13 results on '"Michael A. Amrein"'
Search Results
2. Data from T-cell–Secreted TNFα Induces Emergency Myelopoiesis and Myeloid-Derived Suppressor Cell Differentiation in Cancer
- Author
-
Adrian F. Ochsenbein, Carsten Riether, Ramin Radpour, Anne-Laure Huguenin, Elias D. Bührer, Michael A. Amrein, and Mohamad F. Al Sayed
- Abstract
Hematopoiesis in patients with cancer is characterized by reduced production of red blood cells and an increase in myelopoiesis, which contributes to the immunosuppressive environment in cancer. Some tumors produce growth factors that directly stimulate myelopoiesis such as G-CSF or GM-CSF. However, for a majority of tumors that do not directly secrete hematopoietic growth factors, the mechanisms involved in the activation of myelopoiesis are poorly characterized. In this study, we document in different murine tumor models activated hematopoiesis with increased proliferation of long-term and short-term hematopoietic stem cells and myeloid progenitor cells. As a consequence, the frequency of myeloid-derived suppressor cells and its ratio to CD8+ T cells increased in tumor-bearing mice. Activation of hematopoiesis and myeloid differentiation in tumor-bearing mice was induced by TNFα, which was mainly secreted by activated CD4+ T cells. Therefore, the activated adaptive immune system in cancer induces emergency myelopoiesis and immunosuppression.Significance:These findings characterize a regulatory circuit linking activated T cells to suppression of tumor-specific immune responses, providing a conceptual advance in the understanding of emergency-hematopoiesis in cancer and opening new targets for therapeutic approaches.
- Published
- 2023
- Full Text
- View/download PDF
3. Supplementary Data from T-cell–Secreted TNFα Induces Emergency Myelopoiesis and Myeloid-Derived Suppressor Cell Differentiation in Cancer
- Author
-
Adrian F. Ochsenbein, Carsten Riether, Ramin Radpour, Anne-Laure Huguenin, Elias D. Bührer, Michael A. Amrein, and Mohamad F. Al Sayed
- Abstract
Table S1: suppl. cell cycle analysis to Fig 3; Table S2: suppl. BrDU and AnnexinV analysis to Fig 3; Table S3: suppl. analysis of the compositions of HSPC compartments to Fig 4; Figure S1: suppl. to Fig 1; Figure S2: suppl. to Fig 4; Figure S3: suppl. to Fig 6; Figure S4: suppl. to Fig 7
- Published
- 2023
- Full Text
- View/download PDF
4. LIGHT/LTβR signaling regulates self-renewal and differentiation of hematopoietic and leukemia stem cells
- Author
-
Ramin Radpour, Sabine Höpner, Ana Raykova, Gabriela M. Baerlocher, D. Koller, Michael A. Amrein, Adrian F. Ochsenbein, and Carsten Riether
- Subjects
0301 basic medicine ,Cell ,General Physics and Astronomy ,Symmetric cell division ,Antigens, CD34 ,0302 clinical medicine ,hemic and lymphatic diseases ,Cell Self Renewal ,610 Medicine & health ,Mice, Knockout ,Multidisciplinary ,Cancer stem cells ,Gene Expression Regulation, Leukemic ,Haematopoietic stem cells ,Cell Cycle ,Myeloid leukemia ,Hematopoietic stem cell ,Cell Differentiation ,Cell biology ,Leukemia ,Haematopoiesis ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Neoplastic Stem Cells ,Self-renewal ,Fluorouracil ,Stem cell ,Signal Transduction ,Tumor Necrosis Factor Ligand Superfamily Member 14 ,Science ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Lymphotoxin beta Receptor ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,medicine ,Animals ,Humans ,RNA, Messenger ,Cell Proliferation ,General Chemistry ,medicine.disease ,Hematopoietic Stem Cells ,Mice, Inbred C57BL ,030104 developmental biology ,Lymphotoxin ,DNA Damage - Abstract
The production of blood cells during steady-state and increased demand depends on the regulation of hematopoietic stem cell (HSC) self-renewal and differentiation. Similarly, the balance between self-renewal and differentiation of leukemia stem cells (LSCs) is crucial in the pathogenesis of leukemia. Here, we document that the TNF receptor superfamily member lymphotoxin-β receptor (LTβR) and its ligand LIGHT regulate quiescence and self-renewal of murine and human HSCs and LSCs. Cell-autonomous LIGHT/LTβR signaling on HSCs reduces cell cycling, promotes symmetric cell division and prevents primitive HSCs from exhaustion in serial re-transplantation experiments and genotoxic stress. LTβR deficiency reduces the numbers of LSCs and prolongs survival in a murine chronic myeloid leukemia (CML) model. Similarly, LIGHT/LTβR signaling in human G-CSF mobilized HSCs and human LSCs results in increased colony forming capacity in vitro. Thus, our results define LIGHT/LTβR signaling as an important pathway in the regulation of the self-renewal of HSCs and LSCs., Regulation of self-renewal is critical during steady state and stress in haematpoietic stem cells (HSCs), and underlies leukaemia pathology. Here, the authors show that LIGHT and its receptor LTβR promote quiescence and self-renewal of HSCs and that LTβR deficiency promotes survival in a mouse leukaemia model.
- Published
- 2021
- Full Text
- View/download PDF
5. Chemotherapy negatively impacts the tumor immune microenvironment in NSCLC: an analysis of pre- and post-treatment biopsies in the multi-center SAKK19/09 study
- Author
-
M L Amrein, S Savic-Prince, Adrian F. Ochsenbein, Michael A. Amrein, Elias D. Bührer, Rolf Jaggi, Sacha I. Rothschild, Lukas Bubendorf, Qiyu Li, Carsten Riether, and O. Gautschi
- Subjects
Male ,Cancer Research ,Lung Neoplasms ,LAG3 ,Biopsy ,medicine.medical_treatment ,Immunology ,Gene Expression ,chemical and pharmacologic phenomena ,610 Medicine & health ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Carcinoma, Non-Small-Cell Lung ,Tumor Microenvironment ,Humans ,Chemotherapy ,Immunology and Allergy ,Medicine ,030212 general & internal medicine ,Lung cancer ,TNFRSF18 ,business.industry ,FOXP3 ,Cancer ,medicine.disease ,Immune checkpoint ,Immune microenvironment ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Female ,Original Article ,business ,Non-small-cell lung cancer - Abstract
Background Over the past few years, immune checkpoint inhibitors have changed the therapeutic landscape of non-small-cell lung cancer (NSCLC). Response to immune checkpoint inhibitors correlates with a pre-existing anti-tumoral immune response. Checkpoint inhibitors have been introduced as second-line therapy and are only very recently used as monotherapy or in combination with chemotherapy as first-line treatment of NSCLC. However, the effect of conventional first-line platinum-based chemotherapy on the immune infiltrate in the tumor is largely unknown. Methods We measured the gene expression of a custom set of 201 cancer- and immune-related genes in 100 NSCLC tumor biopsies collected before chemotherapy and 33 re-biopsies after platinum-based chemotherapy at the time point of progression. For 29 patients matched pre- and post-chemotherapy samples could be evaluated. Results We identified a cluster of 47 co-expressed immune genes, including PDCD1 (PD1) and CD274 (PD-L1), along with three other co-expression clusters. Chemotherapy decreased the average gene expression of the immune cluster while no effect was observed on the other three cluster. Within this immune cluster, CTLA4, LAG3, TNFRSF18, CD80 and FOXP3 were found to be significantly decreased in patient-matched samples after chemotherapy. Conclusion Our results suggest that conventional platinum-based chemotherapy negatively impacts the immune microenvironment at the time point of secondary progression.
- Published
- 2020
- Full Text
- View/download PDF
6. Tumor Initiation Capacity and Therapy Resistance Are Differential Features of EMT-Related Subpopulations in the NSCLC Cell Line A549
- Author
-
Gregor J. Kocher, Elias D. Bührer, Yanyun Gao, Laurène Froment, Sean R R Hall, Adrian F. Ochsenbein, Carsten Riether, Colin Tièche, Rémy Bruggmann, Ralph A. Schmid, Stefan Weis, Primo Schär, Kurt Wyler, Nina Hobi, Michael A. Amrein, Sabina Berezowska, Thomas M. Marti, Ren-Wang Peng, and Patrick Dorn
- Subjects
0301 basic medicine ,Cancer Research ,Lung Neoplasms ,Tumor initiation ,POU5F1, OCT4 ,TIC, tumor initiation cell ,Receptor tyrosine kinase ,SNAI1, SNAIL ,Mice ,0302 clinical medicine ,CDH1, E-cadherin ,SCLC, small cell lung cancer ,Carcinoma, Non-Small-Cell Lung ,540 Chemistry ,MYC, cellular Myc ,MMP2, matrix metalloproteinase-2 ,610 Medicine & health ,MYCN, N-Myc ,SOX2+, high SOX2 ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,NSCLC, non–small cell lung cancers ,Phenotype ,Cell Transformation, Neoplastic ,030220 oncology & carcinogenesis ,DNA methylation ,Neoplastic Stem Cells ,GOseq, KEGG pathway enrichment analysis ,medicine.drug ,Original article ,Epithelial-Mesenchymal Transition ,CSCs, cancer stem cells ,ALCAM, CD166 ,hybrid-E/M, hybrid epithelial/mesenchymal ,Biology ,SNAI2, SLUG ,lcsh:RC254-282 ,Immunophenotyping ,CDH17, Cadherin 17 ,CTSB, Cathepsin B ,03 medical and health sciences ,Cancer stem cell ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Epigenetics ,Cell Proliferation ,Cisplatin ,VIM, Vimentin ,CD90−, low CD90 expression ,Gene Expression Profiling ,DNA Methylation ,EGFR, epidermal growth factor receptor ,Disease Models, Animal ,030104 developmental biology ,A549 Cells ,Cell culture ,Cancer research ,biology.protein ,570 Life sciences ,biology ,IR, ionizing radiation ,Transcriptome ,Biomarkers ,EMT, epithelial-to-mesenchymal transition ,DNA Damage - Abstract
Cell lines are essential tools to standardize and compare experimental findings in basic and translational cancer research. The current dogma states that cancer stem cells feature an increased tumor initiation capacity and are also chemoresistant. Here, we identified and comprehensively characterized three morphologically distinct cellular subtypes in the non–small cell lung cancer cell line A549 and challenge the current cancer stem cell dogma. Subtype-specific cellular morphology is maintained during short-term culturing, resulting in the formation of holoclonal, meroclonal, and paraclonal colonies. A549 holoclone cells were characterized by an epithelial and stem-like phenotype, paraclone cells featured a mesenchymal phenotype, whereas meroclone cells were phenotypically intermediate. Cell-surface marker expression of subpopulations changed over time, indicating an active epithelial-to-mesenchymal transition (EMT), in vitro and in vivo. EMT has been associated with the overexpression of the immunomodulators PD-L1 and PD-L2, which were 37- and 235-fold overexpressed in para- versus holoclone cells, respectively. We found that DNA methylation is involved in epigenetic regulation of marker expression. Holoclone cells were extremely sensitive to cisplatin and radiotherapy in vitro, whereas paraclone cells were highly resistant. However, inhibition of the receptor tyrosine kinase AXL, whose expression is associated with an EMT, specifically targeted the otherwise highly resistant paraclone cells. Xenograft tumor formation capacity was 24- and 269-fold higher in holo- than mero- and paraclone cells, respectively. Our results show that A549 subpopulations might serve as a unique system to explore the network of stemness, cellular plasticity, tumor initiation capacity, invasive and metastatic potential, and chemo/radiotherapy resistance.
- Published
- 2019
- Full Text
- View/download PDF
7. Combined mTOR inhibition and chemotherapy as an effective strategy to treat KRAS-mutant lung cancer
- Author
-
Ralph A. Schmid, Laurène Froment, RW Peng, Sabina Berezowska, S-Q. Liang, Zhang Yang, Erik Vassella, Gregor J. Kocher, Adrian F. Ochsenbein, Thomas M. Marti, Elias D. Bührer, Srr Hall, Duo Xu, Carsten Riether, Michael A. Amrein, and Haitang Yang
- Subjects
Chemotherapy ,business.industry ,medicine.medical_treatment ,Mutant ,medicine ,Cancer research ,KRAS ,Lung cancer ,medicine.disease ,medicine.disease_cause ,business ,PI3K/AKT/mTOR pathway - Published
- 2019
- Full Text
- View/download PDF
8. TNIK signaling imprints CD8
- Author
-
Carla A, Jaeger-Ruckstuhl, Magdalena, Hinterbrandner, Sabine, Höpner, Colin E, Correnti, Ursina, Lüthi, Olivier, Friedli, Stefan, Freigang, Mohamad F, Al Sayed, Elias D, Bührer, Michael A, Amrein, Christian M, Schürch, Ramin, Radpour, Carsten, Riether, and Adrian F, Ochsenbein
- Subjects
Mice, Knockout ,Apoptosis ,Cell Differentiation ,CD8-Positive T-Lymphocytes ,Lymphocytic Choriomeningitis ,Protein Serine-Threonine Kinases ,Signal transduction ,Lymphocyte Activation ,Immunological memory ,Article ,Mice ,Viral infection ,Animals ,Humans ,Lymphocytic choriomeningitis virus ,CD8-positive T cells ,Immunologic Memory ,Wnt Signaling Pathway - Abstract
Co-stimulatory signals, cytokines and transcription factors regulate the balance between effector and memory cell differentiation during T cell activation. Here, we analyse the role of the TRAF2-/NCK-interacting kinase (TNIK), a signaling molecule downstream of the tumor necrosis factor superfamily receptors such as CD27, in the regulation of CD8+ T cell fate during acute infection with lymphocytic choriomeningitis virus. Priming of CD8+ T cells induces a TNIK-dependent nuclear translocation of β-catenin with consecutive Wnt pathway activation. TNIK-deficiency during T cell activation results in enhanced differentiation towards effector cells, glycolysis and apoptosis. TNIK signaling enriches for memory precursors by favouring symmetric over asymmetric cell division. This enlarges the pool of memory CD8+ T cells and increases their capacity to expand after re-infection in serial re-transplantation experiments. These findings reveal that TNIK is an important regulator of effector and memory T cell differentiation and induces a population of stem cell-like memory T cells., Coordinate expression of multiple factors play critical roles in the regulation between effector and memory CD8+ T cell differentiation. Here the authors show upon acute viral infection TNIK is critically required as a regulator of effector and memory T cell differentiation.
- Published
- 2019
9. T-cell-Secreted TNFα Induces Emergency Myelopoiesis and Myeloid-Derived Suppressor Cell Differentiation in Cancer
- Author
-
Adrian F. Ochsenbein, Anne-Laure Huguenin, Carsten Riether, Mohamad F. Al Sayed, Elias D. Bührer, Michael A. Amrein, and Ramin Radpour
- Subjects
0301 basic medicine ,CD4-Positive T-Lymphocytes ,Cancer Research ,Myeloid ,T cell ,Bone Marrow Cells ,Mice, Transgenic ,Biology ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Lymphocytes, Tumor-Infiltrating ,medicine ,Animals ,Myeloid Progenitor Cells ,Myelopoiesis ,Tumor Necrosis Factor-alpha ,Myeloid-Derived Suppressor Cells ,Cell Differentiation ,Neoplasms, Experimental ,Acquired immune system ,Hematopoietic Stem Cells ,Mice, Inbred C57BL ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Myeloid-derived Suppressor Cell ,Stem cell - Abstract
Hematopoiesis in patients with cancer is characterized by reduced production of red blood cells and an increase in myelopoiesis, which contributes to the immunosuppressive environment in cancer. Some tumors produce growth factors that directly stimulate myelopoiesis such as G-CSF or GM-CSF. However, for a majority of tumors that do not directly secrete hematopoietic growth factors, the mechanisms involved in the activation of myelopoiesis are poorly characterized. In this study, we document in different murine tumor models activated hematopoiesis with increased proliferation of long-term and short-term hematopoietic stem cells and myeloid progenitor cells. As a consequence, the frequency of myeloid-derived suppressor cells and its ratio to CD8+ T cells increased in tumor-bearing mice. Activation of hematopoiesis and myeloid differentiation in tumor-bearing mice was induced by TNFα, which was mainly secreted by activated CD4+ T cells. Therefore, the activated adaptive immune system in cancer induces emergency myelopoiesis and immunosuppression. Significance: These findings characterize a regulatory circuit linking activated T cells to suppression of tumor-specific immune responses, providing a conceptual advance in the understanding of emergency-hematopoiesis in cancer and opening new targets for therapeutic approaches.
- Published
- 2018
10. mTOR mediates a mechanism of resistance to chemotherapy and defines a rational combination strategy to treat KRAS-mutant lung cancer
- Author
-
Shun-Qing Liang, Ralph A. Schmid, Carsten Riether, Ren-Wang Peng, Laurène Froment, Thomas M. Marti, Elias D. Bührer, Sabina Berezowska, Erik Vassella, Duo Xu, Gregor J. Kocher, Zhang Yang, Adrian F. Ochsenbein, Haitang Yang, Sean Hall, and Michael A. Amrein
- Subjects
0301 basic medicine ,Cancer Research ,Lung Neoplasms ,medicine.medical_treatment ,Adenocarcinoma of Lung ,Drug resistance ,Biology ,medicine.disease_cause ,Proto-Oncogene Proteins p21(ras) ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Genetics ,medicine ,Animals ,Humans ,Lung cancer ,neoplasms ,Molecular Biology ,PI3K/AKT/mTOR pathway ,Mice, Knockout ,Chemotherapy ,Cell growth ,TOR Serine-Threonine Kinases ,medicine.disease ,digestive system diseases ,respiratory tract diseases ,030104 developmental biology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Adenocarcinoma ,KRAS - Abstract
Oncogenic KRAS mutations comprise the largest subset of lung cancer defined by genetic alterations, but in the clinic no targeted therapies are available that effectively control mutational KRAS activation. Consequently, patients with KRAS-driven tumors are routinely treated with cytotoxic chemotherapy, which is often transiently effective owing to development of drug resistance. In this study, we show that hyperactivated mammalian target of rapamycin (mTOR) pathway is a characteristic hallmark of KRAS-mutant lung adenocarcinoma after chemotherapy treatment, and that KRAS-mutant lung cancer cells rely on persistent mTOR signaling to resist chemotherapeutic drugs. Coherently, mTOR inhibition circumvents the refractory phenotype and restores sensitivity of resistant KRAS-mutant lung cancer cells to chemotherapy. Importantly, drug combinations of clinically approved mTOR inhibitors and chemotherapy drugs synergize in inhibiting cell proliferation of KRAS-mutant cancer cells in vitro and in vivo, and the efficacy of this combination treatment correlates with the magnitude of mTOR activity induced by chemotherapy alone. These results pinpoint mTOR as a mechanism of resistance to chemotherapy in KRAS-mutant lung cancer and validate a rational and readily translatable strategy that combines mTOR inhibitors with standard chemotherapy to treat KRAS-mutant adenocarcinoma, the most common and deadliest lung cancer subset.
- Published
- 2018
11. Splenic CD24low Red Pulp Macrophages Provide an Alternate Niche for Chronic Myeloid Leukemia Stem Cells
- Author
-
Salil S. Bhate, Michael A. Amrein, Ramin Radpour, Stephan Isringhausen, Christian M. Schürch, Stefan Forster, Carsten Riether, Mohamad F. Al Sayed, Cesar Nombela Arrieta, Elias D. Bührer, and Adrian F. Ochsenbein
- Subjects
Myeloid ,Immunology ,Myeloid leukemia ,Spleen ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Transplantation ,medicine.anatomical_structure ,medicine ,Cancer research ,Red pulp ,Bone marrow ,Stem cell ,Progenitor cell - Abstract
Chronic myeloid leukemia (CML) is a typical stem-cell driven malignancy, driven by leukemia stem cells (LSCs). LSCs are resistant to conventional therapies. This resistance is mediated by cell-intrinsic mechanisms and interactions with the microenvironment. LSCs depend on signals from a specialized microenvironment, a so called niche, to maintain their stem cell characteristics. In CML the bone marrow (BM) as a niche is well-investigated and several therapeutic targets, which aim at LSCs by interrupting their interaction with the BM-niche are under investigation. However, even though splenomegaly is a hallmark of CML the contribution of the splenic microenvironment to CML development has not been studied so far. This project aims to investigate the role of the splenic microenvironment as an independent secondary LSC niche and its contribution to disease development. To induce a CML-like disease in mice we retrovirally transduced FACS-sorted Lineage- Sca-1+ cKit+ BM cells with pMSCV-p210BCR/ABL-IRES-GFP and injected the transduced cells into non-irradiated mice. To find out if the spleen contributes to disease development we induced CML in splenectomized and sham operated mice. Splenectomized mice survived significantly longer compared to sham operated controls (median survival 31 vs. 22 days; p=0.0006) with 20% of the splenectomized mice surviving longer than 90 days. Moreover, the number of LSCs in the BM of splenectomized mice was reduced 3.7-fold (p=0.002). Flowcytometric analysis of the spleen and BM compartments of CML bearing mice revealed that the majority of the leukemic stem and progenitor cells (LSPCs) were located in the spleen (19-fold more LSCs in the spleen; p =0.007). Moreover we found the leukemic compartment in the spleen to be enriched for LSPCs compared to the BM (20 % spleen vs. 10 % BM; % LSPCs of total leukemic cells; p=0.01). To confirm this phenotypic observation functionally we performed a limiting dilution transplantation of leukemic cells from spleen and BM. In line with the phenotypic observation we found a higher frequency of LSCs in the spleen compared to the BM (1/41'703 vs. 1/432'594; p=0.02). We next analyzed the gene expression of LSPCs from spleen and BM. We found that the gene expression profile of splenic LSPCs showed higher expression of stemness-related genes and reduced expression of myeloid differentiation genes compared to BM LSPCs, indicating that the spleen is more supportive of primitive LSPCs. Knowing that the spleen contributes to disease development by providing an alternate niche for LSCs we next analyzed the spleens using confocal microscopy. We found that the LSCs resided exclusively in the red pulp. Previous studies have shown that HSCs reside in direct contact with red pulp macrophages (RPMs) during extramedullary hematopoiesis (Dutta et al., JEM, 2015). In addition we found that in spleens from human CML patients CD34+ leukemia cells localized together with macrophages (p=0.001). Furthermore we could show that RPMs are capable of producing both SCF and G-CSF. To test the role of RPMs as a potential niche component in vitro we co-incubated LSCs and RPMs overnight before plating the LSCs in a colony formation assay. We found that the co-incubation with RPMs improved the colony formation capacity of LSCs (CFUs 166 vs. 138; p=0.0356). To test the role of RPMs in vivo we depleted macrophages in CML mice using clodronate liposomes. This resulted in significantly reduced splenomegaly (867mg vs. 249mg; p In summary, we found that the spleen provides an alternate niche for LSCs, thereby contributing to CML development. Compared to the BM niche the splenic niche is more supportive of primitive LSPCs, as shown by the higher frequency of LSCSs found in the spleen and the higher expression of stemness related genes in splenic LSCs compared to BM LSCs. Moreover we identified CD24low RPMs as a unique and central component of the splenic LSC niche. Even though we could show that RPMs are capable of producing SCF and G-CSF the exact mechanisms by which RPMs support LSCs remains to be investigated. Disclosures No relevant conflicts of interest to declare.
- Published
- 2019
- Full Text
- View/download PDF
12. Cytotoxic T cells induce proliferation of chronic myeloid leukemia stem cells by secreting interferon-?
- Author
-
Adrian F. Ochsenbein, Christian M. Schürch, Carsten Riether, and Michael A. Amrein
- Subjects
Adult ,Cytotoxicity, Immunologic ,T cell ,Molecular Sequence Data ,Immunology ,610 Medicine & health ,Biology ,Immunotherapy, Adoptive ,Article ,Interferon-gamma ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Immunology and Allergy ,Cytotoxic T cell ,Amino Acid Sequence ,Progenitor cell ,Aged ,Cell Proliferation ,030304 developmental biology ,0303 health sciences ,ABL ,Myeloid leukemia ,Middle Aged ,medicine.disease ,3. Good health ,Mice, Inbred C57BL ,Haematopoiesis ,Leukemia ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Neoplastic Stem Cells ,Stem cell ,T-Lymphocytes, Cytotoxic - Abstract
Cytotoxic T cells increase leukemia stem cell numbers in a murine model of chronic myeloid leukemia., Chronic myeloid leukemia (CML) is a clonal myeloproliferative neoplasia arising from the oncogenic break point cluster region/Abelson murine leukemia viral oncogene homolog 1 translocation in hematopoietic stem cells (HSCs), resulting in a leukemia stem cell (LSC). Curing CML depends on the eradication of LSCs. Unfortunately, LSCs are resistant to current treatment strategies. The host’s immune system is thought to contribute to disease control, and several immunotherapy strategies are under investigation. However, the interaction of the immune system with LSCs is poorly defined. In the present study, we use a murine CML model to show that LSCs express major histocompatibility complex (MHC) and co-stimulatory molecules and are recognized and killed by leukemia-specific CD8+ effector CTLs in vitro. In contrast, therapeutic infusions of effector CTLs into CML mice in vivo failed to eradicate LSCs but, paradoxically, increased LSC numbers. LSC proliferation and differentiation was induced by CTL-secreted IFN-γ. Effector CTLs were only able to eliminate LSCs in a situation with minimal leukemia load where CTL-secreted IFN-γ levels were low. In addition, IFN-γ increased proliferation and colony formation of CD34+ stem/progenitor cells from CML patients in vitro. Our study reveals a novel mechanism by which the immune system contributes to leukemia progression and may be important to improve T cell–based immunotherapy against leukemia.
- Published
- 2013
- Full Text
- View/download PDF
13. MA 15.11 CCNE1, PTGS2, TGFA and WISP2 Predict Benefit from Bevacizumab and Chemotherapy in Patients with Advanced Non-Small Cell Lung Cancer (SAKK19/09)
- Author
-
Rolf A. Stahel, Daniel C. Betticher, Martina Schneider, Miklos Pless, Thilo Zander, Carsten Riether, Martin Früh, Richard Cathomas, N Schuster, Spasenija Savic, Rolf Jaggi, Sacha I. Rothschild, Michael A. Amrein, Christine Biaggi, Qiyu Li, Alfred Zippelius, Martin Brutsche, Daniel Rauch, O. Gautschi, Adrian F. Ochsenbein, and Lukas Bubendorf
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Oncology ,medicine.medical_specialty ,Chemotherapy ,TGF alpha ,Bevacizumab ,business.industry ,medicine.medical_treatment ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,In patient ,Non small cell ,business ,Lung cancer ,medicine.drug - Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.