Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Mol Cancer ; 23(1): 10, 2024 01 10.
Article in English | MEDLINE | ID: mdl-38200602

ABSTRACT

BACKGROUND AND AIMS: This study sought to determine the value of patient-derived organoids (PDOs) from esophago-gastric adenocarcinoma (EGC) for response prediction to neoadjuvant chemotherapy (neoCTx). METHODS: Endoscopic biopsies of patients with locally advanced EGC (n = 120) were taken into culture and PDOs expanded. PDOs' response towards the single substances of the FLOT regimen and the combination treatment were correlated to patients' pathological response using tumor regression grading. A classifier based on FLOT response of PDOs was established in an exploratory cohort (n = 13) and subsequently confirmed in an independent validation cohort (n = 13). RESULTS: EGC PDOs reflected patients' diverse responses to single chemotherapeutics and the combination regimen FLOT. In the exploratory cohort, PDOs response to single 5-FU and FLOT combination treatment correlated with the patients' pathological response (5-FU: Kendall's τ = 0.411, P = 0.001; FLOT: Kendall's τ = 0.694, P = 2.541e-08). For FLOT testing, a high diagnostic precision in receiver operating characteristic (ROC) analysis was reached with an AUCROC of 0.994 (CI 0.980 to 1.000). The discriminative ability of PDO-based FLOT testing allowed the definition of a threshold, which classified in an independent validation cohort FLOT responders from non-responders with high sensitivity (90%), specificity (100%) and accuracy (92%). CONCLUSION: In vitro drug testing of EGC PDOs has a high predictive accuracy in classifying patients' histological response to neoadjuvant FLOT treatment. Taking into account the high rate of successful PDO expansion from biopsies, the definition of a threshold that allows treatment stratification paves the way for an interventional trial exploring PDO-guided treatment of EGC patients.


Subject(s)
Adenocarcinoma , Carbamates , Pyrazines , Pyridines , Stomach Neoplasms , Humans , Stomach Neoplasms/drug therapy , Combined Modality Therapy , Neoadjuvant Therapy , Adenocarcinoma/drug therapy , Organoids , Fluorouracil/pharmacology
2.
Mol Cancer ; 21(1): 78, 2022 03 18.
Article in English | MEDLINE | ID: mdl-35303871

ABSTRACT

Chimeric Antigen Receptor (CAR) T-cells represent a breakthrough in personalized cancer therapy. In this strategy, synthetic receptors comprised of antigen recognition, signaling, and costimulatory domains are used to reprogram T-cells to target tumor cells for destruction. Despite the success of this approach in refractory B-cell malignancies, optimal potency of CAR T-cell therapy for many other cancers, particularly solid tumors, has not been achieved. Factors such as T-cell exhaustion, lack of CAR T-cell persistence, cytokine-related toxicities, and bottlenecks in the manufacturing of autologous products have hampered the safety, effectiveness, and availability of this approach. With the ease and accessibility of CRISPR-Cas9-based gene editing, it is possible to address many of these limitations. Accordingly, current research efforts focus on precision engineering of CAR T-cells with conventional CRISPR-Cas9 systems or novel editors that can install desired genetic changes with or without introduction of a double-stranded break (DSB) into the genome. These tools and strategies can be directly applied to targeting negative regulators of T-cell function, directing therapeutic transgenes to specific genomic loci, and generating reproducibly safe and potent allogeneic universal CAR T-cell products for on-demand cancer immunotherapy. This review evaluates several of the ongoing and future directions of combining next-generation CRISPR-Cas9 gene editing with synthetic biology to optimize CAR T-cell therapy for future clinical trials toward the establishment of a new cancer treatment paradigm.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , CRISPR-Cas Systems , Gene Editing , Humans , Immunotherapy, Adoptive , Neoplasms/drug therapy , Neoplasms/therapy , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes
3.
Int J Cancer ; 147(2): 519-531, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32077087

ABSTRACT

Disseminated tumor cells (dTCs) can frequently be detected in the bone marrow (BM) of colorectal cancer (CRC) patients, raising the possibility that the BM serves as a reservoir for metastatic tumor cells. Identification of dTCs in BM aspirates harbors the potential of assessing therapeutic outcome and directing therapy intensity with limited risk and effort. Still, the functional and prognostic relevance of dTCs is not fully established. We have previously shown that CRC cell clones can be traced to the BM of mice carrying patient-derived xenografts. However, cellular interactions, proliferative state and tumorigenicity of dTCs remain largely unknown. Here, we applied a coculture system modeling the microvascular niche and used immunofluorescence imaging of the murine BM to show that primary CRC cells migrate toward endothelial tubes. dTCs in the BM were rare, but detectable in mice with xenografts from most patient samples (8/10) predominantly at perivascular sites. Comparable to primary tumors, a substantial fraction of proliferating dTCs was detected in the BM. However, most dTCs were found as isolated cells, indicating that dividing dTCs rather separate than aggregate to metastatic clones-a phenomenon frequently observed in the microvascular niche model. Clonal tracking identified subsets of self-renewing tumor-initiating cells in the BM that formed tumors out of BM transplants, including one subset that did not drive primary tumor growth. Our results indicate an important role of the perivascular BM niche for CRC cell dissemination and show that dTCs can be a potential source for tumor relapse and tumor heterogeneity.


Subject(s)
Bone Marrow/pathology , Colorectal Neoplasms/pathology , Green Fluorescent Proteins/metabolism , Mesenchymal Stem Cells/cytology , Neoplastic Cells, Circulating/pathology , Tumor Cells, Cultured/cytology , Animals , Bone Marrow/metabolism , Cell Tracking , Coculture Techniques , Colorectal Neoplasms/metabolism , Green Fluorescent Proteins/genetics , Human Umbilical Vein Endothelial Cells , Humans , Mesenchymal Stem Cells/metabolism , Mice , Neoplastic Cells, Circulating/metabolism , Optical Imaging , Prognosis , Stem Cell Niche , Time-Lapse Imaging , Tumor Cells, Cultured/metabolism , Xenograft Model Antitumor Assays
4.
EMBO J ; 34(18): 2350-62, 2015 Sep 14.
Article in English | MEDLINE | ID: mdl-26271101

ABSTRACT

The Dnmt2 enzyme utilizes the catalytic mechanism of eukaryotic DNA methyltransferases to methylate several tRNAs at cytosine 38. Dnmt2 mutant mice, flies, and plants were reported to be viable and fertile, and the biological function of Dnmt2 has remained elusive. Here, we show that endochondral ossification is delayed in newborn Dnmt2-deficient mice, which is accompanied by a reduction of the haematopoietic stem and progenitor cell population and a cell-autonomous defect in their differentiation. RNA bisulfite sequencing revealed that Dnmt2 methylates C38 of tRNA Asp(GTC), Gly(GCC), and Val(AAC), thus preventing tRNA fragmentation. Proteomic analyses from primary bone marrow cells uncovered systematic differences in protein expression that are due to specific codon mistranslation by tRNAs lacking Dnmt2-dependent methylation. Our observations demonstrate that Dnmt2 plays an important role in haematopoiesis and define a novel function of C38 tRNA methylation in the discrimination of near-cognate codons, thereby ensuring accurate polypeptide synthesis.


Subject(s)
Cell Differentiation/physiology , DNA (Cytosine-5-)-Methyltransferases/metabolism , Hematopoiesis/physiology , Hematopoietic Stem Cells/enzymology , Protein Biosynthesis/physiology , Animals , Animals, Newborn , DNA (Cytosine-5-)-Methyltransferases/genetics , Hematopoietic Stem Cells/cytology , Methylation , Mice , Mice, Knockout , Osteogenesis/physiology , RNA, Transfer/genetics , RNA, Transfer/metabolism
5.
BMC Cancer ; 19(1): 1181, 2019 Dec 03.
Article in English | MEDLINE | ID: mdl-31796022

ABSTRACT

BACKGROUND: While colorectal cancer (CRC) patients with localized disease have a favorable prognosis, the five-year-survival rate in patients with distant spread is still below 15%. Hence, a detailed understanding of the mechanisms regulating metastasis formation is essential to develop therapeutic strategies targeting metastasized CRC. The notch pathway has been shown to be involved in the metastatic spread of various tumor entities; however, the impact of its target gene HEYL remains unclear so far. METHODS: In this study, we functionally assessed the association between high HEYL expression and metastasis formation in human CRC. Therefore, we lentivirally overexpressed HEYL in two human patient-derived CRC cultures differing in their spontaneous metastasizing capacity and analyzed metastasis formation as well as tumor cell dissemination into the bone marrow after xenotransplantation into NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ (NSG) mice. RESULTS: HEYL overexpression decreased tumor cell dissemination and the absolute numbers of formed metastases in a sub-renal capsular spontaneous metastasis formation model, addressing all steps of the metastatic cascade. In contrast, metastatic capacity was not decreased following intrasplenic xenotransplantation where the cells are placed directly into the blood circulation. CONCLUSION: These results suggest that HEYL negatively regulates metastasis formation in vivo presumably by inhibiting intravasation of metastasis-initiating cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone Marrow Neoplasms/secondary , Colorectal Neoplasms/pathology , Repressor Proteins/metabolism , Spheroids, Cellular/pathology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Bone Marrow Neoplasms/genetics , Bone Marrow Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/physiology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Female , Heterografts , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Receptor, Notch1/metabolism , Repressor Proteins/genetics , Spheroids, Cellular/metabolism
6.
Blood ; 127(12): 1575-86, 2016 Mar 24.
Article in English | MEDLINE | ID: mdl-26729896

ABSTRACT

The de novo DNA methyltransferases Dnmt3a and Dnmt3b are of crucial importance in hematopoietic stem cells. Dnmt3b has recently been shown to play a role in genic methylation. To investigate how Dnmt3b-mediated DNA methylation affects leukemogenesis, we analyzed leukemia development under conditions of high and physiological methylation levels in a tetracycline-inducible knock-in mouse model. High expression of Dnmt3b slowed leukemia development in serial transplantations and impaired leukemia stem cell (LSC) function. Forced Dnmt3b expression induced widespread DNA hypermethylation inMyc-Bcl2-induced leukemias, preferentially at gene bodies.MLL-AF9-induced leukemogenesis showed much less pronounced DNA hypermethylation upon Dnmt3b expression. Nonetheless, leukemogenesis was delayed in both models with a shared core set of DNA hypermethylated regions and suppression of stem cell-related genes. Acute myeloid leukemia patients with high expression of Dnmt3b target genes showed inferior survival. Together, these findings indicate a critical role for Dnmt3b-mediated DNA methylation in leukemia development and maintenance of LSC function.


Subject(s)
Carcinogenesis/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation , Gene Expression Regulation, Leukemic , Leukemia/genetics , Animals , Carcinogenesis/pathology , Gene Knock-In Techniques , Hematopoiesis , Humans , Leukemia/diagnosis , Leukemia/pathology , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred C57BL , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Prognosis , Promoter Regions, Genetic , DNA Methyltransferase 3B
7.
Mol Carcinog ; 54(12): 1815-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25491945

ABSTRACT

Even though a large proportion of patients with acute myeloid leukemia (AML) achieve a complete remission upon initial therapy, the majority of them eventually relapse with resistant disease. Overexpression of the gene coding for the transcription factor Ecotropic Virus Integration site 1 (EVI1) is associated with rapid disease recurrence and shortened survival. We therefore sought to identify EVI1 target genes that may play a role in chemotherapy resistance using a previously established in vitro model system for EVI1 positive myeloid malignancies. Gene expression microarray analyses uncovered the Cell Adhesion Molecule 1 (CADM1) gene as a candidate whose deregulation by EVI1 may contribute to drug refractoriness. CADM1 is an apoptosis inducing tumor suppressor gene that is inactivated by methylation in a variety of tumor types. In the present study we provide evidence that it may play a role in chemotherapy induced cell death in AML: CADM1 was induced by drugs used in the treatment of AML in a human myeloid cell line and in primary diagnostic AML samples, and its experimental expression in a cell line model increased the proportion of apoptotic cells. CADM1 up-regulation was abolished by ectopic expression of EVI1, and EVI1 expression correlated with increased CADM1 promoter methylation both in a cell line model and in primary AML cells. Finally, CADM1 induction was repressed in primary samples from AML patients at relapse. In summary, these data suggest that failure to up-regulate CADM1 in response to chemotherapeutic drugs may contribute to therapy resistance in AML.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/genetics , Cell Adhesion Molecules/genetics , Drug Resistance, Neoplasm/genetics , Genes, Tumor Suppressor/physiology , Immunoglobulins/genetics , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Aged , Apoptosis/drug effects , Cell Adhesion Molecule-1 , Cell Line, Tumor , DNA Methylation/drug effects , DNA Methylation/genetics , DNA-Binding Proteins/genetics , Female , Gene Expression/drug effects , Gene Expression/genetics , Humans , MDS1 and EVI1 Complex Locus Protein , Middle Aged , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Proto-Oncogenes/genetics , Transcription Factors/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
8.
Stem Cells ; 30(9): 1961-70, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22696148

ABSTRACT

Hematopoietic stem cells (HSCs) generate all mature blood cells during the whole lifespan of an individual. However, the clonal contribution of individual HSC and progenitor cells in steady-state hematopoiesis is poorly understood. To investigate the activity of HSCs under steady-state conditions, murine HSC and progenitor cells were genetically marked in vivo by integrating lentiviral vectors (LVs) encoding green fluorescent protein (GFP). Hematopoietic contribution of individual marked clones was monitored by determination of lentiviral integration sites using highly sensitive linear amplification-mediated-polymerase chain reaction. A remarkably stable small proportion of hematopoietic cells expressed GFP in LV-injected animals for up to 24 months, indicating stable marking of murine steady-state hematopoiesis. Analysis of the lentiviral integration sites revealed that multiple hematopoietic clones with both myeloid and lymphoid differentiation potential contributed to long-term hematopoiesis. In contrast to intrafemoral vector injection, intravenous administration of LV preferentially targeted short-lived progenitor cells. Myelosuppressive treatment of mice prior to LV-injection did not affect the marking efficiency. Our study represents the first continuous analysis of clonal behavior of genetically marked hematopoietic cells in an unmanipulated system, providing evidence that multiple clones are simultaneously active in murine steady-state hematopoiesis.


Subject(s)
Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Animals , Cell Differentiation/physiology , Cell Lineage , Gene Transfer Techniques , Genetic Vectors , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mice, Inbred C57BL
9.
Mol Ther ; 20(5): 1014-21, 2012 May.
Article in English | MEDLINE | ID: mdl-22434137

ABSTRACT

Lentiviral vectors (LV) are widely used to stably transfer genes into target cells investigating or treating gene functions. In addition, gene transfer into early murine embryos may be improved to efficiently generate transgenic mice. We applied lentiviral gene transfer to generate a mouse model transgenic for SET binding protein-1 (Setbp1) and enhanced green fluorescent protein (eGFP). Neither transgenic founders nor their vector-positive offspring transcribed or expressed the transgenes. Bisulfite sequencing of the internal spleen focus-forming virus (SFFV) promoter demonstrated extensive methylation of all analyzed CpGs in the transgenic mice. To analyze the impact of Setbp1 on epigenetic silencing, embryonic stem cells (ESC) were differentiated into cardiomyocytes (CM) in vitro. In contrast to human promoters in LV, virally derived promoter sequences were strongly methylated during differentiation, independent of the transgene. Moreover, the commonly used SFFV promoter (SFFVp) was highly methylated with remarkable strength and frequency during hematopoietic differentiation in vivo in LV but less in γ-retroviral (γ-RV) backbones. In summary, we conclude that LV using an internal SFFVp are not suitable to generate transgenic mice or perform constitutive expression studies in differentiating cells. Choosing the appropriate promoter is also crucial to allow stable transgene expression in clinical gene therapy.


Subject(s)
Carrier Proteins/genetics , Genetic Vectors , Lentivirus/genetics , Mice, Transgenic/genetics , Spleen Focus-Forming Viruses/genetics , Stem Cells/metabolism , Animals , Cell Differentiation , CpG Islands/genetics , DNA Methylation , Epigenesis, Genetic , Founder Effect , Gene Silencing , Genes, Essential , Green Fluorescent Proteins/genetics , Humans , Mice , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Promoter Regions, Genetic , Sequence Analysis, DNA , Stem Cells/cytology , Transgenes
10.
Sci Transl Med ; 15(714): eadi1145, 2023 09 20.
Article in English | MEDLINE | ID: mdl-37651540

ABSTRACT

In the absence of cell surface cancer-specific antigens, immunotherapies such as chimeric antigen receptor (CAR) T cells, monoclonal antibodies, or bispecific T cell engagers typically target lineage antigens. Currently, such immunotherapies are individually designed and tested for each disease. This approach is inefficient and limited to a few lineage antigens for which the on-target/off-tumor toxicities are clinically tolerated. Here, we sought to develop a universal CAR T cell therapy for blood cancers directed against the pan-leukocyte marker CD45. To protect healthy hematopoietic cells, including CAR T cells, from CD45-directed on-target/off-tumor toxicity while preserving the essential functions of CD45, we mapped the epitope on CD45 that is targeted by the CAR and used CRISPR adenine base editing to install a function-preserving mutation sufficient to evade CAR T cell recognition. Epitope-edited CD45 CAR T cells were fratricide resistant and effective against patient-derived acute myeloid leukemia, B cell lymphoma, and acute T cell leukemia. Epitope-edited hematopoietic stem cells (HSCs) were protected from CAR T cells and, unlike CD45 knockout cells, could engraft, persist, and differentiate in vivo. Ex vivo epitope editing in HSCs and T cells enables the safe and effective use of CD45-directed CAR T cells and bispecific T cell engagers for the universal treatment of hematologic malignancies and might be exploited for other diseases requiring intensive hematopoietic ablation.


Subject(s)
Hematologic Neoplasms , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Humans , Epitopes , Gene Editing , Hematologic Neoplasms/therapy , Immunotherapy
SELECTION OF CITATIONS
SEARCH DETAIL