Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Article in English | MEDLINE | ID: mdl-35197278

ABSTRACT

Evasion from drug-induced apoptosis is a crucial mechanism of cancer treatment resistance. The proapoptotic protein NOXA marks an aggressive pancreatic ductal adenocarcinoma (PDAC) subtype. To identify drugs that unleash the death-inducing potential of NOXA, we performed an unbiased drug screening experiment. In NOXA-deficient isogenic cellular models, we identified an inhibitor of the transcription factor heterodimer CBFß/RUNX1. By genetic gain and loss of function experiments, we validated that the mode of action depends on RUNX1 and NOXA. Of note is that RUNX1 expression is significantly higher in PDACs compared to normal pancreas. We show that pharmacological RUNX1 inhibition significantly blocks tumor growth in vivo and in primary patient-derived PDAC organoids. Through genome-wide analysis, we detected that RUNX1-loss reshapes the epigenetic landscape, which gains H3K27ac enrichment at the NOXA promoter. Our study demonstrates a previously unknown mechanism of NOXA-dependent cell death, which can be triggered pharmaceutically. Therefore, our data show a way to target a therapy-resistant PDAC, an unmet clinical need.


Subject(s)
Apoptosis/genetics , Carcinoma, Pancreatic Ductal/genetics , Core Binding Factor Alpha 2 Subunit/genetics , Gene Expression , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Synthetic Lethal Mutations , Carcinoma, Pancreatic Ductal/pathology , Core Binding Factor Alpha 2 Subunit/antagonists & inhibitors , Humans , Pancreatic Neoplasms/pathology , Promoter Regions, Genetic , Up-Regulation
2.
Mol Cancer ; 23(1): 115, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38811992

ABSTRACT

BACKGROUND: We explored potential predictive biomarkers of immunotherapy response in patients with extensive-stage small-cell lung cancer (ES-SCLC) treated with durvalumab (D) + tremelimumab (T) + etoposide-platinum (EP), D + EP, or EP in the randomized phase 3 CASPIAN trial. METHODS: 805 treatment-naïve patients with ES-SCLC were randomized (1:1:1) to receive D + T + EP, D + EP, or EP. The primary endpoint was overall survival (OS). Patients were required to provide an archived tumor tissue block (or ≥ 15 newly cut unstained slides) at screening, if these samples existed. After assessment for programmed cell death ligand-1 expression and tissue tumor mutational burden, residual tissue was used for additional molecular profiling including by RNA sequencing and immunohistochemistry. RESULTS: In 182 patients with transcriptional molecular subtyping, OS with D ± T + EP was numerically highest in the SCLC-inflamed subtype (n = 10, median 24.0 months). Patients derived benefit from immunotherapy across subtypes; thus, additional biomarkers were investigated. OS benefit with D ± T + EP versus EP was greater with high versus low CD8A expression/CD8 cell density by immunohistochemistry, but with no additional benefit with D + T + EP versus D + EP. OS benefit with D + T + EP versus D + EP was associated with high expression of CD4 (median 25.9 vs. 11.4 months) and antigen-presenting and processing machinery (25.9 vs. 14.6 months) and MHC I and II (23.6 vs. 17.3 months) gene signatures, and with higher MHC I expression by immunohistochemistry. CONCLUSIONS: These findings demonstrate the tumor microenvironment is important in mediating better outcomes with D ± T + EP in ES-SCLC, with canonical immune markers associated with hypothesized immunotherapy mechanisms of action defining patient subsets that respond to D ± T. TRIAL REGISTRATION: ClinicalTrials.gov, NCT03043872.


Subject(s)
Biomarkers, Tumor , Immunotherapy , Lung Neoplasms , Small Cell Lung Carcinoma , Humans , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology , Small Cell Lung Carcinoma/immunology , Small Cell Lung Carcinoma/therapy , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/mortality , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/mortality , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Lung Neoplasms/metabolism , Female , Male , Immunotherapy/methods , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Middle Aged , Aged , Antibodies, Monoclonal/therapeutic use , Treatment Outcome , Neoplasm Staging , Antibodies, Monoclonal, Humanized/therapeutic use , Prognosis , Adult
3.
Haematologica ; 108(2): 555-567, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36134453

ABSTRACT

Aberrant activity of the SUMOylation pathway has been associated with MYC overexpression and poor prognosis in aggressive B-cell lymphoma (BCL) and other malignancies. Recently developed small-molecule inhibitors of SUMOylation (SUMOi) target the heterodimeric E1 SUMO activation complex (SAE1/UBA2). Here, we report that activated MYC signaling is an actionable molecular vulnerability in vitro and in a preclinical murine in vivo model of MYC-driven BCL. While SUMOi conferred direct effects on MYC-driven lymphoma cells, SUMO inhibition also resulted in substantial remodeling of various subsets of the innate and specific immunity in vivo. Specifically, SUMOi increased the number of memory B cells as well as cytotoxic and memory T cells, subsets that are attributed a key role within a coordinated anti-tumor immune response. In summary, our data constitute pharmacologic SUMOi as a powerful therapy in a subset of BCL causing massive remodeling of the normal B-cell and T-cell compartment.


Subject(s)
Lymphoma, B-Cell , Lymphoma , Humans , Mice , Animals , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Lymphoma/drug therapy , Lymphoma, B-Cell/drug therapy , Biomarkers , Ubiquitin-Activating Enzymes/metabolism
4.
Mol Cell ; 56(1): 116-27, 2014 Oct 02.
Article in English | MEDLINE | ID: mdl-25242142

ABSTRACT

Exposure of cells to reactive oxygen species (ROS) causes a rapid and significant drop in intracellular ATP levels. This energy depletion negatively affects ATP-dependent chaperone systems, making ROS-mediated protein unfolding and aggregation a potentially very challenging problem. Here we show that Get3, a protein involved in ATP-dependent targeting of tail-anchored (TA) proteins under nonstress conditions, turns into an effective ATP-independent chaperone when oxidized. Activation of Get3's chaperone function, which is a fully reversible process, involves disulfide bond formation, metal release, and its conversion into distinct, higher oligomeric structures. Mutational studies demonstrate that the chaperone activity of Get3 is functionally distinct from and likely mutually exclusive with its targeting function, and responsible for the oxidative stress-sensitive phenotype that has long been noted for yeast cells lacking functional Get3. These results provide convincing evidence that Get3 functions as a redox-regulated chaperone, effectively protecting eukaryotic cells against oxidative protein damage.


Subject(s)
Adenosine Triphosphatases/physiology , Guanine Nucleotide Exchange Factors/physiology , Oxidative Stress , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/metabolism , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/metabolism , Models, Biological , Molecular Chaperones/metabolism , Molecular Chaperones/physiology , Oxidation-Reduction , Protein Unfolding , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism
5.
Gesundheitswesen ; 84(12): 1158-1164, 2022 Dec.
Article in German | MEDLINE | ID: mdl-34560801

ABSTRACT

BACKGROUND: Supply bottlenecks in midwifery care could intensify due to increasing birth rates in Bavaria. Midwives are already fully booked and can only take care of pregnant women by increasing their working hours. The situation of self-employed midwives is particularly precarious. Bavaria has been supporting self-employed midwives by granting the Bavarian midwife bonus of up to € 1,000 per year since September 2018. This analysis aims to describe the current status of the unique Bavarian midwife bonus scheme and to describe current programmes to improve care in obstetrics in Bavaria. METHODS: All midwives who were self-employed, had their primary residence in Bavaria and had cared for at least four births in the application year were eligible for the Bavarian midwife bonus. We used data from application years 2017 and 2018 to describe the number of applications per year and the socio-demographic characteristics of the applicants and analyzed the regional distribution of the midwives and the regional supply density. RESULTS: In 2017, a total of 723 midwives received the bonus, in 2018 already 852 midwives. The average age of the applicants, who were exclusively female, was 44 years in the application year 2018. In 2018, 37 midwives per 100,000 women aged 15-44 were self-employed in obstetrics in Bavaria. A regional comparison showed an above-average supply density in the Upper Palatinate (52) and Lower Bavaria (54) and a below-average supply density in the Upper Franconia (13). CONCLUSION: The analysis of the Bavarian midwifery bonus enables a glance, albeit limited, into the supply situation of self-employed midwives in Bavaria. Whether the political instruments can improve the supply situation in Bavaria have to be evaluated using newly generated representative data. Central registration of midwives in Bavaria could be useful.


Subject(s)
Pregnancy , Female , Humans , Adult , Germany
6.
Br J Cancer ; 124(3): 531-538, 2021 02.
Article in English | MEDLINE | ID: mdl-33071285

ABSTRACT

An urgent medical need to develop novel treatment strategies for patients with pancreatic ductal adenocarcinoma (PDAC) exists. However, despite various efforts in the histopathological and molecular subtyping of PDAC, novel targeted or specific therapies have not been established. Posttranslational modifications (PTMs) with ubiquitin-like proteins, including small ubiquitin-like modifiers (SUMOs), mediate numerous processes that can contribute to the fitness and survival of cancer cells. The contribution of SUMOylation to transcriptional control, DNA repair pathways, mitotic progression, and oncogenic signalling has been described. Here we review functions of the SUMO pathway in PDAC, with a special focus on its connection to an aggressive subtype of the disease characterised by high MYC activity, and discuss SUMOylation inhibitors under development for precise PDAC therapies.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Pancreatic Neoplasms/metabolism , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-myc/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Survival/physiology , Cysteine Endopeptidases/drug effects , Cysteine Endopeptidases/metabolism , DNA Repair/physiology , Enzyme Inhibitors/pharmacology , Humans , Mitosis/physiology , Pancreatic Neoplasms/pathology , Poly-ADP-Ribose Binding Proteins/metabolism , Promyelocytic Leukemia Protein/metabolism , Protein Inhibitors of Activated STAT/metabolism , Proto-Oncogene Proteins c-myc/genetics , SUMO-1 Protein/antagonists & inhibitors , SUMO-1 Protein/metabolism , Signal Transduction/physiology , Small Ubiquitin-Related Modifier Proteins/antagonists & inhibitors , Sumoylation/drug effects , Synthetic Lethal Mutations , Ubiquitin-Conjugating Enzymes/antagonists & inhibitors , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitins/antagonists & inhibitors , Ubiquitins/metabolism
7.
Gut ; 69(8): 1472-1482, 2020 08.
Article in English | MEDLINE | ID: mdl-32001555

ABSTRACT

OBJECTIVE: Pancreatic ductal adenocarcinoma (PDAC) still carries a dismal prognosis with an overall 5-year survival rate of 9%. Conventional combination chemotherapies are a clear advance in the treatment of PDAC; however, subtypes of the disease exist, which exhibit extensive resistance to such therapies. Genomic MYC amplifications represent a distinct subset of PDAC with an aggressive tumour biology. It is clear that hyperactivation of MYC generates dependencies that can be exploited therapeutically. The aim of the study was to find and to target MYC-associated dependencies. DESIGN: We analysed human PDAC gene expression datasets. Results were corroborated by the analysis of the small ubiquitin-like modifier (SUMO) pathway in a large PDAC cohort using immunohistochemistry. A SUMO inhibitor was used and characterised using human and murine two-dimensional, organoid and in vivo models of PDAC. RESULTS: We observed that MYC is connected to the SUMOylation machinery in PDAC. Components of the SUMO pathway characterise a PDAC subtype with a dismal prognosis and we provide evidence that hyperactivation of MYC is connected to an increased sensitivity to pharmacological SUMO inhibition. CONCLUSION: SUMO inhibitor-based therapies should be further developed for an aggressive PDAC subtype.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins c-myc/genetics , Small Ubiquitin-Related Modifier Proteins/genetics , Small Ubiquitin-Related Modifier Proteins/metabolism , Aged , Animals , Apoptosis , Carcinoma, Pancreatic Ductal/drug therapy , Cell Line, Tumor , Cell Proliferation , Enzyme Inhibitors/pharmacology , Esters/pharmacology , Female , Gene Amplification , Gene Expression , Humans , Male , Mice , Middle Aged , Neoplasm Transplantation , Organoids/metabolism , Pancreatic Neoplasms/drug therapy , Prognosis , Proto-Oncogene Proteins c-myc/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , SUMO-1 Protein/genetics , SUMO-1 Protein/metabolism , Sulfonic Acids , Sumoylation/drug effects , Sumoylation/genetics , Transcriptome/drug effects , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitins/genetics , Ubiquitins/metabolism
8.
Bioinformatics ; 34(20): 3503-3510, 2018 10 15.
Article in English | MEDLINE | ID: mdl-29741575

ABSTRACT

Motivation: Transcriptional regulators play a major role in most biological processes. Alterations in their activities are associated with a variety of diseases and in particular with tumor development and progression. Hence, it is important to assess the effects of deregulated regulators on pathological processes. Results: Here, we present REGulator-Gene Association Enrichment (REGGAE), a novel method for the identification of key transcriptional regulators that have a significant effect on the expression of a given set of genes, e.g. genes that are differentially expressed between two sample groups. REGGAE uses a Kolmogorov-Smirnov-like test statistic that implicitly combines associations between regulators and their target genes with an enrichment approach to prioritize the influence of transcriptional regulators. We evaluated our method in two different application scenarios, which demonstrate that REGGAE is well suited for uncovering the influence of transcriptional regulators and is a valuable tool for the elucidation of complex regulatory mechanisms. Availability and implementation: REGGAE is freely available at https://regulatortrail.bioinf.uni-sb.de. Supplementary information: Supplementary data are available at Bioinformatics online.


Subject(s)
Gene Expression Regulation , Neoplasms/genetics , Transcription, Genetic , Female , Humans , Probability , Software
9.
Br J Haematol ; 179(5): 724-738, 2017 12.
Article in English | MEDLINE | ID: mdl-29171017

ABSTRACT

Identifying and therapeutically targeting cancer cell liabilities is of utmost importance in order to improve the treatment of patients with malignancies of poor prognosis. The MYC family genes (MYC, MYCN and MYCL) are among the most deregulated proto-oncogenes in human cancer. Aberrant MYC expression is frequently associated with poor prognosis. Although many aspects of MYC-mediated tumour biology are well characterized, there are currently no effective means for targeting MYC in a specific manner that have been established for clinical use. This review first discusses the role of MYC in the pathogenesis of haematopoietic malignancies, and secondly summarizes how insight into MYC functions could be translated into therapeutic approaches. In particular, we will address the possibilities of taking advantage of MYC-induced cancer cell vulnerabilities that could be exploited in terms of synthetic lethal interactions.


Subject(s)
Hematologic Neoplasms/genetics , Molecular Targeted Therapy/methods , Proto-Oncogene Proteins c-myc/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Genetic Predisposition to Disease , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/metabolism , Humans , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/genetics , Proto-Oncogene Proteins c-myc/metabolism
10.
Haematologica ; 101(8): 932-40, 2016 08.
Article in English | MEDLINE | ID: mdl-27175029

ABSTRACT

Acute myeloid leukemia originates from leukemia-initiating cells that reside in the protective bone marrow niche. CXCR4/CXCL12 interaction is crucially involved in recruitment and retention of leukemia-initiating cells within this niche. Various drugs targeting this pathway have entered clinical trials. To evaluate CXCR4 imaging in acute myeloid leukemia, we first tested CXCR4 expression in patient-derived primary blasts. Flow cytometry revealed that high blast counts in patients with acute myeloid leukemia correlate with high CXCR4 expression. The wide range of CXCR4 surface expression in patients was reflected in cell lines of acute myeloid leukemia. Next, we evaluated the CXCR4-specific peptide Pentixafor by positron emission tomography imaging in mice harboring CXCR4 positive and CXCR4 negative leukemia xenografts, and in 10 patients with active disease. [(68)Ga]Pentixafor-positron emission tomography showed specific measurable disease in murine CXCR4 positive xenografts, but not when CXCR4 was knocked out with CRISPR/Cas9 gene editing. Five of 10 patients showed tracer uptake correlating well with leukemia infiltration assessed by magnetic resonance imaging. The mean maximal standard uptake value was significantly higher in visually CXCR4 positive patients compared to CXCR4 negative patients. In summary, in vivo molecular CXCR4 imaging by means of positron emission tomography is feasible in acute myeloid leukemia. These data provide a framework for future diagnostic and theranostic approaches targeting the CXCR4/CXCL12-defined leukemia-initiating cell niche.


Subject(s)
Coordination Complexes , Gene Expression , Leukemia, Myeloid, Acute/diagnostic imaging , Leukemia, Myeloid, Acute/genetics , Molecular Imaging , Peptides, Cyclic , Positron-Emission Tomography , Receptors, CXCR4/genetics , Animals , Cell Line, Tumor , Disease Models, Animal , Gene Knockout Techniques , Gene Targeting , Humans , Leukemia, Myeloid, Acute/pathology , Magnetic Resonance Imaging/methods , Molecular Imaging/methods , Positron-Emission Tomography/methods , Receptors, CXCR4/metabolism , Xenograft Model Antitumor Assays
11.
Sci Rep ; 14(1): 12129, 2024 05 27.
Article in English | MEDLINE | ID: mdl-38802399

ABSTRACT

Many targeted cancer therapies rely on biomarkers assessed by scoring of immunohistochemically (IHC)-stained tissue, which is subjective, semiquantitative, and does not account for expression heterogeneity. We describe an image analysis-based method for quantitative continuous scoring (QCS) of digital whole-slide images acquired from baseline human epidermal growth factor receptor 2 (HER2) IHC-stained breast cancer tissue. Candidate signatures for patient stratification using QCS of HER2 expression on subcellular compartments were identified, addressing the spatial distribution of tumor cells and tumor-infiltrating lymphocytes. Using data from trastuzumab deruxtecan-treated patients with HER2-positive and HER2-negative breast cancer from a phase 1 study (NCT02564900; DS8201-A-J101; N = 151), QCS-based patient stratification showed longer progression-free survival (14.8 vs 8.6 months) with higher prevalence of patient selection (76.4 vs 56.9%) and a better cross-validated log-rank p value (0.026 vs 0.26) than manual scoring based on the American Society of Clinical Oncology / College of American Pathologists guidelines. QCS-based features enriched the HER2-negative subgroup by correctly predicting 20 of 26 responders.


Subject(s)
Breast Neoplasms , Patient Selection , Receptor, ErbB-2 , Trastuzumab , Humans , Female , Receptor, ErbB-2/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Trastuzumab/therapeutic use , Middle Aged , Biomarkers, Tumor/metabolism , Adult , Immunoconjugates/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Aged , Immunohistochemistry , Camptothecin/analogs & derivatives
12.
Sci Signal ; 17(824): eadc9662, 2024 02 20.
Article in English | MEDLINE | ID: mdl-38377177

ABSTRACT

The IL-6-gp130-STAT3 signaling axis is a major regulator of inflammation. Activating mutations in the gene encoding gp130 and germline gain-of-function mutations in STAT3 (STAT3GOF) are associated with multi-organ autoimmunity, severe morbidity, and adverse prognosis. To dissect crucial cellular subsets and disease biology involved in activated gp130 signaling, the gp130-JAK-STAT3 axis was constitutively activated using a transgene, L-gp130, specifically targeted to T cells. Activating gp130 signaling in T cells in vivo resulted in fatal, early onset, multi-organ autoimmunity in mice that resembled human STAT3GOF disease. Female mice had more rapid disease progression than male mice. On a cellular level, gp130 signaling induced the activation and effector cell differentiation of T cells, promoted the expansion of T helper type 17 (TH17) cells, and impaired the activity of regulatory T cells. Transcriptomic profiling of CD4+ and CD8+ T cells from these mice revealed commonly dysregulated genes and a gene signature that, when applied to human transcriptomic data, improved the segregation of patients with transcriptionally diverse STAT3GOF mutations from healthy controls. The findings demonstrate that increased gp130-STAT3 signaling leads to TH17-driven autoimmunity that phenotypically resembles human STAT3GOF disease.


Subject(s)
Autoimmunity , CD8-Positive T-Lymphocytes , Humans , Male , Female , Mice , Animals , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/metabolism , Autoimmunity/genetics , CD8-Positive T-Lymphocytes/metabolism , Signal Transduction , Inflammation , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
13.
EMBO Mol Med ; 15(9): e16431, 2023 09 11.
Article in English | MEDLINE | ID: mdl-37485814

ABSTRACT

The DNA damage response (DDR) acts as a barrier to malignant transformation and is often impaired during tumorigenesis. Exploiting the impaired DDR can be a promising therapeutic strategy; however, the mechanisms of inactivation and corresponding biomarkers are incompletely understood. Starting from an unbiased screening approach, we identified the SMC5-SMC6 Complex Localization Factor 2 (SLF2) as a regulator of the DDR and biomarker for a B-cell lymphoma (BCL) patient subgroup with an adverse prognosis. SLF2-deficiency leads to loss of DDR factors including Claspin (CLSPN) and consequently impairs CHK1 activation. In line with this mechanism, genetic deletion of Slf2 drives lymphomagenesis in vivo. Tumor cells lacking SLF2 are characterized by a high level of DNA damage, which leads to alterations of the post-translational SUMOylation pathway as a safeguard. The resulting co-dependency confers synthetic lethality to a clinically applicable SUMOylation inhibitor (SUMOi), and inhibitors of the DDR pathway act highly synergistic with SUMOi. Together, our results identify SLF2 as a DDR regulator and reveal co-targeting of the DDR and SUMOylation as a promising strategy for treating aggressive lymphoma.


Subject(s)
DNA Damage , Lymphoma, B-Cell , Humans , Adaptor Proteins, Signal Transducing , B-Lymphocytes , DNA Repair , Lymphoma, B-Cell/genetics
14.
Blood Adv ; 7(4): 469-481, 2023 02 28.
Article in English | MEDLINE | ID: mdl-35917568

ABSTRACT

Proteasome inhibition is a highly effective treatment for multiple myeloma (MM). However, virtually all patients develop proteasome inhibitor resistance, which is associated with a poor prognosis. Hyperactive small ubiquitin-like modifier (SUMO) signaling is involved in both cancer pathogenesis and cancer progression. A state of increased SUMOylation has been associated with aggressive cancer biology. We found that relapsed/refractory MM is characterized by a SUMO-high state, and high expression of the SUMO E1-activating enzyme (SAE1/UBA2) is associated with poor overall survival. Consistently, continuous treatment of MM cell lines with carfilzomib (CFZ) enhanced SUMO pathway activity. Treatment of MM cell lines with the SUMO E1-activating enzyme inhibitor subasumstat (TAK-981) showed synergy with CFZ in both CFZ-sensitive and CFZ-resistant MM cell lines, irrespective of the TP53 state. Combination therapy was effective in primary MM cells and in 2 murine MM xenograft models. Mechanistically, combination treatment with subasumstat and CFZ enhanced genotoxic and proteotoxic stress, and induced apoptosis was associated with activity of the prolyl isomerase PIN1. In summary, our findings reveal activated SUMOylation as a therapeutic target in MM and point to combined SUMO/proteasome inhibition as a novel and potent strategy for the treatment of proteasome inhibitor-resistant MM.


Subject(s)
Multiple Myeloma , Proteasome Inhibitors , Humans , Animals , Mice , Proteasome Inhibitors/pharmacology , Proteasome Inhibitors/therapeutic use , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Sumoylation , Proteasome Endopeptidase Complex/metabolism , Apoptosis , Ubiquitin-Activating Enzymes/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/pharmacology
15.
Blood Adv ; 6(2): 515-520, 2022 01 25.
Article in English | MEDLINE | ID: mdl-34768284

ABSTRACT

Biomarkers that predict response to lenalidomide maintenance therapy in patients with multiple myeloma (MM) have remained elusive. We have shown that immunomodulatory drugs (IMiDs) exert anti-MM activity via destabilization of MCT1 and CD147. In this study, cell samples of 654 patients with MM who received lenalidomide (n = 455), thalidomide (n = 98), or bortezomib (n = 101) maintenance were assessed by gene expression profiling and RNA sequencing, followed by correlation of MCT1 and CD147 expression with data for progression-free survival (PFS) and overall survival (OS). Patients with high expression levels of MCT1 showed significantly reduced PFS (31.9 months vs 48.2 months in MCT1high vs MCT1low; P = .03) and OS (75.9 months vs not reached [NR] in MCT1high vs MCT1low; P = .001) in cases with lenalidomide maintenance, whereas MCT1 expression had no significant impact on PFS or OS in cases with bortezomib maintenance. We validated the predictive role of MCT1 for IMiD-based maintenance in an independent cohort of patients who received thalidomide (OS, 83.6 months vs NR in MCT1high vs MCT1low; P = .03). Functional validation showed that MCT1 overexpression in human MM cell lines significantly reduced the efficacy of lenalidomide, whereas no change was observed with bortezomib treatment, either in vitro or in a MM xenograft model. Our findings have established MCT1 expression as a predictive marker for response to lenalidomide-based maintenance in patients with MM.


Subject(s)
Multiple Myeloma , Biomarkers , Bortezomib/pharmacology , Bortezomib/therapeutic use , Humans , Lenalidomide/therapeutic use , Multiple Myeloma/therapy , Thalidomide/pharmacology , Thalidomide/therapeutic use
16.
J Clin Invest ; 132(9)2022 05 02.
Article in English | MEDLINE | ID: mdl-35499080

ABSTRACT

Activated SUMOylation is a hallmark of cancer. Starting from a targeted screening for SUMO-regulated immune evasion mechanisms, we identified an evolutionarily conserved function of activated SUMOylation, which attenuated the immunogenicity of tumor cells. Activated SUMOylation allowed cancer cells to evade CD8+ T cell-mediated immunosurveillance by suppressing the MHC class I (MHC-I) antigen-processing and presentation machinery (APM). Loss of the MHC-I APM is a frequent cause of resistance to cancer immunotherapies, and the pharmacological inhibition of SUMOylation (SUMOi) resulted in reduced activity of the transcriptional repressor scaffold attachment factor B (SAFB) and induction of the MHC-I APM. Consequently, SUMOi enhanced the presentation of antigens and the susceptibility of tumor cells to CD8+ T cell-mediated killing. Importantly, SUMOi also triggered the activation of CD8+ T cells and thereby drove a feed-forward loop amplifying the specific antitumor immune response. In summary, we showed that activated SUMOylation allowed tumor cells to evade antitumor immunosurveillance, and we have expanded the understanding of SUMOi as a rational therapeutic strategy for enhancing the efficacy of cancer immunotherapies.


Subject(s)
Antigen Presentation , Neoplasms , Histocompatibility Antigens Class I , Humans , Immune Evasion , Neoplasms/pathology , Sumoylation
17.
Nat Commun ; 13(1): 281, 2022 01 12.
Article in English | MEDLINE | ID: mdl-35022408

ABSTRACT

SUMOylation is a post-translational modification of proteins that regulates these proteins' localization, turnover or function. Aberrant SUMOylation is frequently found in cancers but its origin remains elusive. Using a genome-wide transposon mutagenesis screen in a MYC-driven B-cell lymphoma model, we here identify the SUMO isopeptidase (or deconjugase) SENP6 as a tumor suppressor that links unrestricted SUMOylation to tumor development and progression. Notably, SENP6 is recurrently deleted in human lymphomas and SENP6 deficiency results in unrestricted SUMOylation. Mechanistically, SENP6 loss triggers release of DNA repair- and genome maintenance-associated protein complexes from chromatin thereby impairing DNA repair in response to DNA damages and ultimately promoting genomic instability. In line with this hypothesis, SENP6 deficiency drives synthetic lethality to Poly-ADP-Ribose-Polymerase (PARP) inhibition. Together, our results link SENP6 loss to defective genome maintenance and reveal the potential therapeutic application of PARP inhibitors in B-cell lymphoma.


Subject(s)
Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Mutation , Sumoylation/physiology , Animals , Biomarkers, Tumor , Carbon-Nitrogen Lyases/genetics , Carbon-Nitrogen Lyases/metabolism , Chromatin , DNA Damage/drug effects , DNA Repair/drug effects , Female , Genomic Instability , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/drug effects , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Protein Processing, Post-Translational , Sumoylation/drug effects , Sumoylation/genetics , Synthetic Lethal Mutations , Xenograft Model Antitumor Assays
18.
Leukemia ; 35(10): 2895-2905, 2021 10.
Article in English | MEDLINE | ID: mdl-34363012

ABSTRACT

Aberrant CXCR4 activity has been implicated in lymphoma pathogenesis, disease progression, and resistance to therapies. Using a mouse model with a gain-of-function CXCR4 mutation (CXCR4C1013G) that hyperactivates CXCR4 signaling, we identified CXCR4 as a crucial activator of multiple key oncogenic pathways. CXCR4 hyperactivation resulted in an expansion of transitional B1 lymphocytes, which represent the precursors of chronic lymphocytic leukemia (CLL). Indeed, CXCR4 hyperactivation led to a significant acceleration of disease onset and a more aggressive phenotype in the murine Eµ-TCL1 CLL model. Hyperactivated CXCR4 signaling cooperated with TCL1 to cause a distinct oncogenic transcriptional program in B cells, characterized by PLK1/FOXM1-associated pathways. In accordance, Eµ-TCL1;CXCR4C1013G B cells enriched a transcriptional signature from patients with Richter's syndrome, an aggressive transformation of CLL. Notably, MYC activation in aggressive lymphoma was associated with increased CXCR4 expression. In line with this finding, additional hyperactive CXCR4 signaling in the Eµ-Myc mouse, a model of aggressive B-cell cancer, did not impact survival. In summary, we here identify CXCR4 hyperactivation as a co-driver of an aggressive lymphoma phenotype.


Subject(s)
Cell Cycle Proteins/metabolism , Forkhead Box Protein M1/metabolism , Gene Expression Regulation, Leukemic , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Mutation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/physiology , Receptors, CXCR4/metabolism , Animals , Cell Cycle Proteins/genetics , Disease Progression , Female , Forkhead Box Protein M1/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Receptors, CXCR4/genetics , Polo-Like Kinase 1
19.
Cancers (Basel) ; 12(12)2020 Dec 14.
Article in English | MEDLINE | ID: mdl-33327527

ABSTRACT

Multiple myeloma is a genetically heterogeneous plasma cell malignancy characterized by organ damage and a massive production of (in-)complete monoclonal antibodies. Coping with protein homeostasis and post-translational regulation is therefore essential for multiple myeloma cells to survive. Furthermore, post-translational modifications such as ubiquitination and SUMOylation play key roles in essential pathways in multiple myeloma, including NFκB signaling, epigenetic regulation, as well as DNA damage repair. Drugs modulating the ubiquitin-proteasome system, such as proteasome inhibitors and thalidomide analogs, are approved and highly effective drugs in multiple myeloma. In this review, we focus on ubiquitin and ubiquitin-like modifications in the biology and current developments of new treatments for multiple myeloma.

20.
Biomedicines ; 8(12)2020 Dec 09.
Article in English | MEDLINE | ID: mdl-33317212

ABSTRACT

The reliable authentication of cell lines is a prerequisite for the reproducibility and replicability of experiments. A common method of cell line authentication is the fragment length analysis (FLA) of short-tandem repeats (STR) by capillary electrophoresis. However, this technique is not always accessible and is often costly. Using a microfluidic electrophoresis system, we analyzed the quality and integrity of different murine cell lines by STR profiling. As a proof of concept, we isolated and immortalized hematopoietic progenitor cells (HPC) of various genotypes through retroviral transduction of the fusion of the estrogen receptor hormone-binding domain with the coding sequence of HoxB8. Cell lines were maintained in the HPC state with Flt3 ligand (FL) and estrogen treatment and could be characterized upon differentiation. In a validation cohort, we applied this technique on primary mutant Kras-driven pancreatic cancer cell lines, which again allowed for clear discrimination. In summary, our study provides evidence that FLA of STR-amplicons by microfluidic electrophoresis allows for stringent quality control and the tracking of cross-contaminations in both genetically stable HPC lines and cancer cell lines, making it a simple and cost-efficient alternative to traditional capillary electrophoresis.

SELECTION OF CITATIONS
SEARCH DETAIL