33 results on '"Gjerstorff, Morten F"'
Search Results
2. MCM3 upregulation confers endocrine resistance in breast cancer and is a predictive marker of diminished tamoxifen benefit.
- Author
-
Løkkegaard, Sanne, Elias, Daniel, Alves, Carla L, Bennetzen, Martin V, Lænkholm, Anne-Vibeke, Bak, Martin, Gjerstorff, Morten F, Johansen, Lene E, Vever, Henriette, Bjerre, Christina, Kirkegaard, Tove, Nordenskjöld, Bo, Fornander, Tommy, Stål, Olle, Lindström, Linda S, Esserman, Laura J, Lykkesfeldt, Anne E, Andersen, Jens S, Leth-Larsen, Rikke, and Ditzel, Henrik J
- Abstract
Resistance to endocrine therapy in estrogen receptor-positive (ER+) breast cancer is a major clinical problem with poorly understood mechanisms. There is an unmet need for prognostic and predictive biomarkers to allow appropriate therapeutic targeting. We evaluated the mechanism by which minichromosome maintenance protein 3 (MCM3) influences endocrine resistance and its predictive/prognostic potential in ER+ breast cancer. We discovered that ER+ breast cancer cells survive tamoxifen and letrozole treatments through upregulation of minichromosome maintenance proteins (MCMs), including MCM3, which are key molecules in the cell cycle and DNA replication. Lowering MCM3 expression in endocrine-resistant cells restored drug sensitivity and altered phosphorylation of cell cycle regulators, including p53(Ser315,33), CHK1(Ser317), and cdc25b(Ser323), suggesting that the interaction of MCM3 with cell cycle proteins is an important mechanism of overcoming replicative stress and anti-proliferative effects of endocrine treatments. Interestingly, the MCM3 levels did not affect the efficacy of growth inhibitory by CDK4/6 inhibitors. Evaluation of MCM3 levels in primary tumors from four independent cohorts of breast cancer patients receiving adjuvant tamoxifen mono-therapy or no adjuvant treatment, including the Stockholm tamoxifen (STO-3) trial, showed MCM3 to be an independent prognostic marker adding information beyond Ki67. In addition, MCM3 was shown to be a predictive marker of response to endocrine treatment. Our study reveals a coordinated signaling network centered around MCM3 that limits response to endocrine therapy in ER+ breast cancer and identifies MCM3 as a clinically useful prognostic and predictive biomarker that allows personalized treatment of ER+ breast cancer patients.
- Published
- 2021
3. Stochastic demethylation and redundant epigenetic suppressive mechanisms generate highly heterogeneous responses to pharmacological DNA methyltransferase inhibition.
- Author
-
Jakobsen, Mie K., Traynor, Sofie, Nielsen, Aaraby Y., Dahl, Christina, Staehr, Mette, Jakobsen, Simon T., Madsen, Maria S., Siersbaek, Rasmus, Terp, Mikkel G., Jensen, Josefine B., Pedersen, Christina B., Shrestha, Anup, Brewer, Jonathan R., Duijf, Pascal H. G., Gammelgaard, Odd L., Ditzel, Henrik J., Kirkin, Alexei F., Guldberg, Per, and Gjerstorff, Morten F.
- Subjects
CANCER cell culture ,HISTONE deacetylase inhibitors ,DNA methylation ,LIFE sciences ,GENETIC variation ,DNA methyltransferases - Abstract
Background: Despite promising preclinical studies, the application of DNA methyltransferase inhibitors in treating patients with solid cancers has thus far produced only modest outcomes. The presence of intratumoral heterogeneity in response to DNA methyltransferase inhibitors could significantly influence clinical efficacy, yet our understanding of the single-cell response to these drugs in solid tumors remains very limited. Methods: In this study, we used cancer/testis antigen genes as a model for methylation-dependent gene expression to examine the activity of DNA methyltransferase inhibitors and their potential synergistic effect with histone deacetylase inhibitors at the single-cancer cell level. The analysis was performed on breast cancer patient-derived xenograft tumors and cell lines, employing a comprehensive set of techniques, including targeted single-cell mRNA sequencing. Mechanistic insights were further gained through DNA methylation profiling and chromatin structure analysis. Results: We show that breast cancer tumors and cell cultures exhibit a highly heterogenous response to DNA methyltransferase inhibitors, persisting even under high drug concentrations and efficient DNA methyltransferase depletion. The observed variability in response to DNA methyltransferase inhibitors was independent of cancer-associated aberrations and clonal genetic diversity. Instead, these variations were attributed to stochastic demethylation of regulatory CpG sites and the DNA methylation-independent suppressive function of histone deacetylases. Conclusions: Our findings point to intratumoral heterogeneity as a limiting factor in the use of DNA methyltransferase inhibitors as single agents in treatment of solid cancers and highlight histone deacetylase inhibitors as essential partners to DNA methyltransferase inhibitors in the clinic. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
4. SSX2 promotes the formation of a novel type of intranuclear lamin bodies
- Author
-
Traynor, S., Jakobsen, N.D., Ebbesen, M.F., Bennedsen, S.N., Johansen, S., Ebstrup, M.L., Pedersen, C.B., Ditzel, H.J., Brewer, J.R., and Gjerstorff, Morten F.
- Published
- 2022
- Full Text
- View/download PDF
5. MiR-142-3p targets HMGA2 and suppresses breast cancer malignancy
- Author
-
Mansoori, Behzad, Duijf, Pascal H.G., Mohammadi, Ali, Safarzadeh, Elham, Ditzel, Henrik J., Gjerstorff, Morten F., Cho, William Chi-Shing, and Baradaran, Behzad
- Published
- 2021
- Full Text
- View/download PDF
6. Augmenting engineered T-cell strategies in solid cancers through epigenetic priming
- Author
-
Nielsen, Aaraby Y., Ormhøj, Maria, Traynor, Sofie, and Gjerstorff, Morten F.
- Published
- 2020
- Full Text
- View/download PDF
7. Photodynamic therapy for cancer: Role of natural products
- Author
-
Mansoori, Behzad, Mohammadi, Ali, Amin Doustvandi, Mohammad, Mohammadnejad, Fatemeh, Kamari, Farzin, Gjerstorff, Morten F., Baradaran, Behzad, and Hamblin, Michael R.
- Published
- 2019
- Full Text
- View/download PDF
8. A functional genetic screen identifies the Mediator complex as essential for SSX2-induced senescence
- Author
-
Brückmann, Nadine H., Bennedsen, Sofie N., Duijf, Pascal H. G., Terp, Mikkel G., Thomassen, Mads, Larsen, Martin, Pedersen, Christina B., Kruse, Torben, Alcaraz, Nicolas, Ditzel, Henrik J., and Gjerstorff, Morten F.
- Published
- 2019
- Full Text
- View/download PDF
9. Ectopic expression of cancer/testis antigen SSX2 induces DNA damage and promotes genomic instability
- Author
-
Greve, Katrine B.V., Lindgreen, Jonas N., Terp, Mikkel G., Pedersen, Christina B., Schmidt, Steffen, Mollenhauer, Jan, Kristensen, Stine B., Andersen, Rikke S., Relster, Mette M., Ditzel, Henrik J., and Gjerstorff, Morten F.
- Published
- 2015
- Full Text
- View/download PDF
10. Expression, purification and characterization of the cancer-germline antigen GAGE12I: A candidate for cancer immunotherapy
- Author
-
Gjerstorff, Morten F., Besir, Hüseyin, Larsen, Martin R., and Ditzel, Henrik J.
- Published
- 2010
- Full Text
- View/download PDF
11. PDX Models: A Versatile Tool for Studying the Role of Myeloid-Derived Suppressor Cells in Breast Cancer.
- Author
-
Gjerstorff, Morten F., Traynor, Sofie, Gammelgaard, Odd L., Johansen, Simone, Pedersen, Christina B., Ditzel, Henrik J., and Terp, Mikkel G.
- Subjects
- *
XENOGRAFTS , *IN vivo studies , *MATHEMATICAL models , *ANIMAL experimentation , *MYELOID-derived suppressor cells , *THEORY , *BREAST tumors , *MICE - Abstract
Simple Summary: Myeloid-derived suppressive cells (MDSCs) are important for the progression of human tumors and represent potential targets for novel therapeutic strategies in breast and other cancers. To develop such strategies, pre-clinical models mimicking patient tumors are needed. In this study, we demonstrate that tumor models established by transplanting breast cancer patient tumor biopsies into the mammary tissues of mice represent excellent tools for studying and targeting MDSCs. Using molecular and genetic analyses, we show that these patient-derived xenograft (PDX) tumors produce signaling proteins that actively recruit MDSCs to the tumors, with differences between models that reflect those seen in breast cancer patients. Furthermore, MDSCs were associated with the spread of cancer cells to distant sites in the mice, similar to what is observed in patients. In conclusion, the use of breast cancer PDX models may greatly facilitate the development of novel therapeutic strategies for breast cancer and other cancer types. The pivotal role of myeloid-derived suppressive cells (MDSCs) in cancer has become increasingly apparent over the past few years. However, to fully understand how MDSCs can promote human tumor progression and to develop strategies to target this cell type, relevant models that closely resemble the clinical complexity of human tumors are needed. Here, we show that mouse MDSCs of both the monocytic (M-MDCS) and the granulocytic (PMN-MDSC) lineages are recruited to human breast cancer patient-derived xenograft (PDX) tumors in mice. Transcriptomic analysis of FACS-sorted MDSC-subpopulations from the PDX tumors demonstrated the expression of several MDSC genes associated with both their mobilization and immunosuppressive function, including S100A8/9, Ptgs2, Stat3, and Cxcr2, confirming the functional identity of these cells. By combining FACS analysis, RNA sequencing, and immune florescence, we show that the extent and type of MDSC infiltration depend on PDX model intrinsic factors such as the expression of chemokines involved in mobilizing and recruiting tumor-promoting MDSCs. Interestingly, MDSCs have been shown to play a prominent role in breast cancer metastasis, and in this context, we demonstrate increased recruitment of MDSCs in spontaneous PDX lung metastases compared to the corresponding primary PDX tumors. We also demonstrate that T cell-induced inflammation enhances the recruitment of MDSC in experimental breast cancer metastases. In conclusion, breast cancer PDX models represent a versatile tool for studying molecular mechanisms that drive myeloid cell recruitment to primary and metastatic tumors and facilitate the development of innovative therapeutic strategies targeting these cells. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
12. Biweekly cetuximab and irinotecan as second-line therapy in patients with gastro-esophageal cancer previously treated with platinum
- Author
-
Schoennemann, Katrine R., Bjerregaard, Jon K., Hansen, Tine P., De Stricker, Karin, Gjerstorff, Morten F., Jensen, Helle A., Vestermark, Lene W., and Pfeiffer, Per
- Published
- 2011
- Full Text
- View/download PDF
13. Distinct GAGE and MAGE-A expression during early human development indicate specific roles in lineage differentiation
- Author
-
Gjerstorff, Morten F., Harkness, Linda, Kassem, Moustapha, Frandsen, Ulrik, Nielsen, Ole, Lutterodt, Melissa, Møllgård, Kjeld, and Ditzel, Henrik J.
- Published
- 2008
14. MAGE-A1, GAGE and NY-ESO-1 cancer/testis antigen expression during human gonadal development
- Author
-
Gjerstorff, Morten F., Kock, Kirsten, Nielsen, Ole, and Ditzel, Henrik J.
- Published
- 2007
15. Sustained compensatory p38 MAPK signaling following treatment with MAPK inhibitors induces the immunosuppressive protein CD73 in cancer: combined targeting could improve outcomes.
- Author
-
Terp, Mikkel G., Gammelgaard, Odd L., Vever, Henriette, Gjerstorff, Morten F., and Ditzel, Henrik J.
- Abstract
RAS‐MAPK signaling promotes immune evasion and cancer cell survival, and MAPK inhibitors (MAPKis) are frequently used as cancer therapies. Despite progress elucidating the direct effects of MAPKi on immune cells, their indirect effect on the tumor microenvironment (TME) through changes in tumor cells remains incompletely understood. Here, we present evidence of a rapid compensatory response to MAPKi that is driven by sustained p38 MAPK signaling and by which cancer cells can upregulate the immunosuppressive protein CD73 to reduce the antitumor immune response. This compensatory response also results in decreased sensitivity toward MAPKi, and, accordingly, combining anti‐CD73 antibodies and MAPKi significantly enhances the antitumor effect compared to single‐agent treatment in vivo. Combining MAPKi and anti‐CD73 was accompanied by significant alterations in intratumor immune cell composition, supporting the effect of MAPKi‐induced CD73 expression on the TME. We show that high CD73 expression significantly correlates with worse outcome in MAPKi‐treated colorectal cancer patients, highlighting the potential clinical importance of increased CD73 expression following MAPKi treatment. Our findings may explain the diminished effect of MAPKi in cancer patients and provides further rationale for combined anti‐CD73 and MAPKi treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
16. The Cancer/Testis Antigen Gene VCX2 Is Rarely Expressed in Malignancies but Can Be Epigenetically Activated Using DNA Methyltransferase and Histone Deacetylase Inhibitors.
- Author
-
Jakobsen, Mie K., Traynor, Sofie, Stæhr, Mette, Duijf, Pascal G., Nielsen, Aaraby Y., Terp, Mikkel G., Pedersen, Christina B., Guldberg, Per, Ditzel, Henrik J., and Gjerstorff, Morten F.
- Subjects
HISTONE deacetylase inhibitors ,DNA methyltransferases ,GONAD development ,TESTIS ,IMMUNOSTAINING ,ANTIGENS ,GERM cells - Abstract
Identification of novel tumor-specific targets is important for the future development of immunotherapeutic strategies using genetically engineered T cells or vaccines. In this study, we characterized the expression of VCX2, a member of the VCX/Y cancer/testis antigen family, in a large panel of normal tissues and tumors from multiple cancer types using immunohistochemical staining and RNA expression data. In normal tissues, VCX2 was detected in the germ cells of the testis at all stages of maturation but not in any somatic tissues. Among malignancies, VCX2 was only found in tumors of a small subset of melanoma patients and thus rarely expressed compared to other cancer/testis antigens such as GAGE and MAGE-A. The expression of VCX2 correlated with that of other VCX/Y genes. Importantly, we found that expression of VCX2 was inversely correlated with promoter methylation and could be activated by treatment with a DNA methyltransferase inhibitor in multiple breast cancer and melanoma cell lines and a breast cancer patient-derived xenograft. The effect could be further potentiated by combining the DNA methyltransferase inhibitor with a histone deacetylase inhibitor. Our results show that the expression of VCX2 can be epigenetically induced in cancer cells and therefore could be an attractive target for immunotherapy of cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
17. CAR T-Cell Cancer Therapy Targeting Surface Cancer/Testis Antigens.
- Author
-
Jakobsen, Mie K. and Gjerstorff, Morten F.
- Subjects
T cell receptors ,DECITABINE ,CANCER treatment ,ANTIGENS ,CYTOTOXIC T cells ,TUMOR antigens ,TESTIS ,CHIMERIC antigen receptors - Published
- 2020
- Full Text
- View/download PDF
18. HMGA2 and Bach‐1 cooperate to promote breast cancer cell malignancy.
- Author
-
Mansoori, Behzad, Mohammadi, Ali, Asadzadeh, Zahra, Shirjang, Solmaz, Minouei, Mahsa, Abedi Gaballu, Fereydoon, Shajari, Neda, Kazemi, Tohid, Gjerstorff, Morten F., Duijf, Pascal H.G., and Baradaran, Behzad
- Subjects
BREAST cancer ,CANCER ,CANCER cells ,CELL cycle ,CELL death - Abstract
During breast cancer progression, tumor cells acquire multiple malignant features. The transcription factors and cell cycle regulators high mobility group A2 (HMGA2) and BTB and CNC homology 1 (Bach‐1) are overexpressed in several cancers, but the mechanistic understanding of how HMGA2 and Bach‐1 promote cancer development has been limited. We found that HMGA2 and Bach‐1 are overexpressed in breast cancer tissues and their expression correlates positively in tumors but not in normal tissues. Individual HMGA2 or Bach‐1 knockdown downregulates expression of both proteins, suggesting a mutual stabilizing effect between the two proteins. Importantly, combined HMGA2 and Bach‐1 knockdown additively decrease cell proliferation, migration, epithelial‐to‐mesenchymal transition, and colony formation, while promoting apoptotic cell death via upregulation of caspase‐3 and caspase‐9. First the first time, we show that HMGA2 and Bach‐1 overexpression in tumors correlate positively and that the proteins cooperatively suppress a broad range of malignant cellular properties, such as proliferation, migration, clonogenicity, and evasion of apoptotic cell death. Thus, our observations suggest that combined targeting of HMGA2 and Bach1 may be an effective therapeutic strategy to treat breast cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
19. miR‐142‐3p is a tumor suppressor that inhibits estrogen receptor expression in ER‐positive breast cancer.
- Author
-
Mansoori, Behzad, Mohammadi, Ali, Gjerstorff, Morten F., Shirjang, Solmaz, Asadzadeh, Zahra, Khaze, Vahid, Holmskov, Uffe, Kazemi, Tohid, Duijf, Pascal H. G., and Baradaran, Behzad
- Subjects
ESTROGEN receptors ,BREAST cancer ,CELL migration inhibition ,WESTERN immunoblotting ,MESSENGER RNA ,CELL migration - Abstract
Estrogen receptors (ERs) are involved in the development of many types of malignant tumors, in particular, breast cancer. Among others, ERs affect cell growth, proliferation, and differentiation. The microRNA (miRNA) miR‐142‐3p has been shown to inhibit carcinogenesis by regulating various cellular processes, including cell cycle progression, cell migration, apoptosis, and invasion. It does so via targeting molecules involved in a range of signaling pathways. We surgically collected 20 ER‐positive breast cancer samples, each with matched adjacent normal breast tissue, and measured the expression of miR‐142‐3p via quantitative real‐time polymerase chain reaction (qRT‐PCR). Bioinformatics methods, luciferase reporter assay, qRT‐PCR, and western blot analysis were used to assess whether miR‐142‐3p could target ESR1, which encodes the estrogen receptor, in ER‐positive breast cancer cells and patient samples. We also restored miRNA expression and performed cell viability, cytotoxicity, and colony formation assays. Western blot analysis and qRT‐PCR were used to study the expression of apoptosis and stemness markers. We found that miR‐142‐3p is downregulated in ER‐positive breast cancers. Restoration of miR‐142‐3p expression in ER‐positive breast cancer cells reduced cell viability, induced apoptosis via the intrinsic pathway and decreased both colony formation and the expression of stem cell markers. Bioinformatic analysis predicted miR‐142‐3p could bind to 3′‐untranslated region ESR1 messenger RNA (mRNA). Consistently, we demonstrated that miR‐142‐3p reduced luciferase activity in ER‐positive breast cancer cells, and decreased ESR1 expression in both mRNA and protein levels. The results revealed miR‐142‐3p and ESR1 expression correlated negatively in ER‐positive breast cancer samples. The results suggest miR‐142‐3p acts as a tumor suppressor via multiple mechanisms. Thus, restoration of miR‐142‐3p expression, for example, via miRNA replacement therapy, may represent an effective strategy for the treatment of ER‐positive breast cancer patients. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
20. Remodeling and destabilization of chromosome 1 pericentromeric heterochromatin by SSX proteins.
- Author
-
Traynor, Sofie, Møllegaard, Niels Erik, Jørgensen, Mikkel G, Brückmann, Nadine H, Pedersen, Christina B, Terp, Mikkel G, Johansen, Simone, Dejardin, Jerome, Ditzel, Henrik J, and Gjerstorff, Morten F
- Published
- 2019
- Full Text
- View/download PDF
21. Adoptive cancer immunotherapy using DNA-demethylated T helper cells as antigen-presenting cells.
- Author
-
Kirkin, Alexei F., Dzhandzhugazyan, Karine N., Guldberg, Per, Jon Fang, Johnny, Andersen, Rikke S., Dahl, Christina, Mortensen, Jann, Lundby, Tim, Wagner, Aase, Law, Ian, Broholm, Helle, Madsen, Line, Lundell-Ek, Christer, Gjerstorff, Morten F., Ditzel, Henrik J., Jensen, Martin R., and Fischer, Walter
- Subjects
CYTOTOXIC T cells ,T helper cells ,KILLER cells ,DNA demethylation ,CANCER cells ,GLIOBLASTOMA multiforme - Abstract
In cancer cells, cancer/testis (CT) antigens become epigenetically derepressed through DNA demethylation and constitute attractive targets for cancer immunotherapy. Here we report that activated CD4
+ T helper cells treated with a DNA-demethylating agent express a broad repertoire of endogenous CT antigens and can be used as antigen-presenting cells to generate autologous cytotoxic T lymphocytes (CTLs) and natural killer cells. In vitro, activated CTLs induce HLA-restricted lysis of tumor cells of different histological types, as well as cells expressing single CT antigens. In a phase 1 trial of 25 patients with recurrent glioblastoma multiforme, cytotoxic lymphocytes homed to the tumor, with tumor regression ongoing in three patients for 14, 22, and 27 months, respectively. No treatment-related adverse effects were observed. This proof-of-principle study shows that tumor-reactive effector cells can be generated ex vivo by exposure to antigens induced by DNA demethylation, providing a novel, minimally invasive therapeutic strategy for treating cancer. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
22. Lack of ADAM2, CALR3 and SAGE1 Cancer/Testis Antigen Expression in Lung and Breast Cancer.
- Author
-
Maheswaran, Emeaga, Pedersen, Christina B., Ditzel, Henrik J., and Gjerstorff, Morten F.
- Subjects
BREAST cancer treatment ,IMMUNOTHERAPY ,ANTIGEN analysis ,IMMUNOGLOBULINS ,DNA methyltransferase regulation - Abstract
Immunotherapy is emerging as a supplement to conventional cancer treatment, and identifying antigen targets for specific types of cancer is critical to optimizing therapeutic efficacy. Cancer/testis antigens are highly promising targets for immunotherapy due to their cancer-specific expression and antigenic properties, but the expression patterns of most of the more than 200 identified cancer/testis antigens in various cancers remain largely uncharacterized. In this study, we investigated the expression of the cancer/testis antigens ADAM2, CALR3 and SAGE1 in lung and breast cancer, the two most frequent human cancers, with the purpose of providing novel therapeutic targets for these diseases. We used a set of previously uncharacterized antibodies against the cancer/testis antigens ADAM2, CALR3 and SAGE1 to investigate their expression in a large panel of normal tissues as well as breast and lung cancers. Staining for the well-characterized MAGE-A proteins was included for comparison. Immunohistochemical staining confirmed previous mRNA analysis demonstrating that ADAM2, CALR3 and SAGE1 proteins are confined to testis in normal individuals. Negative tissues included plancenta, which express many other CT antigens, such as MAGE-A proteins. Surprisingly, we detected no ADAM2, CALR3 and SAGE1 in the 67 lung cancers (mainly non-small lung cancer) and 189 breast cancers, while MAGE-A proteins were present in 15% and 7–16% of these tumor types, respectively. Treatment with DNA methyltransferase inhibitors has been proposed as an attractive strategy to increase the expression of cancer/testis antigens in tumors before immunotargeting; however, neither ADAM2, CALR3 nor SAGE1 could be significantly induced in lung and breast cancer cell lines using this strategy. Our results suggest that ADAM2, CALR3 and SAGE1 cancer/testis antigens are not promising targets for immunotherapy of breast and lung cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
23. Analysis of GAGE, NY-ESO-1 and SP17 cancer/testis antigen expression in early stage non-small cell lung carcinoma.
- Author
-
Gjerstorff, Morten F., Pøhl, Mette, Olsen, Karen E., and Ditzel, Henrik J.
- Subjects
- *
TUMOR antigens , *LUNG cancer patients , *BIOPSY , *IMMUNOHISTOCHEMISTRY , *REGRESSION analysis , *HISTOPATHOLOGY , *SQUAMOUS cell carcinoma - Abstract
Background: The unique expression pattern and immunogenic properties of cancer/testis antigens make them ideal targets for immunotherapy of cancer. The MAGE-A3 cancer/testis antigen is frequently expressed in non-small cell lung cancer (NSCLC) and vaccination with MAGE-A3 in patients with MAGE-A3-positive NSCLC has shown promising results. However, little is known about the expression of other cancer/testis antigens in NSCLC. In the present study the expression of cancer/testis antigens GAGE, NY-ESO-1 and SP17 was investigated in patients with completely resected, early stage, primary NSCLC.Methods: Tumor biopsies from normal lung tissue and from a large cohort (n = 169) of NSCLC patients were examined for GAGE, NY-ESO-1 and SP17 protein expression by immunohistochemical analysis. The expression of these antigens was further matched to clinical and pathological features using univariate cox regression analysis.Results: GAGE and NY-ESO-1 cancer/testis antigens were not expressed in normal lung tissue, while SP17 was expressed in ciliated lung epithelia. The frequency of GAGE, NY-ESO-1 and SP17 expression in NSCLC tumors were 26.0% (44/169), 11.8% (20/169) and 4.7% (8/169), respectively, and 33.1% (56/169) of the tumors expressed at least one of these antigens. In general, the expression of GAGE, NY-ESO-1 and SP17 was not significantly associated with a specific histotype (adenocarcinoma vs. squamous cell carcinoma), but high-level GAGE expression (>50%) was more frequent in squamous cell carcinoma (p = 0.02). Furthermore, the frequency of GAGE expression was demonstrated to be significantly higher in stage II-IIIa than stage I NSCLC (17.0% vs. 35.8%; p = 0.02). Analysis of the relation between tumor expression of GAGE and NY-ESO-1 and survival endpoints revealed no significant associations.Conclusion: Our study demonstrates that GAGE, NY-ESO-1 and SP17 cancer/testis antigens are candidate targets for immunotherapy of NSCLC and further suggest that multi-antigen vaccines may be beneficial. [ABSTRACT FROM AUTHOR]- Published
- 2013
- Full Text
- View/download PDF
24. GAGE Cancer-Germline Antigens Are Recruited to the Nuclear Envelope by Germ Cell-Less (GCL).
- Author
-
Gjerstorff, Morten F., Rósner, Heike I., Pedersen, Christina B., Greve, Katrine B. V., Schmidt, Steffen, Wilson, Katherine L., Mollenhauer, Jan, Besir, Hüseyin, Poulsen, Flemming M., Møllegaard, Niels Erik, Ditzel, Henrik J., and Permyakov, Eugene A.
- Subjects
- *
PROTEINS , *GERM cells , *NUCLEAR membranes , *MESSENGER RNA , *CANCER cells , *CHROMATIN - Abstract
GAGE proteins are highly similar, primate-specific molecules with unique primary structure and undefined cellular roles. They are restricted to cells of the germ line in adult healthy individuals, but are broadly expressed in a wide range of cancers. In a yeast two-hybrid screen we identified the metazoan transcriptional regulator, Germ cell-less (GCL), as an nteraction partner of GAGE12I. GCL directly binds LEM-domain proteins (LAP2β, emerin, MAN1) at the nuclear envelope, and we found that GAGE proteins were recruited to the nuclear envelope inner membrane by GCL. Based on yeast two- hybrid analysis and pull-down experiments of GCL polypeptides, GCL residues 209-320 (which includes the BACK domain) were deduced sufficient for association with GAGE proteins. GAGE mRNAs and GCL mRNA were demonstrated in human testis and most types of cancers, and at the protein level GAGE members and GCL were co-expressed in cancer cell lines. Structural studies of GAGE proteins revealed no distinct secondary or tertiary structure, suggesting they are intrinsically disordered. Interestingly GAGE proteins formed stable complexes with dsDNA in vitro at physiological concentrations, and GAGE12I bound several different dsDNA fragments, suggesting sequence-nonspecific binding. Dual association of GAGE family members with GCL at the nuclear envelope inner membrane in cells, and with dsDNA in vitro, implicate GAGE proteins in chromatin regulation in germ cells and cancer cells. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
25. HMGA2 as a Critical Regulator in Cancer Development.
- Author
-
Mansoori, Behzad, Mohammadi, Ali, Ditzel, Henrik J., Duijf, Pascal H. G., Khaze, Vahid, Gjerstorff, Morten F., Baradaran, Behzad, and Machín, Félix
- Subjects
HIGH mobility group proteins ,EMBRYONIC stem cells ,CANCER cell proliferation ,CELL cycle ,DNA repair ,ONCOGENES - Abstract
The high mobility group protein 2 (HMGA2) regulates gene expression by binding to AT-rich regions of DNA. Akin to other DNA architectural proteins, HMGA2 is highly expressed in embryonic stem cells during embryogenesis, while its expression is more limited at later stages of development and in adulthood. Importantly, HMGA2 is re-expressed in nearly all human malignancies, where it promotes tumorigenesis by multiple mechanisms. HMGA2 increases cancer cell proliferation by promoting cell cycle entry and inhibition of apoptosis. In addition, HMGA2 influences different DNA repair mechanisms and promotes epithelial-to-mesenchymal transition by activating signaling via the MAPK/ERK, TGFβ/Smad, PI3K/AKT/mTOR, NFkB, and STAT3 pathways. Moreover, HMGA2 supports a cancer stem cell phenotype and renders cancer cells resistant to chemotherapeutic agents. In this review, we discuss these oncogenic roles of HMGA2 in different types of cancers and propose that HMGA2 may be used for cancer diagnostic, prognostic, and therapeutic purposes. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
26. Single-cell sequencing unveils extensive intratumoral heterogeneity of cancer/testis antigen expression in melanoma and lung cancer.
- Author
-
Traynor S, Jakobsen MK, Green TM, Komic H, Palarasah Y, Pedersen CB, Ditzel HJ, Thoren FB, Guldberg P, and Gjerstorff MF
- Subjects
- Humans, Male, Gene Expression Regulation, Neoplastic, Melanoma genetics, Melanoma pathology, Melanoma immunology, Melanoma metabolism, Lung Neoplasms genetics, Lung Neoplasms pathology, Lung Neoplasms immunology, Antigens, Neoplasm genetics, Antigens, Neoplasm metabolism, Antigens, Neoplasm immunology, Single-Cell Analysis methods
- Abstract
Cancer/testis antigens (CTAs) are widely expressed in melanoma and lung cancer, emerging as promising targets for vaccination strategies and T-cell-based therapies in these malignancies. Despite recognizing the essential impact of intratumoral heterogeneity on clinical responses to immunotherapy, our understanding of intratumoral heterogeneity in CTA expression has remained limited. We employed single-cell mRNA sequencing to delineate the CTA expression profiles of cancer cells in clinically derived melanoma and lung cancer samples. Our findings reveal a high degree of intratumoral transcriptional heterogeneity in CTA expression. In melanoma, every cell expressed at least one CTA. However, most individual CTAs, including the widely used therapeutic targets NY-ESO-1 and MAGE, were confined to subpopulations of cells and were uncoordinated in their expression, resulting in mosaics of cancer cells with diverse CTA profiles. Coordinated expression was observed, however, mainly among highly structurally and evolutionarily related CTA genes. Importantly, a minor subset of CTAs, including PRAME and several members of the GAGE and MAGE-A families, were homogenously expressed in melanomas, highlighting their potential as therapeutic targets. Extensive heterogeneity in CTA expression was also observed in lung cancer. However, the frequency of CTA-positive cancer cells was notably lower and homogenously expressed CTAs were only identified in one of five tumors in this cancer type. Our findings underscore the need for careful CTA target selection in immunotherapy development and clinical testing and offer a rational framework for identifying the most promising candidates., Competing Interests: Competing interests: No, there are no competing interests., (© Author(s) (or their employer(s)) 2024. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2024
- Full Text
- View/download PDF
27. Correction: Chimeric Antigen Receptor T Cells Targeting CD79b Show Efficacy in Lymphoma with or without Cotargeting CD19.
- Author
-
Ormhøj M, Scarfò I, Cabral ML, Bailey SR, Lorrey SJ, Bouffard AA, Castano AP, Larson RC, Riley LS, Schmidts A, Choi BD, Andersen RS, Cédile O, Nyvold CG, Christensen JH, Gjerstorff MF, Ditzel HJ, Weinstock DM, Barington T, Frigault MJ, and Maus MV
- Published
- 2024
- Full Text
- View/download PDF
28. Age-related differences in cancer biology in older patients.
- Author
-
Gammelgaard OL, Guldberg P, Gjerstorff MF, and Ditzel HJ
- Subjects
- Humans, Aged, Biology, Immune System, Tumor Microenvironment genetics, Aging genetics, Neoplasms genetics, Neoplasms therapy
- Abstract
Aging impacts cancer development with incidence rising exponentially. Age-related genetic and epigenetic changes, along with the aging microenvironment, contribute to cancer development. In older individuals, tumours manifest a heightened mutational burden and distinct genetic changes which differ significantly from tumours observed in younger patients. The aging microenvironment accumulates senescent cells, altered matrix, and a dysregulated immune system. Age-related changes in the immune system fuel tumour development and treatment resistance. Understanding these processes is crucial for optimized treatment of older patients with cancer, as discussed in this review., (Published under Open Access CC-BY-NC-BD 4.0. https://creativecommons.org/licenses/by-nc-nd/4.0/.)
- Published
- 2024
- Full Text
- View/download PDF
29. Targeting two distinct epitopes on human CD73 with a bispecific antibody improves anticancer activity.
- Author
-
Gammelgaard OL, Terp MG, Renn C, Labrijn AF, Hamaker O, Nielsen AY, Vever H, Hansen SW, Gjerstorff MF, Müller CE, Parren PW, and Ditzel HJ
- Subjects
- Adenosine, Animals, Epitopes, Humans, Mice, Antibodies, Bispecific pharmacology, Antibodies, Bispecific therapeutic use, Neoplasms
- Abstract
Background: Immunosuppressive extracellular adenosine is generated by the enzymatic activity of CD73. In preclinical models, antibodies (Abs) targeting different epitopes on CD73 exert anticancer activity through distinct mechanisms such as inhibition of enzymatic activity, engagement of Fc receptors, and spatial redistribution of CD73., Methods: Using controlled Fab arm exchange, we generated biparatopic bispecific antibodies (bsAbs) from parental anti-CD73 Abs with distinct anticancer activities. The resulting anticancer activity was evaluated using in vitro and in vivo models., Results: We demonstrate that different anticancer activities can be combined in a biparatopic bsAb. Remarkably, the bsAb significantly improved the enzyme inhibitory activity compared with the parental Abs, which led to neutralization of adenosine-mediated T-cell suppression as demonstrated by proliferation and interferon gamma (IFN-γ) production and prolonged survival of tumor-bearing mice. Additionally, the bsAb caused more efficient internalization of cell surface CD73 and stimulated potent Fc-mediated engagement of human immune effector cells in vitro and in vivo., Conclusions: Our data collectively demonstrate that complementary anticancer mechanisms of action of distinct anti-CD73 Abs can be combined and enhanced in a biparatopic bsAb. The multiple mechanisms of action and superior activity compared with the monospecific parental Abs make the bsAb a promising candidate for therapeutic targeting of CD73 in cancer. This concept may greatly improve future Ab design., Competing Interests: Competing interests: All authors except AFL and PP declare no competing interests. AFL is an employee of Genmab, a biotechnology company that commercializes the DuoBody technology platform. He owns warrants and stock. PP is a co-inventor of patents relating to the DuoBody technology., (© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2022
- Full Text
- View/download PDF
30. Overexpression of HMGA2 in breast cancer promotes cell proliferation, migration, invasion and stemness.
- Author
-
Mansoori B, Duijf PHG, Mohammadi A, Najafi S, Roshani E, Shanehbandi D, Hajiasgharzadeh K, Shirjang S, Ditzel HJ, Kazemi T, Mokhtarzadeh A, Gjerstorff MF, and Baradaran B
- Abstract
Despite improved therapeutic strategies for early-stage breast cancer, the most common cancer type in women, relapse remains common and the underlying mechanisms for this progression remain poorly understood. To gain more insight, we studied the DNA-binding protein HMGA2 in breast cancer development and stemness. We demonstrated that HMGA2 is overexpressed in breast cancer tissues at the mRNA and protein levels (P value <0.0001). HMGA2 knockdown and overexpression in breast cancer cells revealed that HMGA2 promotes cell proliferation and protects against apoptosis via the intrinsic pathway. HMGA2 knockdown also causes cell cycle arrest in G2/M phase. In addition, we found that HMGA2 increases breast cancer cell migration and invasion (P value <0.001) and promotes the acquisition of cancer stem cell features, both in vitro, in colony formation (P value <0.01) and spheroid assays, and in breast cancer tissues. Overexpression of HMGA2 in breast cancer spurs the acquisition of several hallmarks of cancer, including increased cell proliferation, migration, invasion and stemness, and decreased apoptosis. Thus, targeting HMGA2 could represent an effective strategy to block breast cancer progression.
- Published
- 2020
- Full Text
- View/download PDF
31. Chimeric Antigen Receptor T Cells Targeting CD79b Show Efficacy in Lymphoma with or without Cotargeting CD19.
- Author
-
Ormhøj M, Scarfò I, Cabral ML, Bailey SR, Lorrey SJ, Bouffard AA, Castano AP, Larson RC, Riley LS, Schmidts A, Choi BD, Andersen RS, Cédile O, Nyvold CG, Christensen JH, Gjerstorff MF, Ditzel HJ, Weinstock DM, Barington T, Frigault MJ, and Maus MV
- Subjects
- Animals, Apoptosis, Cell Proliferation, Humans, Lymphocyte Activation, Lymphoma, Mantle-Cell immunology, Lymphoma, Mantle-Cell metabolism, Mice, Mice, Inbred NOD, Mice, SCID, Prognosis, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Antigens, CD19 immunology, CD79 Antigens immunology, Immunotherapy, Adoptive methods, Lymphoma, Mantle-Cell therapy, Receptors, Chimeric Antigen immunology, T-Lymphocytes immunology
- Abstract
Purpose: T cells engineered to express a chimeric antigen receptor (CAR) against CD19 have recently been FDA approved for the treatment of relapsed or refractory large B-cell lymphoma. Despite the success and curative potential of CD19 CAR T cells, several reports describing disease relapse due to antigen loss are now emerging., Experimental Design: We developed a novel CAR construct directed against CD79b, a critical receptor for successful B-cell development that remains highly expressed in several subtypes of B-cell lymphoma, including mantle cell lymphoma (MCL). We tested CAR T cells directed against CD79b alone or in combination with CD19 targeting in a single construct, against cell line- and patient-derived xenograft models., Results: We demonstrate CAR79b antigen-specific recognition and cytotoxicity against a panel of cell lines and patient-derived xenograft models of MCL. Importantly, we show that downregulation of CD19 does not influence surface expression of CD79b and that anti-CD79b CAR T cells alone or arranged in a dual-targeting format with a CD19 single-chain variable fragment (scFv) are able to recognize and eliminate CD19
+ , CD19- , and mixed CD19+ /CD19- B-cell lymphoma., Conclusions: Our findings demonstrate that CAR T cells targeting CD79b alone or in combination have promise for treating and preventing CD19 antigen escape in B-cell lymphomas., (©2019 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
32. Oncogenic cancer/testis antigens: prime candidates for immunotherapy.
- Author
-
Gjerstorff MF, Andersen MH, and Ditzel HJ
- Subjects
- Humans, Male, Antigens, Neoplasm immunology, Immunotherapy methods, Neoplasms immunology, Neoplasms therapy, Testis immunology
- Abstract
Recent developments have set the stage for immunotherapy as a supplement to conventional cancer treatment. Consequently, a significant effort is required to further improve efficacy and specificity, particularly the identification of optimal therapeutic targets for clinical testing. Cancer/testis antigens are immunogenic, highly cancer-specific, and frequently expressed in various types of cancer, which make them promising candidate targets for cancer immunotherapy, including cancer vaccination and adoptive T-cell transfer with chimeric T-cell receptors. Our current understanding of tumor immunology and immune escape suggests that targeting oncogenic antigens may be beneficial, meaning that identification of cancer/testis antigens with oncogenic properties is of high priority. Recent work from our lab and others provide evidence that many cancer/testis antigens, in fact, have oncogenic functions, including support of growth, survival and metastasis. This novel insight into the function of cancer/testis antigens has the potential to deliver more effective cancer vaccines. Moreover, immune targeting of oncogenic cancer/testis antigens in combination with conventional cytotoxic therapies or novel immunotherapies such as checkpoint blockade or adoptive transfer, represents a highly synergistic approach with the potential to improve patient survival.
- Published
- 2015
- Full Text
- View/download PDF
33. Identification of genes with altered expression in medullary breast cancer vs. ductal breast cancer and normal breast epithelia.
- Author
-
Gjerstorff MF, Benoit VM, Laenkholm AV, Nielsen O, Johansen LE, and Ditzel HJ
- Subjects
- Antigens, Neoplasm genetics, DNA Primers, Female, Humans, Insulin-Like Growth Factor Binding Proteins genetics, Reference Values, Somatomedins genetics, Breast cytology, Breast physiology, Breast Neoplasms genetics, Carcinoma, Ductal, Breast genetics, Carcinoma, Medullary genetics, Epithelial Cells physiology, Gene Expression Regulation, Gene Expression Regulation, Neoplastic
- Abstract
Medullary breast cancer (MCB) is a morphologically and biologically distinct subtype that, despite cytologically highly malignant characteristics, has a favorable prognosis compared to the more common infiltrating ductal breast carcinoma. MCB metastasizes less frequently, which has been attributed to both immunological and endogenous cellular factors, although little is known about the distinct biology of MCB that may contribute to the improved outcome of MCB patients. To identify candidate genes, we performed gene array expression analysis of cell lines of MCB, ductal breast cancer and normal breast epithelia, and the differential expression of a panel of candidate genes was further validated by quantitative PCR and immunohistochemical analysis of cell lines and tumor biopsies. A limited number of genes, including several members of the GAGE and insulin growth factor binding protein (IGFBP) gene families, Vav1, monoglyceride lipase and NADP+-dependent malic enzyme, exhibited altered expression in MCB vs. ductal breast cancer, and the differences for some of these genes were confirmed on an extended panel of cell lines by quantitative PCR. Immunohistochemical analysis further established that the expression of monoglyceride lipase was restricted to ductal breast cancer and present in 77% of these tumors, while Vav1 was restricted to MCB and present in 60% of tumors. In this study, we have identified genes that are differentially expressed in MCB vs. ductal breast cancer and further analysis of the gene products should illuminate the biological differences between MCB and ductal breast cancer.
- Published
- 2006
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.