CD163, an M2 macrophage marker, and increased CD80, expressed on M1 macrophage. Full activation of the GoCAR-T cells with both Rim and the target antigen fully repolarized M2 macrophage to an M1 marked phenotype (CD163lowCD80high). This repolarization could be directed partially in the absence of cell-cell contact by diffusion of soluble factors through Transwell membranes. M2 macrophages repolarized by conditioning media from activated GoCAR-T cells also exhibited the functionality of M1 macrophages and acquired cytotoxicity against tumor cells. Furthermore, cultures of conditioned macrophages and limiting dilutions of GoCAR-T cells demonstrated a cooperative enhancement of the cytotoxicity of PSCA GoCAR-T cells toward Panc-1 targets. This cooperation was more effective for GoCAR-T cells than CD28- or 4-1BB-enhanced 2nd generation PSCA CAR-T cells. Conclusions: These results predict that GoCAR-T activation with Rim will convert TAMs within a solid tumor microenvironment from T cell inhibitors to tumor-caustic agents. Citation Format: Xiaohong Wang, Konrad Gabrusiewicz, David M. Spencer, Aaron E. Foster, J. Henri Bayle. Small molecule inducible MyD88/CD40 (iMC) in CAR-T cells can repolarize M2 macrophage to an anti-tumor M1 phenotype [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 2193.
- Published
- 2020
20. Abstract 2180: Genetically engineered chimeric antigen receptor (CAR) monocytes demonstrate targeted anti-tumor activity and differentiate into M1-polarized CAR macrophages
- Author
-
Yumi Ohtani, Saar Gill, Daniel Cushing, Konrad Gabrusiewicz, Maggie Schmierer, Andrew Best, Michael Klichinsky, Linara Gabitova, and Martha Zeeman
- Subjects
Cancer Research ,medicine.medical_treatment ,Monocyte ,T cell ,CD14 ,Antigen presentation ,Immunotherapy ,Biology ,Chimeric antigen receptor ,medicine.anatomical_structure ,Oncology ,Cancer immunotherapy ,Cancer research ,medicine ,CD80 - Abstract
Despite rapid advances in cancer immunotherapy, clinical responses in metastatic solid tumors have been limited. Macrophages are the most abundant immune cell in the solid tumor microenvironment (TME) and are primarily recruited as monocytes by TME-derived chemokines. When not under the control of the TME, macrophages are potent immune effector cells capable of phagocytosis, T cell recruitment, and antigen presentation. We have previously demonstrated that CAR macrophages (CAR-M) have potent anti-tumor activity and overcome several of the barriers to success in solid tumor immunotherapy - trafficking, immunosuppression, and antigen heterogeneity. Currently, CAR-M are generated via ex vivo differentiation of peripheral blood monocytes into macrophages prior to genetic manipulation. To more closely recapitulate normal biologic behavior, we attempted to create CAR monocytes that could traffic and differentiate into CAR macrophages upon tumor penetration. Toward that goal, we genetically engineered CD14+ human monocytes without ex vivo differentiation and with minimal cell culture. Using the chimeric adenoviral vector Ad5f35, we engineered human CAR-monocytes targeted against HER2. CAR expression and viability both exceeded 90%. Ad5f35 transduced CAR monocytes survived and maintained CAR expression ex vivo for at least 21 days. CAR monocytes efficiently differentiated into CAR-expressing macrophages when treated with GM-CSF as determined by FACS-based phenotypic characterization and Wright-Giemsa staining. Anti-HER2 CAR monocytes eradicated HER2 expressing tumor cells in a time and dose-dependent manner, and had comparable potency to anti-HER2 CAR-M. Additionally, the CAR monocyte manufacturing process offered the logistical advantage of a short manufacturing process (approximately two days). We have previously demonstrated that CAR-M are polarized toward a pro-inflammatory M1 phenotype after transduction with Ad5f35. Similarly, CAR monocytes demonstrated elevated expression of M1 markers, and intriguingly after differentiation into CAR-expressing macrophages, HLA-DR, CD80, CD86, and other M1 markers remained elevated - suggesting that transduction prior to differentiation does not impact the pro-inflammatory impact of adenoviral vectors on myeloid cells. Taken together, this abstract describes the successful development of CAR-monocytes with the potential for a rapid manufacturing process. In addition to direct anti-tumor activity while in the monocyte phase, CAR monocytes have the capacity to differentiate into CAR macrophages in situ, which are in turn capable of phagocytosis, T cell recruitment, TME activation, and antigen presentation. Given the previously demonstrated pre-clinical efficacy of CT-0508 (an anti-HER2 CAR macrophage), the CAR monocyte platform described herein offers a shortened manufacturing process and a potential advantage in tumor penetration, which will be directly evaluated in upcoming studies. Citation Format: Konrad Gabrusiewicz, Maggie Schmierer, Andrew Best, Martha Zeeman, Yumi Ohtani, Linara Gabitova, Daniel Cushing, Saar Gill, Michael Klichinsky. Genetically engineered chimeric antigen receptor (CAR) monocytes demonstrate targeted anti-tumor activity and differentiate into M1-polarized CAR macrophages [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 2180.
- Published
- 2020
21. Abstract B65: CT-0508, a novel CAR macrophage product directed against HER2, promotes a proinflammatory tumor microenvironment
- Author
-
Nicholas R. Anderson, Martha Zeeman, Maggie Schmierer, Andrew Best, Saar Gill, Yumi Ohtani, Nicholas E. Petty, Katherine D. Cummins, Olga Shestova, Feng Shen, Xueqing Lu, Konrad Gabrusiewicz, Michael Klichinsky, and Xinhe Shan
- Subjects
Cancer Research ,Adoptive cell transfer ,Tumor microenvironment ,medicine.medical_treatment ,Immunology ,Antigen presentation ,Immunotherapy ,Dendritic cell ,Biology ,Chimeric antigen receptor ,Proinflammatory cytokine ,Immune system ,medicine ,Cancer research - Abstract
Despite recent advances in T cell immunotherapy for the treatment of human cancer, metastatic solid tumors remain an intractable challenge. Macrophages are usually the most abundant immune cell in the tumor microenvironment (TME) where, as immunosuppressive tumor-associated macrophages (TAMs), they participate in disease progression. The current goals of macrophage-based immunotherapies are to reduce TAM infiltration or enhance TAM phagocytosis. In contrast, we have developed a new paradigm based on the adoptive transfer of genetically engineered chimeric antigen receptor (CAR) macrophages (CAR-M) for the treatment of human cancer. CAR-M can only be produced using a unique adenoviral vector, since human macrophages are highly resistant to other methods of gene transfer. We have previously shown that the primary mechanism of action of CAR-M is phagocytosis, and that a single dose of primary human anti-HER2 CAR-M led to significantly improved overall survival in multiple xenograft models. We now establish that Ad5f35-transduced anti-HER2 CAR-M (CT-0508) adopt a unique proinflammatory and antitumor M1 phenotype. Functional evaluation and RNA sequencing revealed that CT-0508 maintain a proinflammatory M1 phenotype despite challenge with immunosuppressive environments in vitro, highlighting their resistance to subversion. By engrafting immunodeficient mice with human hematopoietic cells and human cancer cells, we established a novel xenografted human TME model. We demonstrate with single-cell resolution that CT-0508 maintain their phenotype within the human TME. Additionally, CT-0508 activated the human TME and generated an activated human dendritic cell signature. To further investigate the potential of CT-0508 for TME activation, we modeled the interaction of CT-0508 with immunosuppressive macrophages, dendritic cells, and T cells. CT-0508 shifted bystander macrophages toward a proinflammatory phenotype, induced activation and maturation markers on DCs, and recruited resting as well as activated T cells in chemotaxis assays. Lastly, CT-0508 demonstrated enhanced antigen presentation when compared to control human macrophages. These results show that in addition to direct antitumor activity, the anti-HER2 CAR macrophage cell product CT-0508 is capable of activating the solid cancer TME and promoting a proinflammatory phenotype. The safety of CT-0508 will be evaluated in an upcoming first-in-human phase I clinical trial. Citation Format: Konrad Gabrusiewicz, Nicholas Anderson, Xueqing Lu, Xinhe Shan, Olga Shestova, Nicholas Petty, Feng Shen, Maggie Schmierer, Andrew Best, Martha Zeeman, Yumi Ohtani, Katherine Cummins, Saar Gill, Michael Klichinsky. CT-0508, a novel CAR macrophage product directed against HER2, promotes a proinflammatory tumor microenvironment [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2019 Nov 17-20; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2020;8(3 Suppl):Abstract nr B65.
- Published
- 2020
22. Glioblastoma stem cell-derived exosomes induce M2 macrophages and PD-L1 expression on human monocytes
- Author
-
Shouhao Zhou, Anwar Hossain, Johnny C. Akers, Yuzaburo Shimizu, Erik P. Sulman, Laurence J.N. Cooper, Shulin Li, Jason T. Huse, Ganesh Rao, M. Anna Zal, Joy Gumin, Changiz Geula, Jack P. Antel, Anantha Marisetty, Raghu Kalluri, Aras Rezavanian, Luke M. Healy, Gregory N. Fuller, Jun Wei, Fei Wang, Sourindra Maiti, Tomasz Zal, Felix Nwajei, Amy B. Heimberger, Kenneth Dunner, Shinji Yamashita, Martina Ott, Yuuri Hashimoto, Jared K. Burks, Raymond Sawaya, Clark C. Chen, David H. Hawke, John Yu, Konrad Gabrusiewicz, and Xu Li
- Subjects
0301 basic medicine ,cancer stem cells ,lcsh:Immunologic diseases. Allergy ,CD14 ,Immunology ,Exosome ,lcsh:RC254-282 ,stat3 ,03 medical and health sciences ,immune cells ,pd-l1 ,PD-L1 ,medicine ,Immunology and Allergy ,exosome ,Original Research ,biology ,Chemistry ,Monocyte ,glioblastoma ,Actin cytoskeleton ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Microvesicles ,3. Good health ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,biology.protein ,STAT protein ,Stem cell ,lcsh:RC581-607 - Abstract
Exosomes can mediate a dynamic method of communication between malignancies, including those sequestered in the central nervous system and the immune system. We sought to determine whether exosomes from glioblastoma (GBM)-derived stem cells (GSCs) can induce immunosuppression. We report that GSC-derived exosomes (GDEs) have a predilection for monocytes, the precursor to macrophages. The GDEs traverse the monocyte cytoplasm, cause a reorganization of the actin cytoskeleton, and skew monocytes toward the immune suppresive M2 phenotype, including programmed death-ligand 1 (PD-L1) expression. Mass spectrometry analysis demonstrated that the GDEs contain a variety of components, including members of the signal transducer and activator of transcription 3 (STAT3) pathway that functionally mediate this immune suppressive switch. Western blot analysis revealed that upregulation of PD-L1 in GSC exosome-treated monocytes and GBM-patient-infiltrating CD14+ cells predominantly correlates with increased phosphorylation of STAT3, and in some cases, with phosphorylated p70S6 kinase and Erk1/2. Cumulatively, these data indicate that GDEs are secreted GBM-released factors that are potent modulators of the GBM-associated immunosuppressive microenvironment.
- Published
- 2018
23. Down-regulation of IKKβ expression in glioma-infiltrating microglia/macrophages is associated with defective inflammatory/immune gene responses in glioblastoma
- Author
-
Tomasz Tykocki, Marta Maleszewska, Marcin Roszkowski, Wenson D. Rajan, Wiesława Grajkowska, Bozena Kaminska, Konrad Gabrusiewicz, Malgorzata Sielska, Marta Kocyk, Paweł Nauman, Katarzyna Kotulska, Dominika Pszczolkowska, Bogusław Kostkiewicz, Jakub Mieczkowski, and Piotr Przanowski
- Subjects
Male ,global gene expression profiling ,Angiogenesis ,medicine.medical_treatment ,Down-Regulation ,Biology ,IKKβ/NFkB signaling ,Immune system ,Glioma ,medicine ,Animals ,Humans ,glioma-associated microglia/macrophages ,Rats, Wistar ,immunosuppression ,Microglia ,Brain Neoplasms ,Gene Expression Profiling ,Macrophages ,glioblastoma ,Immunosuppression ,medicine.disease ,Molecular medicine ,nervous system diseases ,I-kappa B Kinase ,Rats ,Gene expression profiling ,medicine.anatomical_structure ,Oncology ,Immunology ,Signal transduction ,Research Paper - Abstract
// Jakub Mieczkowski 1, * , Marta Kocyk 1, 2, * , Pawel Nauman 3 , Konrad Gabrusiewicz 1 , Malgorzata Sielska 1 , Piotr Przanowski 1 , Marta Maleszewska 1 , Wenson D. Rajan 1 , Dominika Pszczolkowska 1 , Tomasz Tykocki 3 , Wieslawa Grajkowska 4 , Katarzyna Kotulska 5 , Marcin Roszkowski 6 , Boguslaw Kostkiewicz 7 , Bozena Kaminska 1 1 Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland 2 Postgraduate School of Molecular Medicine, Medical University of Warsaw, Poland 3 Department of Neurosurgery, Institute Psychiatry and Neurology, Warsaw, Poland 4 Departments of Pathology, The Children's Memorial Health Institute, Warsaw, Poland 5 Neurology, The Children's Memorial Health Institute, Warsaw, Poland 6 Neurosurgery, The Children's Memorial Health Institute, Warsaw, Poland 7 Central Clinical Hospital Ministry of Interior, Warsaw, Poland * These authors have contributed equally to this work Correspondence to: Bozena Kaminska, e-mail: bozenakk@nencki.gov.pl Keywords: immunosuppression, glioma-associated microglia/macrophages, global gene expression profiling, glioblastoma, IKKβ/NFkB signaling Received: April 03, 2015 Accepted: September 17, 2015 Published: September 29, 2015 ABSTRACT Glioblastoma (GBM) is an aggressive malignancy associated with profound host immunosuppression. Microglia and macrophages infiltrating GBM acquire the pro-tumorigenic, M2 phenotype and support tumor invasion, proliferation, survival, angiogenesis and block immune responses both locally and systematically. Mechanisms responsible for immunological deficits in GBM patients are poorly understood. We analyzed immune/inflammatory gene expression in five datasets of low and high grade gliomas, and performed Gene Ontology and signaling pathway analyses to identify defective transcriptional responses. The expression of many immune/inflammatory response and TLR signaling pathway genes was reduced in high grade gliomas compared to low grade gliomas. In particular, we found the reduced expression of the IKBKB, a gene coding for IKKβ, which phosphorylates IκB proteins and represents a convergence point for most signal transduction pathways leading to NFκB activation. The reduced IKBKB expression and IKKβ levels in GBM tissues were demonstrated by qPCR, Western blotting and immunohistochemistry. The IKKβ expression was down-regulated in microglia/macrophages infiltrating glioblastoma. NFκB activation, prominent in microglia/macrophages infiltrating low grade gliomas, was reduced in microglia/macrophages in glioblastoma tissues. Down-regulation of IKBKB expression and NFκB signaling in microglia/macrophages infiltrating glioblastoma correlates with defective expression of immune/inflammatory genes and M2 polarization that may result in the global impairment of anti-tumor immune responses in glioblastoma.
- Published
- 2015
24. PD-L1 expression and prognostic impact in glioblastoma
- Author
-
Jie Qing Chen, Charles A. Conrad, Xiaoyang Ling, Neal Huang, Jun Wei, Ling Yuan Kong, Shouhao Zhou, Gregory N. Fuller, Caitlin Creasy, Jared K. Burks, Konrad Gabrusiewicz, Amy B. Heimberger, Cristina Ivan, Edjah K. Nduom, Laszlo Radvanyi, Nasser K. Yaghi, Krit Ritthipichai, and George A. Calin
- Subjects
Male ,0301 basic medicine ,Oncology ,Cancer Research ,Pathology ,T-Lymphocytes ,B7-H1 Antigen ,Immunoenzyme Techniques ,Mice ,0302 clinical medicine ,biology ,medicine.diagnostic_test ,Brain Neoplasms ,Middle Aged ,Flow Cytometry ,Prognosis ,Gene Expression Regulation, Neoplastic ,Survival Rate ,030220 oncology & carcinogenesis ,Basic and Translational Investigations ,Immunohistochemistry ,Female ,Adult ,medicine.medical_specialty ,Flow cytometry ,03 medical and health sciences ,Cancer stem cell ,Cell Line, Tumor ,Internal medicine ,PD-L1 ,Biomarkers, Tumor ,medicine ,Animals ,Humans ,RNA, Messenger ,Survival rate ,Aged ,Cell Proliferation ,Neoplasm Staging ,Messenger RNA ,Cell growth ,business.industry ,030104 developmental biology ,biology.protein ,Neurology (clinical) ,Glioblastoma ,business ,Ex vivo ,Follow-Up Studies - Abstract
Therapeutic targeting of the immune checkpoints cytotoxic T-lymphocyte-associated molecule-4 (CTLA-4) and PD-1/PD-L1 has demonstrated tumor regression in clinical trials, and phase 2 trials are ongoing in glioblastoma (GBM). Previous reports have suggested that responses are more frequent in patients with tumors that express PD-L1; however, this has been disputed. At issue is the validation of PD-L1 biomarker assays and prognostic impact.Using immunohistochemical analysis, we measured the incidence of PD-L1 expression in 94 patients with GBM. We categorized our results according to the total number of PD-L1-expressing cells within the GBMs and then validated this finding in ex vivo GBM flow cytometry with further analysis of the T cell populations. We then evaluated the association between PD-L1 expression and median survival time using the protein expression datasets and mRNA from The Cancer Genome Atlas.The median percentage of PD-L1-expressing cells in GBM by cell surface staining is 2.77% (range: 0%-86.6%; n = 92), which is similar to the percentage found by ex vivo flow cytometry. The majority of GBM patients (61%) had tumors with at least 1% or more PD-L1-positive cells, and 38% had at least 5% or greater PD-L1 expression. PD-L1 is commonly expressed on the GBM-infiltrating T cells. Expression of both PD-L1 and PD-1 are negative prognosticators for GBM outcome.The incidence of PD-L1 expression in GBM patients is frequent but is confined to a minority subpopulation, similar to other malignancies that have been profiled for PD-L1 expression. Higher expression of PD-L1 is correlated with worse outcome.
- Published
- 2015
25. TMIC-22. DECIPHERING GLIOMA INTRINSIC TRANSCRIPTIONAL SUBTYPES IDENTIFIES TUMOR EVOLUTION ASSOCIATES WITH CHANGES IN IMMUNE-MICROENVIRONMENT
- Author
-
Jian Hu, Joanna J. Phillips, Qianghu Wang, Floris P. Barthel, Amy B. Heimberger, Yan Li, Siyuan Zheng, Konrad Gabrusiewicz, Yu-Hsi Lin, Baoli Hu, Nikuj Satani, Erik P. Sulman, Pengping Li, Florian L. Muller, Ana C. deCarvalho, Guocan Wang, Gaetano Finocchiaro, Xin Hu, Sali Lyu, Massimo Squatrito, Edward F. Chang, Adriana Olar, Do-Hyun Nam, Zhengdao Lan, Tom Mikkelsen, Emmanuel Martinez-Ledesma, Roel G.W. Verhaak, Ronald A. DePinho, Eskil Eskilsson, Mitchel S. Berger, Hee Jin Cho, and Lisa Scarpace
- Subjects
0301 basic medicine ,03 medical and health sciences ,Cancer Research ,Abstracts ,030104 developmental biology ,Oncology ,Glioma ,Immune microenvironment ,Cancer research ,medicine ,Neurology (clinical) ,Biology ,medicine.disease - Abstract
Glioblastoma expression subtypes have been previously been associated with genomic abnormalities, treatment response, and differences in tumor microenvironment. We leveraged IDH wild-type glioblastomas, derivative neurospheres, and single cell gene expression profiles to define three tumor-intrinsic transcriptional subtypes designated as proneural, mesenchymal, and classical, a revision of the previously reported TCGA subtypes. Transcriptomic subtype multiplicity correlated with increased intratumoral heterogeneity and the presence of tumor microenvironment. In silico cell sorting identified macrophages/microglia, CD4+ T lymphocytes, and neutrophils in the glioma microenvironment. NF1 deficiency resulted in increased tumor-associated macrophages/microglia infiltration. Comparison of matching primary and recurrent gliomas elucidated treatment-induced phenotypic tumor evolution, including expression subtype switching, in 45% of our cohort as well as associations between microenvironmental components and treatment response. Gene signature-based tumor microenvironment inference revealed a decrease in invading monocytes and a subtype-dependent increase in macrophages/microglia cells upon disease recurrence. Hypermutation at diagnosis or at recurrence was associated with CD8+ T cell enrichment. Frequency of M2 macrophage detection was associated with short-term relapse after radiation therapy. Our study provides a comprehensive transcriptional and cellular landscape of IDH wild-type glioblastoma during treatment modulated tumor evolution. Characterization of the evolving glioblastoma transcriptome and tumor microenvironment aids in designing more effective immunotherapy trials.
- Published
- 2017
26. Cell surface vimentin-targeted monoclonal antibody 86C increases sensitivity to temozolomide in glioma stem cells
- Author
-
Shulin Li, Ling Yuan Kong, Qingnan Zhao, Sungguan Hong, Hyangsoon Noh, Amy B. Heimberger, Jun Yan, Konrad Gabrusiewicz, and Xueqing Xia
- Subjects
0301 basic medicine ,Cancer Research ,Programmed cell death ,endocrine system ,medicine.drug_class ,Cell Survival ,medicine.medical_treatment ,Brain tumor ,Monoclonal antibody ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Glioma ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Temozolomide ,Animals ,Humans ,Vimentin ,Cell Proliferation ,Chemotherapy ,business.industry ,Brain Neoplasms ,fungi ,Antibodies, Monoclonal ,Drug Synergism ,medicine.disease ,Xenograft Model Antitumor Assays ,030104 developmental biology ,Oncology ,Apoptosis ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer research ,Neoplastic Stem Cells ,Stem cell ,business ,Glioblastoma ,medicine.drug - Abstract
Glioblastoma multiforme (GBM) is the most prevalent and aggressive brain tumor. The current standard therapy, which includes radiation and chemotherapy, is frequently ineffective partially because of drug resistance and poor penetration of the blood-brain barrier. Reducing resistance and increasing sensitivity to chemotherapy may improve outcomes. Glioma stem cells (GSCs) are a source of relapse and chemoresistance in GBM; sensitization of GSCs to temozoliomide (TMZ), the primary chemotherapeutic agent used to treat GBM, is therefore integral for therapeutic efficacy. We previously discovered a unique tumor-specific target, cell surface vimentin (CSV), on patient-derived GSCs. In this study, we found that the anti-CSV monoclonal antibody 86C efficiently increased GSC sensitivity to TMZ. The combination TMZ+86C induced significantly greater antitumor effects than TMZ alone in eight of 12 GSC lines. TMZ+86C–sensitive GSCs had higher CSV expression overall and faster CSV resurfacing among CSV(−) GSCs compared with TMZ+86C–resistant GSCs. Finally, TMZ+86C increased apoptosis of tumor cells and prolonged survival compared with either drug alone in GBM mouse models. The combination of TMZ+86C represents a promising strategy to reverse GSC chemoresistance.
- Published
- 2017
27. Tumor Evolution of Glioma-Intrinsic Gene Expression Subtypes Associates with Immunological Changes in the Microenvironment
- Author
-
Baoli Hu, Massimo Squatrito, Nikunj Satani, Eskil Eskilsson, Jian Hu, Charles Etienne Gabriel Sauvé, Mitchel S. Berger, Emmanuel Martinez-Ledesma, Roel G.W. Verhaak, Hoon Kim, Amy B. Heimberger, Yan Li, Siyuan Zheng, Konrad Gabrusiewicz, Do Hyun Nam, Joanna J. Phillips, Ronald A. DePinho, Hee Jin Cho, Floris P. Barthel, Edward I. Chang, Gaetano Finocchiaro, Florian L. Muller, Zheng D. Lan, Adriana Olar, Qianghu Wang, Ana C. deCarvalho, Pengping Li, Erik P. Sulman, Tom Mikkelsen, Guocan Wang, Sali Lyu, Yu Hsi Lin, Lisa Scarpace, Xin Hu, and Neurosurgery
- Subjects
0301 basic medicine ,Cancer Research ,tumor evolution ,T-Lymphocytes ,0302 clinical medicine ,Recurrence ,Gene expression ,Tumor Microenvironment ,2.1 Biological and endogenous factors ,Aetiology ,Cancer ,Regulation of gene expression ,Microglia ,mesenchymal subtype ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,Phenotype ,Oncology ,030220 oncology & carcinogenesis ,T cell ,Oncology and Carcinogenesis ,Biology ,Article ,03 medical and health sciences ,disease recurrence ,immune cells ,Rare Diseases ,Glioma ,medicine ,Genetics ,Humans ,Oncology & Carcinogenesis ,macrophages/microglia ,Tumor microenvironment ,Neoplastic ,Gene Expression Profiling ,glioblastoma ,Neurosciences ,Cell Biology ,Gene signature ,medicine.disease ,Survival Analysis ,Brain Disorders ,Gene expression profiling ,proneural to mesenchymal transition ,Brain Cancer ,030104 developmental biology ,Gene Expression Regulation ,Cancer cell ,Cancer research ,Glioblastoma ,CD8 - Abstract
We leveraged IDH wild-type glioblastomas, derivative neurospheres, and single-cell gene expression profiles to define three tumor-intrinsic transcriptional subtypes designated as proneural, mesenchymal, and classical. Transcriptomic subtype multiplicity correlated with increased intratumoral heterogeneity and presence of tumor microenvironment. In silico cell sorting identified macrophages/microglia, CD4+ T lymphocytes, and neutrophils in the glioma microenvironment. NF1 deficiency resulted in increased tumor-associated macrophages/microglia infiltration. Longitudinal transcriptome analysis showed that expression subtype is retained in 55% of cases. Gene signature-based tumor microenvironment inference revealed a decrease in invading monocytes and a subtype-dependent increase in macrophages/microglia cells upon disease recurrence. Hypermutation at diagnosis or at recurrence associated with CD8+ Tcell enrichment. Frequency of M2 macrophages detection associated with short-term relapse after radiation therapy.
- Published
- 2017
28. The Role of Myeloid-Derived Suppressor Cells in Immunosuppression in Brain Tumors
- Author
-
A. B. Heimberger, N. A. Colwell, and Konrad Gabrusiewicz
- Subjects
0301 basic medicine ,education.field_of_study ,Tumor microenvironment ,Myeloid ,Microglia ,T cell ,Cell ,Population ,Biology ,law.invention ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,law ,030220 oncology & carcinogenesis ,Immunology ,medicine ,Myeloid-derived Suppressor Cell ,Suppressor ,education - Abstract
Myeloid-derived suppressor cells (MDSCs) represent a heterogeneous population of immature myeloid cells with impaired ability to differentiate into macrophages, granulocytes, or dendritic cells. MDSCs have been identified in peripheral blood and tumor microenvironment of glioma-bearing mice and glioblastoma (GBM) patients. MDSCs together with M2-polarized microglia/macrophages play a crucial role in suppression of innate and adaptive antitumor immunity. MDSCs utilize multiple of mechanisms to suppress T cell function; however their interactions with other myeloid cell have not yet been fully elucidated. In this chapter, we describe the origin of the myeloid population, the mechanism of activation and expansion of MDSCs, suppressive mechanisms mediated by MDSCs, and the role of MDSCs in GBM development, with a primary focus on potential therapeutic strategies targeting MDSCs in GBM.
- Published
- 2017
29. Anti-vascular endothelial growth factor therapy-induced glioma invasion is associated with accumulation of Tie2-expressing monocytes
- Author
-
Kenneth Aldape, Dan Liu, Frank C. Marini, Mark R. Gilbert, Candelaria Gomez-Manzano, Konrad Gabrusiewicz, Nahir Cortes-Santiago, Miguel Angel Idoate, Charles A. Conrad, Gregory N. Fuller, Marta M. Alonso, Juan Fueyo, and Mohammad B. Hossain
- Subjects
Vascular Endothelial Growth Factor A ,Oncology ,Pathology ,Myeloid ,Fluorescent Antibody Technique ,Angiogenesis Inhibitors ,Monocytes ,Mice ,0302 clinical medicine ,0303 health sciences ,education.field_of_study ,Microscopy, Confocal ,Brain Neoplasms ,Glioma ,Flow Cytometry ,Immunohistochemistry ,Receptor, TIE-2 ,3. Good health ,Bevacizumab ,Vascular endothelial growth factor A ,Tie2 ,medicine.anatomical_structure ,Matrix Metalloproteinase 9 ,030220 oncology & carcinogenesis ,monocyte ,Monoclonal ,Research Paper ,medicine.drug ,medicine.medical_specialty ,Population ,Mice, Nude ,Enzyme-Linked Immunosorbent Assay ,Biology ,Antibodies, Monoclonal, Humanized ,Brain tumors ,03 medical and health sciences ,Internal medicine ,medicine ,tumor microenvironment ,Animals ,Humans ,Neoplasm Invasiveness ,education ,030304 developmental biology ,Tumor microenvironment ,Cancer ,medicine.disease ,Xenograft Model Antitumor Assays ,Drug Resistance, Neoplasm ,Neoplasm Recurrence, Local ,antiangiogenesis - Abstract
// Konrad Gabrusiewicz 1,* , Dan Liu 1,* , Nahir Cortes-Santiago 1,* , Mohammad B. Hossain 1 , Charles A. Conrad 1 , Kenneth D. Aldape 2 , Gregory N. Fuller 2 , Frank C. Marini 3 , Marta M. Alonso 4 , Miguel Angel Idoate 5 , Mark R. Gilbert 1 , Juan Fueyo 1 , and Candelaria Gomez-Manzano 1,6 1 Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA 2 Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA 3 Institute of Regenerative Medicine, Wake Forest University, Winston-Salem, North Carolina, USA 4 Department of Medical Oncology, University Hospital of Navarra, Pamplona, Spain 5 Department of Pathology, University Hospital of Navarra, Pamplona, Spain 6 Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA * contributed equally to the work Correspondence: Candelaria Gomez-Manzano, email: // Keywords : Brain tumors, antiangiogenesis, Tie2, tumor microenvironment, monocyte Received : January 6, 2014 Accepted : April 11, 2014 Published : April 11, 2014 Abstract The addition of anti-angiogenic therapy to the few treatments available to patients with malignant gliomas was based on the fact that these tumors are highly vascularized and on encouraging results from preclinical and clinical studies. However, tumors that initially respond to this therapy invariably recur with the acquisition of a highly aggressive and invasive phenotype. Although several myeloid populations have been associated to this pattern of recurrence, a specific targetable population has not been yet identified. Here, we present evidence for the accumulation of Tie2-expressing monocytes/macrophages (TEMs) at the tumor/normal brain interface of mice treated with anti-VEGF therapies in regions with heightened tumoral invasion. Furthermore, we describe the presence of TEMs in malignant glioma surgical specimens that recurred after bevacizumab treatment. Our studies showed that TEMs enhanced the invasive properties of glioma cells and secreted high levels of gelatinase enzymatic proteins. Accordingly, Tie2 + MMP9 + monocytic cells were consistently detected in the invasive tumor edge upon anti-VEGF therapies. Our results suggest the presence of a specific myeloid/monocytic subpopulation that plays a pivotal role in the mechanism of escape of malignant gliomas from anti-VEGF therapies and therefore constitutes a new cellular target for combination therapies in patients selected for anti-angiogenesis treatment.
- Published
- 2014
30. TMIC-26. MiR-181a CONTROLS THE OSTEOPONTIN-MEDIATED IMMUNE CIRCUIT IN GLIOBLASTOMA
- Author
-
Konrad Gabrusiewicz, Ling Kong, Amy B. Heimberger, Yuuri Hashimoto, Jun Wei, Martina Ott, and Anantha Marisetty
- Subjects
Cancer Research ,biology ,business.industry ,medicine.disease ,Abstracts ,Immune system ,Oncology ,biology.protein ,Cancer research ,Medicine ,Neurology (clinical) ,Osteopontin ,business ,Glioblastoma - Abstract
INTRODUCTION: MiRNAs can silence a broad gene set, which may benefit heterogeneous tumors such as glioblastoma. Osteopontin has been shown to have an oncogenic role in a variety of cancers and may have immune modulatory effects on macrophages. The current study focuses on using miRNAs to target osteopontin in tumor cells and to modulate immune cells to elicit an antitumor effect. METHODS: Genome-wide profiling followed by evaluation of expression levels of miRNAs and osteopontin were measured using quantitative real time PCR and ELISA in mouse and human glioblastoma and macrophages. Luciferase assays were used to determine the binding potential of miRNAs to osteopontin mRNA. miRNA mimics and forced overexpression using lentiviruses were used to target osteopontin in immune competent murine models of glioblastoma. Nano string profiling and Gene Set Enrichment Analysis (GSEA) were conducted to identify differences in biological states. RESULTS: Microarray analysis demonstrated that osteopontin was the most significantly upregulated gene in human glioblastoma-associated infiltrating macrophages that had originated from matched circulating monocytes, and this was validated using qPCR. Based on the 3`UTR sequence of osteopontin, bioinformatics tools identified 5 miRNAs that are conserved and would be able to modulate osteopontin expression. Luciferase assays confirmed that the miR-181 family regulates osteopontin expression. Overexpression of miR-181a in glioblastoma cells led to their decreased proliferation and increased apoptosis in vitro. miR-181a treatment of immune competent mice bearing intracranial glioblastoma demonstrated a 22% increase in median survival time relative to control mice (p=0.006). CONCLUSIONS: miR-181a controls osteopontin expression in both glioblastoma and macrophages by regulating their proliferation and apoptosis.
- Published
- 2018
31. Abstract 2907: Exosome secretion is an inheritable property of cancer cells: Single-cell profiling of exosome secretion
- Author
-
Konrad Gabrusiewicz, Ankit Mahendra, Xingyue An, Melisa Martinez-Paniagua, Robiya Joseph, Navin Varadarajan, Sendurai A. Mani, Jay R Adolacion, Mohsen Fathi, and Sujash S. Chatterjee
- Subjects
Cancer Research ,Cell ,Biology ,Exosome ,In vitro ,Microvesicles ,medicine.anatomical_structure ,Immune system ,Oncology ,Cell culture ,Cancer cell ,medicine ,Cancer research ,Secretion - Abstract
Decades of research on exosomes, nano-sized vesicles secreted by cells, have revealed novel roles of these vesicles in the formation of pre-metastasis niches that enhances the migration of tumor cells to those sites. Paradoxically, more recent work has suggested that these tumor-derived exosomes can also have an immunostimulatory role, depending on the model studied. The rate of secretion of exosomes by single cells is likely heterogeneous, and a deep profiling of the secretion capacity of individual tumor cells has been largely unexplored. We have developed a high-throughput single-cell methodology to quantify the dynamic secretion of exosomes from single cells using a modified immunoassay and sought to define: (a) heterogeneity among individual cells within the tumor cell population, and (b) the nature of the cells secreting exosomes within a tumor cell population. In order to investigate these, we chose to study models of triple negative breast cancer: the metastatic tumor lines, 4T1 (mouse) and MDA-MB-231(human); and the non-metastatic lines 67NR (mouse) and MCF7 (human). Our method revealed that MDA-MB-231 single cells secreted exosomes at a rate, ~2 fold higher than MCF7 cells. Surprisingly, the non-metastatic 67NR cells showed a higher secretion rate (~1.5 fold) than metastatic 4T1 cells. Next, we performed single-cell RNA-seq on 67NR and 4T1 single cells. Consistent with our single-cell exosomal profiling results, 67NR cells were significantly (p-value < 0.01) enriched for genes correlated to exosome formation in comparison to the 4T1 cells. Next, in order to study exosomes of single cells, we isolated single cells using an automated micromanipulator which allowed us to establish monoclonal cell lines. Measurement of these monoclonal cells showed that secretor cells of 67NR secrete exosomes with ~2 fold higher rate than non-secretor cells. Since our in vitro results clearly demonstrated that the secretor clonal cell lines had a higher frequency of exosome secreting single cells, we sought to define the in vivo relevance of these results. Consistent with the hypothesis that the exosomes from 67NR are immune-stimulatory, injection of the secretor clones into Balb/c mice led to lack of primary tumor formation (2/15 mice had tumors) whereas the injection of the non-secretor clones led to tumor formation in 7/15 mice. In aggregate, our results show that the higher rate of secretion of exosomes from non-metastatic cells can facilitate tumor rejection in vivo. We are currently performing in vitro studies with the 67NR and MDA-MB-231 exosomes to study their impact on immune cells. Citation Format: Mohsen Fathi, Robiya Joseph, Melisa Martinez-Paniagua, Jay R Adolacion, Xingyue An, Ankit Mahendra, Konrad Gabrusiewicz, Sujash Chatterjee, Sendurai A. Mani, Navin Varadarajan. Exosome secretion is an inheritable property of cancer cells: Single-cell profiling of exosome secretion [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 2907.
- Published
- 2019
32. Knockout immune regulator FGL2 in tumor cells impairs tumor progression in the CNS by facilitating CD103+ dendritic cell differentiation
- Author
-
Jun Yan, Qingnan Zhao, Konrad Gabrusiewicz, Ling-Yuan Kong, Xueqing Xia, Jian Wang, Martina Ott, Jingda Xu, Eric Richard Davis, Longfei Huo, Ganesh Rao, Shao-Cong Sun, Stephanie S Watowich, Amy B. Heimberger, and Shulin Li
- Subjects
Immunology ,Immunology and Allergy - Abstract
Although many studies link brain tumor progression to oncogene activation and tumor-suppressor gene inactivation in tumor cells, few studies implicate immune regulatory gene expression in tumor cells in arbitrating brain tumor progression. Here we show that fibrinogen-like protein 2 (FGL2) is highly expressed in glioma stem cells and primary glioblastoma (GBM) cells. FGL2 knockout (FGL2KO) in GL261, DBT, and LLC tumor cells did not affect tumor cell proliferation in vitro or tumor progression in immunodeficient NSG mice, but completely impaired GBM progression in immune-competent C57bl/6 mice. This impairment was reversed in mice with a defect in Batf3 (a key transcription factor for CD103+ DCs differentiation). Mechanistic studies revealed that FGL2KO in tumor cells induces CD103+ DCs differentiation in both the central nervous system (CNS) and in tumor draining lymph nodes (TDLN). The increased CD103+ DCs population in the CNS and TDLNs induce CD8+ T cells priming and activation and thereby gliomas regress. More specifically, the presence of FGL2 in tumor cells inhibited granulocyte-macrophage colony-stimulating factor (GM-CSF)–induced CD103+ DC differentiation by suppressing NF-κB, STAT1/5, and p38 activation. These findings are relevant to GBM patients because a low level of FGL2 expression with concurrent high GM-CSF expression is associated with higher CD8B expression and longer survival. These data provide a rationale for therapeutic inhibition of FGL2 in brain tumors.
- Published
- 2019
33. Author Correction: FGL2 promotes tumor progression in the CNS by suppressing CD103+ dendritic cell differentiation
- Author
-
Qingnan Zhao, Stephanie S. Watowich, R. Eric Davis, Jian Wang, Ling Yuan Kong, Jingda Xu, Xueqing Xia, Amy B. Heimberger, Shulin Li, Shao Cong Sun, Longfei Huo, Jun Yan, Ganesh Rao, Martina Ott, and Konrad Gabrusiewicz
- Subjects
0301 basic medicine ,Multidisciplinary ,business.industry ,Science ,General Physics and Astronomy ,02 engineering and technology ,General Chemistry ,Dendritic cell differentiation ,021001 nanoscience & nanotechnology ,General Biochemistry, Genetics and Molecular Biology ,FGL2 ,03 medical and health sciences ,030104 developmental biology ,Tumor progression ,Cancer research ,Medicine ,lcsh:Q ,0210 nano-technology ,business ,lcsh:Science - Abstract
The original version of this Article contained errors in the author affiliations. Qingnan Zhao, Xueqing Xia, Longfei Huo and Shulin Li were incorrectly associated with Beijing Institute for Brain Disorders, 100069, Beijing, China.This has now been corrected in both the PDF and HTML versions of the Article.
- Published
- 2019
34. Distinct roles of CSF family cytokines in macrophage infiltration and activation in glioma progression and injury response
- Author
-
Malgorzata Zawadzka, Katarzyna Kotulska, Konrad Gabrusiewicz, Marta Maleszewska, Piotr Przanowski, Bartosz Wylot, Joanna Kucharska, Helmut Kettenmann, Katyayni Vinnakota, Wiesława Grajkowska, Malgorzata Sielska, Bozena Kaminska, and Magdalena Kijewska
- Subjects
Macrophage colony-stimulating factor ,Regulation of gene expression ,Gene knockdown ,Pathology ,medicine.medical_specialty ,Microglia ,Angiogenesis ,Biology ,medicine.disease ,Colony-stimulating factor ,Pathology and Forensic Medicine ,medicine.anatomical_structure ,Cell culture ,Glioma ,Cancer research ,medicine - Abstract
Gliomas attract brain-resident (microglia) and peripheral macrophages and reprogram these cells into immunosuppressive, pro-invasive cells. M-CSF (macrophage colony-stimulating factor, encoded by the CSF1 gene) has been implicated in the control of recruitment and polarization of macrophages in several cancers. We found that murine GL261 glioma cells overexpress GM-CSF (granulocyte-macrophage colony-stimulating factor encoded by the CSF2 gene) but not M-CSF when compared to normal astrocytes. Knockdown of GM-CSF in GL261 glioma cells strongly reduced microglia-dependent invasion in organotypical brain slices and growth of intracranial gliomas and extended animal survival. The number of infiltrating microglia/macrophages (Iba1(+) cells) and intratumoural angiogenesis were reduced in murine gliomas depleted of GM-CSF. M1/M2 gene profiling in sorted microglia/macrophages suggests impairment of their pro-invasive activation in GM-CSF-depleted gliomas. Deficiency of M-CSF (op/op mice) did not affect glioma growth in vivo and the accumulation of Iba1(+) cells, but impaired accumulation of Iba1(+) cells in response to demyelination. These results suggest that distinct cytokines of the CSF family contribute to macrophage infiltration of tumours and in response to injury. The expression of CSF2 (but not CSF1) was highly up-regulated in glioblastoma patients and we found an inverse correlation between CSF2 expression and patient survival. Therefore we propose that GM-CSF triggers and drives the alternative activation of tumour-infiltrating microglia/macrophages in which these cells support tumour growth and angiogenesis and shape the immune microenvironment of gliomas.
- Published
- 2013
35. TIE2-mediated tyrosine phosphorylation of H4 regulates DNA damage response by recruiting ABL1
- Author
-
Candelaria Gomez-Manzano, Nahir Cortes-Santiago, Zhimin Lu, Xuemei Luo, Shawn S.-C. Li, Frederick F. Lang, David G. Johnson, Mohammad B. Hossain, Mien Chie Hung, Rehnuma Shifat, Juan Fueyo, Konrad Gabrusiewicz, Ravesanker Ezhilarasan, Hong Jiang, Jessica K. Tyler, Mark T. Bedford, and Erik P. Sulman
- Subjects
0301 basic medicine ,DNA End-Joining Repair ,DNA damage ,DNA repair ,oncogenes ,Genotoxic Stress ,Biology ,Proto-Oncogene Mas ,Radiation Tolerance ,Receptor tyrosine kinase ,Histones ,03 medical and health sciences ,chemistry.chemical_compound ,Cell Line, Tumor ,Radiation, Ionizing ,Humans ,Proto-Oncogene Proteins c-abl ,Research Articles ,NHEJ ,Multidisciplinary ,SciAdv r-articles ,Tyrosine phosphorylation ,Cell Biology ,DNA repair protein XRCC4 ,Receptor, TIE-2 ,3. Good health ,Proliferating cell nuclear antigen ,ANG1 ,Protein Transport ,030104 developmental biology ,Histone ,TIE2 ,chemistry ,biology.protein ,Cancer research ,Tyrosine ,Angiotensin I ,Research Article ,ABL1 ,DNA Damage ,Protein Binding - Abstract
Membrane-bound enzyme relocates to the cell nucleus to modify chromatin, inducing cancer resistance to radiotherapy., DNA repair pathways enable cancer cells to survive DNA damage induced after genotoxic therapies. Tyrosine kinase receptors (TKRs) have been reported as regulators of the DNA repair machinery. TIE2 is a TKR overexpressed in human gliomas at levels that correlate with the degree of increasing malignancy. Following ionizing radiation, TIE2 translocates to the nucleus, conferring cells with an enhanced nonhomologous end-joining mechanism of DNA repair that results in a radioresistant phenotype. Nuclear TIE2 binds to key components of DNA repair and phosphorylates H4 at tyrosine 51, which, in turn, is recognized by the proto-oncogene ABL1, indicating a role for nuclear TIE2 as a sensor for genotoxic stress by action as a histone modifier. H4Y51 constitutes the first tyrosine phosphorylation of core histones recognized by ABL1, defining this histone modification as a direct signal to couple genotoxic stress with the DNA repair machinery.
- Published
- 2016
36. Glioblastoma-infiltrated innate immune cells resemble M0 macrophage phenotype
- Author
-
Sujit S. Prabhu, Jeffrey S. Weinberg, Nasser K. Yaghi, Ganesh Rao, Ahmed Elakkad, Luke M. Healy, Qianghu Wang, Krishna P. Bhat, Ravesanker Ezhilarasan, Rivka R. Colen, Jun Wei, Benjamin Rodriguez, Gregory N. Fuller, Konrad Gabrusiewicz, Lauren A. Langford, Yuuri Hashimoto, Sourindra Maiti, Brandon D. Liebelt, Wei Li, Laurence J.N. Cooper, Shouhao Zhou, Raymond Sawaya, Amit Bar-Or, Ginu Thomas, Erik P. Sulman, Michael A. Curran, Amy B. Heimberger, Janet M. Bruner, Neal Huang, and Jack P. Antel
- Subjects
0301 basic medicine ,Cell biology ,Innate immune system ,medicine.diagnostic_test ,Microglia ,Microarray analysis techniques ,lcsh:R ,lcsh:Medicine ,General Medicine ,Biology ,Phenotype ,3. Good health ,Flow cytometry ,Gene expression profiling ,03 medical and health sciences ,030104 developmental biology ,Immune system ,medicine.anatomical_structure ,Integrin alpha M ,Immunology ,Cancer research ,medicine ,biology.protein ,Research Article - Abstract
Glioblastomas are highly infiltrated by diverse immune cells, including microglia, macrophages, and myeloid-derived suppressor cells (MDSCs). Understanding the mechanisms by which glioblastoma-associated myeloid cells (GAMs) undergo metamorphosis into tumor-supportive cells, characterizing the heterogeneity of immune cell phenotypes within glioblastoma subtypes, and discovering new targets can help the design of new efficient immunotherapies. In this study, we performed a comprehensive battery of immune phenotyping, whole-genome microarray analysis, and microRNA expression profiling of GAMs with matched blood monocytes, healthy donor monocytes, normal brain microglia, nonpolarized M0 macrophages, and polarized M1, M2a, M2c macrophages. Glioblastoma patients had an elevated number of monocytes relative to healthy donors. Among CD11b+ cells, microglia and MDSCs constituted a higher percentage of GAMs than did macrophages. GAM profiling using flow cytometry studies revealed a continuum between the M1- and M2-like phenotype. Contrary to current dogma, GAMs exhibited distinct immunological functions, with the former aligned close to nonpolarized M0 macrophages.
- Published
- 2016
37. Tipping a favorable CNS intratumoral immune response using immune stimulation combined with inhibition of tumor-mediated immune suppression
- Author
-
Gregory N. Fuller, Shouhao Zhou, Brett Schrand, Eli Gilboa, Amy B. Heimberger, Ganesh Rao, Jun Wei, George A. Calin, Konrad Gabrusiewicz, and Ling Yuan Kong
- Subjects
0301 basic medicine ,Effector ,animal diseases ,Brief Report ,Immunology ,Lymphokine ,CCL18 ,chemical and pharmacologic phenomena ,Biology ,biochemical phenomena, metabolism, and nutrition ,03 medical and health sciences ,030104 developmental biology ,Immune system ,Oncology ,Antigen ,STAT protein ,biology.protein ,Cancer research ,Immunology and Allergy ,Macrophage ,bacteria ,STAT3 - Abstract
High-grade gliomas are notoriously heterogeneous regarding antigen expression, effector responses, and immunosuppressive mechanisms. Therefore, combinational immune therapeutic approaches are more likely to impact a greater number of patients and result in longer, durable responses. We have previously demonstrated the monotherapeutic effects of miR-124, which inhibits the signal transducer and activator of transcription 3 (STAT3) immune suppressive pathway, and immune stimulatory 4-1BB aptamers against a variety of malignancies, including genetically engineered immune competent high-grade gliomas. To evaluate potential synergy, we tested an immune stimulatory aptamer together with microRNA-124 (miRNA-124), which blocks tumor-mediated immune suppression, and found survival to be markedly enhanced, including beyond that produced by monotherapy. The synergistic activity appeared to be not only secondary to enhanced CD3(+) cell numbers but also to reduced macrophage immune tumor trafficking, indicating that a greater therapeutic benefit can be achieved with approaches that both induce immune activation and inhibit tumor-mediated immune suppression within the central nervous system (CNS) tumors.
- Published
- 2015
38. Silencing of cellular prion protein (PrPC) expression by DNA-antisense oligonucleotides induces autophagy-dependent cell death in glioma cells
- Author
-
Elena Sbalchiero, Alessandro Spedito, Luigi Pirtoli, Silvia Palumbo, Sergio Comincini, Clelia Miracco, Marco Biggiogera, Giulia Barbieri, Giuliano Mazzini, Alberto Azzalin, Konrad Gabrusiewicz, Bozena Kaminska, and Nicoletta Marchesi
- Subjects
Programmed cell death ,Cell Survival ,animal diseases ,Central nervous system ,Down-Regulation ,Apoptosis ,Ubiquitin-Activating Enzymes ,Biology ,Autophagy-Related Protein 7 ,Mice ,chemistry.chemical_compound ,Downregulation and upregulation ,Cell Line, Tumor ,Glioma ,mental disorders ,Autophagy ,medicine ,Carcinoma ,Animals ,Humans ,Gene silencing ,PrPC Proteins ,Gene Silencing ,RNA, Messenger ,Cell Shape ,Molecular Biology ,Cell Proliferation ,Membrane Proteins ,DNA ,Cell Biology ,Oligonucleotides, Antisense ,medicine.disease ,Rats ,nervous system diseases ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,chemistry ,Cytoprotection ,Cancer research ,Beclin-1 ,Apoptosis Regulatory Proteins - Abstract
Malignant gliomas are the most common and lethal primary central nervous system neoplasms. Several intriguing lines of evidence have recently emerged indicating that the cellular prion protein (PrPC) may exert neuro- and cyto-protective functions: PrPC overexpression protects cultured neurons and also tumor cell lines exposed to various pro-apoptotic stimuli while, on the contrary, PrPC silencing sensitizes Adriamycin-resistant human breast carcinoma cells to TRAIL-mediated cell death. In order to determine if PrPC is involved in the resistance of glial tumors to cell death, the effects of cellular prion protein downregulation by antisense approach were investigated in different human malignant glioma cell lines. PrPC downregulation induced profound morphological changes and significant cell death. In addition, a significant tumor volume reduction was noted after PrPC silencing in a EGFP-GL261 glioma murine model. Investigations of the molecular effects induced by PrPC silencing were carried out on T98G human glioma cells by analysing autophagic as well as typical apoptotic markers (nuclear morphology, caspase-3/7, p53 and PARP-1). The results indicated that apoptosis was not induced after PrPC downregulation while, on the contrary, electron microscopy analysis, and an accumulation of GFP-LC3-II in autophagosomal membranes of GFP-LC3 transfected cells, indicated a predominant activation of autophagy. PrPC silencing also led to induction of LC3-II, increase in Beclin-1 and a concomitant decrease in p62, Bcl-2 and in the phosphorylation of 4E-BP1, a target of mTOR autophagy signaling. In conclusion, our results show for the first time that interfering with the cellular prion protein expression could modulate autophagy-dependent cell death pathways in glial tumor cells.
- Published
- 2011
39. The Antitumorigenic Response of Neural Precursors Depends on Subventricular Proliferation and Age
- Author
-
Wolfgang Uckert, Konrad Gabrusiewicz, Leszek Kaczmarek, Gerd Kempermann, Kazuaki Yoshikawa, Michael Synowitz, Boris Engels, Evgeni Nikolaev, Helmut Kettenmann, Rainer Glass, Darko Markovic, Joo Hee Walzlein, and Bozena Kaminska
- Subjects
Angiogenesis ,Subventricular zone ,Biology ,Mice ,Cyclin D1 ,Cyclin D2 ,Cyclins ,Precursor cell ,otorhinolaryngologic diseases ,medicine ,Animals ,Cells, Cultured ,Cell Proliferation ,Cyclin ,Neurons ,Tumor microenvironment ,Brain Neoplasms ,Cell growth ,Stem Cells ,Age Factors ,Cell Biology ,Mice, Inbred C57BL ,stomatognathic diseases ,medicine.anatomical_structure ,Immunology ,Cancer research ,Molecular Medicine ,Glioblastoma ,Stem Cell Transplantation ,Developmental Biology - Abstract
Glioblastomas, the most aggressive primary brain tumors, occur almost exclusively in adult patients. Neural precursor cells (NPCs) are antitumorigenic in mice, as they can migrate to glioblastomas and induce tumor cell death. Here, we show that the antitumor effect of NPCs is age-dependently controlled by cell proliferation in the subventricular zone (SVZ) and that NPCs accumulating at a glioblastoma are diverted from their normal migratory path to the olfactory bulb. Experimentally induced cortical glioblastomas resulted in decreased subventricular proliferation in adult (postnatal day 90) but not in young (postnatal day 30) mice. Adult mice supplied fewer NPCs to glioblastomas and had larger tumors than young mice. Apart from the difference in proliferation, there was neither a change in cell number and death rate in the SVZ nor a change in angiogenesis and immune cell density in the tumors. The ability to kill glioblastomas was similar in NPCs isolated from young and adult mice. The proliferative response of NPCs to glioblastomas depended on the expression of D-type cyclins. In young mice, NPCs express the cyclins D1 and D2, but the expression of cyclin D1 is lost during aging, and in adult NPCs only cyclin D2 remains. In young and adult cyclin D2-deficient mice we observed a reduced supply of NPCs to glioblastomas and the generation of larger tumors compared with wild-type mice. We conclude that cyclin D1 and D2 are nonredundant for the antitumor response of subventricular NPCs. Loss of a single D-type cyclin results in a smaller pool of proliferating NPCs, lower number of NPCs migrating to the tumor, and reduced antitumor activity. Disclosure of potential conflicts of interest is found at the end of this article.
- Published
- 2008
40. The invasion promoting effect of microglia on glioblastoma cells is inhibited by cyclosporin A
- Author
-
Aleksandra Wesolowska, Michael Synowitz, Helmut Kettenmann, Konrad Gabrusiewicz, Rainer Glass, Darko Markovic, Malgorzata Zawadzka, Bozena Kaminska, and Marcin Sliwa
- Subjects
Male ,Pathology ,medicine.medical_specialty ,Antineoplastic Agents ,Biology ,Tissue Culture Techniques ,Mice ,Cell Movement ,In vivo ,Cyclosporin a ,Glioma ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,Rats, Wistar ,Matrigel ,Microglia ,Brain Neoplasms ,medicine.disease ,In vitro ,Rats ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Mice, Inbred DBA ,Cyclosporine ,Cancer research ,Neuroglia ,Neurology (clinical) ,Signal transduction ,Glioblastoma ,Cell Division ,Neoplasm Transplantation ,Signal Transduction - Abstract
The invasion of tumour cells into brain tissue is a pathologic hallmark of WHO grades II-IV gliomas and contributes significantly to the failure of current therapeutic treatments. Activated microglial cells are abundant in brain tumours and may support tumour invasiveness. We have previously demonstrated that cyclosporin A (CsA) can affect growth of glioma cells in vitro by inhibiting signalling pathways, which are essential for tumour proliferation and invasiveness. In this work, we demonstrate that migration of EGFP-transfected glioblastoma cells in organotypic brain slices was significantly inhibited by treatment with CsA. On average 77% of untreated cells migrated beyond 500 mum, while only 28-33% cells migrated as far in the brain slices treated with CsA (P < 0.001). This inhibitory effect on glioblastoma invasion was lost when glioblastoma cells were injected into microglia-depleted brain slices. Moreover, CsA significantly inhibits intracranial glioma growth in vivo. We demonstrate that microglia-derived factors increase glioma invasiveness in Matrigel assay in vitro and this is associated with activation of the PI-3K/Akt signalling pathway. The invasion promoting effect of microglia is abolished in the presence of CsA. Furthermore, glioma-derived soluble factors induce morphological transformation of microglia and activate MAPK signalling, although production of pro-inflammatory factors was not observed. Our findings that CsA interferes at clinically relevant concentrations with the tumour-promoting role of microglia and impairs invasive growth of glioma cells in vivo may provide a novel therapeutic strategy against gliomas.
- Published
- 2007
41. TMIC-10PLEIOTROPY OF TUMOR-ASSOCIATED MYELOID CELLS WITHIN HUMAN GLIOBLASTOMA
- Author
-
Frederick Lang, Yuuri Hashimoto, Shouhao Zhou, Jack P. Antel, Maiti Sourindra, Ganesh Rao, Konrad Gabrusiewicz, Jun Wei, Luke M. Healy, Raymond Sawaya, Benjamin Rodriguez, Amy B. Heimberger, Raversanker Ezhilarasan, E.P. Sulman, Brandon D. Liebelt, Laurence J.N. Cooper, Sujit S. Prabhu, Neal Huang, Jefffrey Weinberg, and Roeland Verhaak
- Subjects
Cancer Research ,business.industry ,Biology ,medicine.disease ,Text mining ,Oncology ,Pleiotropism ,Myeloid cells ,Cancer research ,medicine ,Neurology (clinical) ,business ,Abstracts from the 20th Annual Scientific Meeting of the Society for Neuro-Oncology ,Glioblastoma - Published
- 2015
42. IMPS-28PD-L1 EXPRESSION AND PROGNOSTIC IMPACT IN GLIOBLASTOMA
- Author
-
Greg Fuller, Ling-Yuan Kong, Shouhao Zhou, Nasser K. Yaghi, Amy B. Heimberger, Neal Huang, Konrad Gabrusiewicz, Jared K. Burks, Xiaoyang Ling, George A. Calin, Cristina Ivan, Jie Qing Chen, Caitlin Creasy, Laszlo Radvanyi, Charles A. Conrad, Jun Wei, Edjah K. Nduom, and Krit Ritthipichai
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,biology ,medicine.diagnostic_test ,CD3 ,T cell ,Flow cytometry ,Immune system ,medicine.anatomical_structure ,Oncology ,Cancer research ,biology.protein ,medicine ,Granzyme A ,Immunohistochemistry ,Neurology (clinical) ,Abstracts from the 20th Annual Scientific Meeting of the Society for Neuro-Oncology ,CD8 ,Ex vivo - Abstract
BACKGROUND: Therapeutic targeting of the immune checkpoints CTLA-4 and PD-1/PD-L1 have demonstrated tumor regression in clinical trials, and phase II trials are ongoing in glioblastoma. Previous reports have suggested that responses are more frequent in patients with tumors expressing PD-L1; however, this has been disputed. At issue is the validation of PD-L1 biomarker assays and prognostic impact. METHODS: Using immunohistochemical analysis, we measured the incidence of PD-L1 expression in 94 patients with glioblastomas. We determined the total number of PD-L1-expressing cells within the glioblastomas and validated this finding using ex vivo glioblastoma flow cytometry, with further analysis of T cell populations. We evaluated the association between PD-L1 expression and median survival time using the protein expression data sets and mRNA from The Cancer Genome Atlas (TCGA). RESULTS: The median percentage of PD-L1-expressing cells in glioblastoma by cell surface staining is 2.77% (range 0-86.6%, n = 92), similar to the percentage found by ex vivo flow cytometry. The majority of glioblastoma patients (61%) had tumors with at least 1% PD-L1-positive cells, and 38% had at least 5% PD-L1 expression. PD-L1 and PD-1 are commonly expressed on glioblastoma-infiltrating T cells. Expression of PD-L1 is prognostic by multivariate analysis for a negative glioblastoma outcome by both immunohistochemistry (p = 0.018) and mRNA expression analysis of TCGA data (p = 0.009). Expression of PD-1 mRNA is correlated with perforin 1 (P 0.0001), both markers of cytolytic activity, as well as the T cell markers CD3 (P < 0.0001), CD4 (P < 0.0001) and CD8 (P < 0.0001). PD-L1 mRNA expression is correlated with granzyme A (P < 0.0001), CD3 (P = 0.0099) and CD4 (P < 0.001). CONCLUSIONS: The incidence of PD-L1 expression in glioblastoma patients is frequent but is confined to a minority subpopulation-similar to other malignancies that have been profiled for PD-L1 expression. Higher expression of PD-L1 is correlated with worse outcome.
- Published
- 2015
43. IMPS-22FGL2 AS A MULTI-MODALITY REGULATOR OF TUMOR-MEDIATED IMMUNE SUPPRESSION
- Author
-
Shulin Li, Xueqing Xia, Ling-Yuan Kong, Amy B. Heimberger, Jun Yan, Konrad Gabrusiewicz, Denada Dibra, and Jiemiao Hu
- Subjects
Therapeutic immunosuppression ,Cancer Research ,Immune system ,Oncology ,Immunology ,Regulator ,Neurology (clinical) ,Biology ,Abstracts from the 20th Annual Scientific Meeting of the Society for Neuro-Oncology ,Multi modality - Published
- 2015
44. TMIC-09GLIOBLASTOMA STEM CELL-DERIVED EXOSOMES PROMOTE M2 POLARIZATION OF HUMAN MONOCYTES
- Author
-
Tomasz Zal, David H. Hawke, Felix Nwajei, Shouhao Zhou, Joy Gumin, Maiti Sourindra, Amy B. Heimberger, Xu Li, Yuuri Hashimoto, Shinji Yamashita, Frederick Lang, Jun Wei, Laurence J.N. Cooper, Konrad Gabrusiewicz, Anna Zal, and John Yu
- Subjects
Cancer Research ,medicine.diagnostic_test ,Monocyte ,CD14 ,medicine.medical_treatment ,Biology ,Peripheral blood mononuclear cell ,Microvesicles ,Flow cytometry ,Cell biology ,medicine.anatomical_structure ,Cytokine ,Oncology ,Cancer cell ,medicine ,Neurology (clinical) ,Stem cell ,Abstracts from the 20th Annual Scientific Meeting of the Society for Neuro-Oncology - Abstract
INTRODUCTION: Exosomes secreted by cancer cells have pleiotropic functions, and can promote autocrine signaling to distant cells. Elucidating the mechanistic modulation of the immune system by these exosomes provides insight into potential biomarkers for detection, recurrence, and response, and identifies potential new therapeutic targets. METHODS: Exosomes were isolated from human glioblastoma stem cells (GSC) and fibroblasts (control) using differential centrifugation. Fluorescent-labeled exosomes were co-cultured with human peripheral blood mononuclear cells (PBMCs). Flow cytometry and confocal microscopy methods were utilized to determine the ability of immune cells to uptake exosomes and to evaluate subsequent intracellular trafficking. The exosomal protein and RNA content was analyzed by mass spectrometry and Nanostring Counter System, respectively. The phenotypic and functional skewing of the monocyte lineage was analyzed after exposing these cells to exosomes. The cytokine array was used to analyze the cytokines generated following treatment of human normal monocytes with exosomes. RESULTS: The GSC-secreted exosomes were preferentially absorbed by CD14+ monocytes and Gr-1+ derived myeloid cells isolated from healthy volunteers and/or glioblastoma patients. When activated, CD4+ and CD8+ T cells could also uptake GSC-secreted exosomes. Confocal microscopy revealed that only monocytes could internalize GSC-secreted exosomes but not fibroblast-secreted exosomes. The exposure to GSC-secreted exosomes induces a phenotypic change in monocytes and prevents them from undergoing apoptosis. GSC-secreted exosomes, but not the fibroblast-secreted exosomes, increased expression of CD163, CD206, and decreased expression of MHC class II. Monocytes treated with GSC-secreted exosomes release IL-6, IL-1RA, CCL3, and CCL4 when compared to cells exposed to fibroblast exosomes. GSC-secreted exosomes contained distinct protein composition in contrast to fibroblast-secreted exosomes that may affect the anti-tumor function of monocyte-derived macrophages. CONCLUSIONS: Monocytes demonstrated a preferential uptake of GSC-secreted exosomes which then induced a glioma-supportive M2 phenotype. GSC-secreted exosomes can be a contributing factor in the M2 skewing within glioma microenvironment.
- Published
- 2015
45. FGL2 as a Multimodality Regulator of Tumor-Mediated Immune Suppression and Therapeutic Target in Gliomas
- Author
-
Denada Dibra, Shulin Li, Jiemiao Hu, Konrad Gabrusiewicz, Ling Yuan Kong, Jun Yan, Amy B. Heimberger, and Xueqing Xia
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_treatment ,Regulator ,Mice, SCID ,Biology ,T-Lymphocytes, Regulatory ,Article ,Mice ,Immune system ,Mice, Inbred NOD ,Cell Line, Tumor ,Glioma ,Biomarkers, Tumor ,medicine ,Animals ,Humans ,Immunologic Factors ,RNA, Messenger ,Aged ,Immunosuppression Therapy ,Regulation of gene expression ,Analysis of Variance ,Mice, Inbred BALB C ,Brain Neoplasms ,Fibrinogen ,Cancer ,Immunosuppression ,Middle Aged ,medicine.disease ,Survival Analysis ,nervous system diseases ,Gene Expression Regulation, Neoplastic ,Mice, Inbred C57BL ,Oncology ,Immunology ,Myeloid-derived Suppressor Cell ,Cancer research ,biology.protein ,Female ,Antibody ,Glioblastoma - Abstract
Fibrinogen-like protein 2 (FGL2) may promote glioblastoma multiforme (GBM) cancer development by inducing multiple immune-suppression mechanisms.The biological significance of FGL2 expression was assessed using the The Cancer Genome Atlast (TCGA) glioma database and tumor lysates analysis. The therapeutic effects of an anti-Fgl2 antibody and the role of immune suppression regulation by Fgl2 were determined in immune-competent, NOD-scid IL2Rgammanull (NSG), and FcɣRIIB-/- mice (n = 3-18 per group). Data were analyzed with two-way analysis of variance, log-rank survival analysis, and Pearson correlation. All statistical tests were two-sided.In low-grade gliomas, 72.5% of patients maintained two copies of the FGL2 gene, whereas 83.8% of GBM patients had gene amplification or copy gain. Patients with high levels of FGL2 mRNA in glioma tissues had a lower overall survival (P = .009). Protein levels of FGL2 in GBM lysates were higher relative to low-grade glioma lysates (11.48±5.75ng/mg vs 3.96±1.01ng/mg, P = .003). In GL261 mice treated with an anti-FGL2 antibody, median survival was 27 days compared with only 17 days for mice treated with an isotype control antibody (P = .01). The anti-FGL2 antibody treatment reduced CD39(+) Tregs, M2 macrophages, programmed cell death protein 1 (PD-1), and myeloid-derived suppressor cells (MDSCs). FGL2-induced increases in M2, CD39, and PD-1 were ablated in FcɣRIIB-/- mice.FGL2 augments glioma immunosuppression by increasing the expression levels of PD-1 and CD39, expanding the frequency of tumor-supportive M2 macrophages via the FcγRIIB pathway, and enhancing the number of MDSCs and CD39(+) regulatory T cells. Collectively, these results show that FGL2 functions as a key immune-suppressive modulator and has potential as an immunotherapeutic target for treating GBM.
- Published
- 2015
46. TMIC-13. ELUCIDATION OF MicroRNA-OSTEOPONTIN CIRCUIT IN GLIOBLASTOMA ASSOCIATED INFILTRATING MACROPHAGES
- Author
-
Yuuri Hashimoto, Anantha Marisetty, Amy B. Heimberger, Konrad Gabrusiewicz, Ling-Yuan Kong, Jun Wei, and Martina Ott
- Subjects
0301 basic medicine ,Cancer Research ,Tumor microenvironment ,Cell cycle checkpoint ,biology ,business.industry ,Cancer ,medicine.disease ,law.invention ,Abstracts ,03 medical and health sciences ,030104 developmental biology ,Text mining ,Oncology ,law ,microRNA ,biology.protein ,medicine ,Cancer research ,Neurology (clinical) ,Osteopontin ,business ,Polymerase chain reaction ,Glioblastoma - Published
- 2017
47. Abstract 2259: Evaluation of polymorphisms in myeloid-associated genes and glioma survival
- Author
-
Cristina Ivan, Paige M. Bracci, Konrad Gabrusiewicz, Anna M. Dahlin, E. Susan Amirian, Beatrice Melin, Melissa L. Bondy, Helen M. Hansen, Renke Zhou, Jun Wei, Amy B. Heimberger, Terri Rice, Spiridon Tsavachidis, Margaret Wrensch, George A. Calin, Daniel I. Jacobs, John K. Wiencke, Yanhong Liu, Georgina Armstrong, and Michael E. Scheurer
- Subjects
Cancer Research ,Myeloid ,medicine.anatomical_structure ,Oncology ,Glioma ,medicine ,Cancer research ,Biology ,medicine.disease ,Gene - Abstract
BACKGROUND: Gliomas are highly infiltrated by immune cells including microglia, macrophages, and myeloid-derived suppressor cells (collectively, glioma-associated myeloid cells). These cells have been shown to be induced by the tumor to be immune-suppressive and tumor-supportive, and are a negative prognosticator for survival in mouse models. Here, we examine whether inherited variants in genes important to the function of glioma-associated myeloid cells are associated with survival following low-grade glioma diagnosis. METHODS: Subjects for this study were 484 patients with WHO grade II or grade III glioma treated at The University of Texas MD Anderson Cancer Center in Houston, Texas between 1992 and 2008 and followed up for survival through August, 2016. We selected 100 genes for analysis including transcription factors, cytokines and chemokines, receptors, enzymes, and other genes central to the function of glioma-associated myeloid cells. Genotyping was originally performed using the Illumina Human 610-Quad Bead Chip platform and 2,040 tagging SNPs as determined by Haploview Tagger software were selected for analysis with minor allele frequency (AF) ≥ 1%. Associations between selected SNPs and survival were evaluated by Cox regression analysis under an additive allelic model adjusting for age, sex, extent of surgery (biopsy only/partial resection/gross total resection), radiotherapy (yes/no), and chemotherapy (yes/no). Models were examined to ensure that proportional hazards assumptions were not violated. RESULTS: Median survival among low-grade glioma patients was 6.7 years. Age at diagnosis, extent of surgery, and having received radiotherapy or chemotherapy were each significantly associated with survival. Five SNPs were associated with survival at a significance level of p CONCLUSIONS: Here we provide preliminary evidence of an association between polymorphisms in two genes related to glioma-associated myeloid cell function and low-grade glioma survival. CD163 is a receptor that is highly expressed on macrophages and may play a role in macrophage-mediated anti-inflammatory responses, while MET is a receptor tyrosine kinase and well-studied proto-oncogene that is also involved in the expansion of myeloid-derived suppressor cell populations. Further investigation of these associations is warranted, and validation of these findings is planned in an independent population. Citation Format: Daniel I. Jacobs, Yanhong Liu, Konrad Gabrusiewicz, Spiridon Tsavachidis, E. Susan Amirian, Georgina N. Armstrong, Renke Zhou, Jun Wei, Cristina Ivan, George Calin, Michael Scheurer, Anna Dahlin, Terri Rice, Paige M. Bracci, Helen M. Hansen, John K. Wiencke, Margaret R. Wrensch, Beatrice Melin, Amy B. Heimberger, Melissa L. Bondy. Evaluation of polymorphisms in myeloid-associated genes and glioma survival [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 2259. doi:10.1158/1538-7445.AM2017-2259
- Published
- 2017
48. Abstract 2949: TGF-β is a key mediator of NK cell dysfunction in gliolastoma
- Author
-
Pinaki P. Banerjee, Richard E. Champlin, Hila Shaim, Enli Liu, Lucila Nassif Kerbauy, Elizabeth J. Shpall, Abdullah Alsuliman, John S. Yu, Jun Wei, Li Li, Mayela Mendt, Amy B. Heimberger, Katayoun Rezvani, May Daher, David Marin, Konrad Gabrusiewicz, Young Jin Gi, Muharrem Muftuoglu, Rafet Basar, Sonny Ang, and Nobuhiko Imahashi
- Subjects
Cancer Research ,Cell ,Biology ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Mediator ,Oncology ,Immunology ,medicine ,Key (cryptography) ,Cancer research ,030215 immunology ,Transforming growth factor - Abstract
Background: NK cells play a crucial role in the antitumor immune response and are involved in controlling tumor formation, progression and metastases. Glioblastoma (GBM) is the most devastating brain tumor, associated with very poor prognosis. While immunotherapy emerged as a promising approach for anti cancer therapy, GBM appears immune resistant. Here, we studied the role of NK cells in targeting glioblastoma and possible mechanisms of NK immune evasion. Methods: NK cells were isolated from freshly resected GBM tissue, matching peripheral blood and the blood of healthy controls. Flow cytometry was used to characterize the cells and their ability to produce cytokines, and chromium release assay was perfumed to assess their cytotoxicity. In vitro assays were performed by co-culturing GBM stem cells (GSCs) with healthy donor NK cells. Results: We found that whereas NK cells were abundant in primary GBM tissue and could efficiently target GBM stem cells (GSCs), GBM infiltrating NK cells (TiNKs) displayed an abnormal phenotype with downregulation of many activating receptors including CD16, NKG2D, DNAM, NKp30, NKp46, 2B4 and NKG2C and upregulation of inhibitory proteins such as PD-1. This inhibitory phenotype was associated with impaired NK cell function when compared with NK cells isolated from the peripheral blood of patients and healthy donors. GSC-NK cell-cell contact resulted in release of TGF-β by GCSs, which in turn led to NK dysfunction through constitutive activating of the p-Smad pathway. TGF-β activation, in turn, is partially mediated by the matrix metalloproteases MMP-2 and MMP-9, secreted by GSCs upon contact with NK cells and enhanced upon TGF-β exposure. We demonstrated that inhibition of the TGF-β axis, in particular by the small molecule inhibitor, galunisertib, can prevent GSC-induced NK cell dysfunction but is unable to inactivate the p-Smad pathway, thus, cannot reverse existing dysfunction. Conclusions: Our results indicate that NK-GBM cross-talk plays an important role in tumor escape and highlight the importance of developing future adoptive transfer therapies with the intent of limiting tumor escape from antitumor immunity. Note: This abstract was not presented at the meeting. Citation Format: Hila Shaim, Abdullah Alsuliman, Konrad Gabrusiewicz, Jun Wei, John Yu, Rafet Basar, May Daher, Lucila Kerbauy, Mayela Mendt, Muharrem Muftuoglu, Li Li, Enli Liu, Nobuhiko Imahashi, Sonny Ang, Young Gi, Pinaki Banerjee, David Marin, Richard Champlin, Elizabeth Shpall, Amy Heimberger, Katayoun Rezvani. TGF-β is a key mediator of NK cell dysfunction in gliolastoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 2949. doi:10.1158/1538-7445.AM2017-2949
- Published
- 2017
49. Abstract 3869: Cell surface vimentin targeted mAb 86C increases sensitivity to temozolomide mediated cell death in glioma stem cells
- Author
-
Konrad Gabrusiewicz, Shulin Li, Jun Yan, and Hyangsoon Noh
- Subjects
endocrine system ,Cancer Research ,Programmed cell death ,Chemotherapy ,Temozolomide ,biology ,business.industry ,medicine.drug_class ,medicine.medical_treatment ,fungi ,Brain tumor ,medicine.disease ,Monoclonal antibody ,Molecular biology ,Oncology ,Glioma ,medicine ,Cancer research ,biology.protein ,Stem cell ,Antibody ,business ,medicine.drug - Abstract
Glioblastoma multiforme (GBM) is one of the most aggressive brain tumor in pediatric patients. The current standard of care including radiation and chemotherapy is not effective in most patients due to multiple factors such as resistance and poor blood-brain barrier (BBB) penetration. The development of strategies to reduce resistance and to increase sensitivity to chemotherapy may ameliorate the problems. Glioma stem cells (GSCs) are considered the source of relapse and chemoresistance. Sensitization of temozolomide TMZ resistance in GSC is therefore integral for therapeutic efficacy. Our lab has discovered that cell surface vimentin (CSV) is presented on patient-derived GSCs. In this study, we demonstrate that the treatment of GSCs with anti-CSV antibody (86C) sensitizes them to TMZ. We found that the combination of 86C and TMZ induced additional antitumor effects in 8 out of 12 GSCs. Mechanistic study of the four resistance GSCs revealed slow re-surface rate of CSV from CSV- GSCs and low CSV expression on GSCs as possible contributing factors. GSCs with rapid CSV resurfacing from CSV- GSCs was more sensitive to combination treatment compared to GSCs with a slow recovering from CSV- GSCs. Furthermore, the metabolism study shows these four resistance cells have high intrinsic mitochondria activity compared to sensitive cells. The combination of TMZ with 86C may represent a valuable strategy to reverse GSC chemoresistance. Citation Format: Hyangsoon Noh, Jun Yan, Konrad Gabrusiewicz, Shulin Li. Cell surface vimentin targeted mAb 86C increases sensitivity to temozolomide mediated cell death in glioma stem cells [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 3869. doi:10.1158/1538-7445.AM2017-3869
- Published
- 2017
50. PRESENCE OF A DISTINCTIVE MYELOID POPULATION IS ASSOCIATED WITH THE INVASIVE TUMOR PHENOTYPE OBSERVED AFTER ANTI-ANGIOGENESIS THERAPIES
- Author
-
Kenneth Aldape, Nahir Cortes-Santiago, Frederick Lang, Konrad Gabrusiewicz, W. K. Alfred Yung, Candelaria Gomez-Manzano, Charles A. Conrad, Mohammad B. Hossain, Gregory N. Fuller, Juan Fueyo, and Mark R. Gilbert
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,education.field_of_study ,Temozolomide ,Myeloid ,MMP2 ,business.industry ,Angiogenesis ,Population ,medicine.disease ,abstracts ,Antiangiogenesis Therapy ,medicine.anatomical_structure ,Oncology ,Glioma ,medicine ,Macrophage ,Neurology (clinical) ,business ,education ,medicine.drug - Abstract
BACKGROUND: The addition of anti-angiogenic therapy to the few treatments that are available to patients with malignant gliomas was based on the fact that these tumors are highly vascularized and on findings from initial preclinical and clinical studies which showed encouraging results. However, tumors that initially respond to this therapy invariably recur with the acquisition of highly aggressive and invasive phenotype. Although several myeloid populations have been associated to the recurrence of these tumors, a specific targetable population has not been yet identified as responsible for the heightened invasion observed upon antiangiogenesis therapies. METHODS: Immunocompromised mice and immunocompetent GFP macrophage Fas-induced apoptosis (MAFIA) transgenic mice were used to model the response of malignant gliomas to several antiangiogenesis therapies, such as bevacizumab, VEGF Trap, DC101, and to temozolomide. Analysis of tumors and monocytic populations were performed by immunohistochemistry and double immunofluorescence. In vitro studies encompass the use of isolated monocytes from donors and a novel experimental model based on a THP1 human monocytic cell line. Data was corroborated in surgical specimens from human malignant gliomas RESULTS: Here we present evidence for the accumulation of Tie2-expressing cells at the tumor/normal brain interface of immunocompromised mice treated with anti-VEGF therapies in regions with heightened tumoral invasion. This cell population was further characterized as having markers of M2 polarized monocytes/macrophages compatible with Tie2-expressing monocytes (TEMs). We validated these results using the immunocompetent MAFIA transgenic mice treated with an anti-VEGFR compound. In contrast, TEMs were almost undetectable in the brains of glioma-bearing mice treated with temozolomide or anti-VEGF therapies that did not result in an invasive tumoral pattern. In vitro studies showed that TEMs enhanced the invasive properties of glioma cells compared with monocytes that did not express Tie2 (non-TEMs). Moreover, TEMs displayed a higher gelatinase enzymatic activity and, specifically, secreted higher levels of MMP2 and MMP9 than non-TEMs did. Consistently Tie2/MMP9 monocytic cells were detected in the invasive tumoral edge of mice treated with anti-VEGF therapies. Of clinical relevance, we detected the presence of TEMs in human surgical specimens after anti-VEGF therapy. CONCLUSIONS: Collectively, our results described the association of a specific myeloid/monocytic subpopulation characterized as Tie2-expressing monocytes with the heightened invasion observed after antiangiogenic therapy. Our results suggest that his distinctive population might explain the mechanism of escape of malignant gliomas to anti-angiogenesis therapies. Therefore, combination of antiangiogenesis therapies with approaches to decrease the tumor recruitment of TEMs might be required to obtain a therapeutic effect. SECONDARY CATEGORY: Preclinical Experimental Therapeutics.
- Published
- 2014
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.