Agnete Engelsen
University of Bergen
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Agnete Engelsen.
Basic & Clinical Pharmacology & Toxicology | 2015
Gro Vatne Røsland; Agnete Engelsen
New molecular insight reveals novel points of attack for targeted cancer therapy. The recent advances in cancer genomics and novel insight into the complex biology of cancer make the promise of personalized, targeted cancer medicine closer than ever. The massive parallel sequencing endeavours performed by The Cancer Genome Atlas, the International Cancer Genome Consortium and by numerous individual investigators have provided a comprehensive genomic characterization of a wide range of cancers. The joint efforts enabled by the improved sequencing technology have demonstrated that individual cancers comprise mutational repertoires with only a few frequently recurrent driver genes. Thus, the identification of new drug targets and novel drugs have accelerated and renewed the hopes of personalized cancer therapy achieving clinical reality for a wider range of cancers. Together with cost-effective sequencing technology to perform comprehensive mutational profiling of each individual cancer, this provides the basis for a personalized cancer medicine revolution within the next few years. The aim of this MiniReview is to provide an overview of the history and evolution of targeted cancer therapy, exemplified by molecularly targeted drugs successfully implemented in the clinic. Furthermore, we aim to highlight novel molecular targets for therapeutic intervention, as well as the main present challenges including inter- and intratumor heterogeneity and cellular plasticity in addition to the importance of the tumor micro-environment. Many cancer patients already receive some form of tailored therapy, and recent evidence suggests that novel and highly innovative, targeted approaches are on their way into the clinic.
Journal of Immunology | 2014
Andrea Gras Navarro; Justyna Kmiecik; Lina Leiss; Mateusz Zelkowski; Agnete Engelsen; Øystein Bruserud; Jacques Zimmer; Per Øyvind Enger; Martha Chekenya
Glioblastomas (GBMs) are lethal brain cancers that are resistant to current therapies. We investigated the cytotoxicity of human allogeneic NK cells against patient-derived GBM in vitro and in vivo, as well as mechanisms mediating their efficacy. We demonstrate that KIR2DS2 immunogenotype NK cells were more potent killers, notwithstanding the absence of inhibitory killer Ig–like receptor (KIR)-HLA ligand mismatch. FACS-sorted and enriched KIR2DS2+ NK cell subpopulations retained significantly high levels of CD69 and CD16 when in contact with GBM cells at a 1:1 ratio and highly expressed CD107a and secreted more soluble CD137 and granzyme A. In contrast, KIR2DS2− immunogenotype donor NK cells were less cytotoxic against GBM and K562, and, similar to FACS-sorted or gated KIR2DS2− NK cells, significantly diminished CD16, CD107a, granzyme A, and CD69 when in contact with GBM cells. Furthermore, NK cell–mediated GBM killing in vitro depended upon the expression of ligands for the activating receptor NKG2D and was partially abrogated by Ab blockade. Treatment of GBM xenografts in NOD/SCID mice with NK cells from a KIR2DS2+ donor lacking inhibitory KIR-HLA ligand mismatch significantly prolonged the median survival to 163 d compared with vehicle controls (log-rank test, p = 0.0001), in contrast to 117.5 d (log-rank test, p = 0.0005) for NK cells with several inhibitory KIR-HLA ligand mismatches but lacking KIR2DS2 genotype. Significantly more CD56+CD16+ NK cells from a KIR2DS2+ donor survived in nontumor-bearing brains 3 wk after infusion compared with KIR2DS2− NK cells, independent of their proliferative capacity. In conclusion, KIR2DS2 identifies potent alloreactive NK cells against GBM that are mediated by commensurate, but dominant, activating signals.
Frontiers in Cell and Developmental Biology | 2018
Tiina Jokela; Agnete Engelsen; Agata Rybicka; Fanny A. Pelissier Vatter; James C. Garbe; Masaru Miyano; Crina Tiron; Dan Ferariu; Lars A. Akslen; Martha R. Stampfer; James B. Lorens; Mark A. LaBarge
The existence of rare cancer cells that sporadically acquire drug-tolerance through epigenetic mechanisms is proposed as one mechanism that drives cancer therapy failure. Here we provide evidence that specific microenvironments impose non-sporadic expression of proteins related to epithelial plasticity and drug resistance. Microarrays of robotically printed combinatorial microenvironments of known composition were used to make cell-based functional associations between microenvironments, which were design-inspired by normal and tumor-burdened breast tissues, and cell phenotypes. We hypothesized that specific combinations of microenvironment constituents non-sporadically impose the induction of the AXL and cKIT receptor tyrosine kinase proteins, which are known to be involved in epithelial plasticity and drug-tolerance, in an isogenic human mammary epithelial cell (HMEC) malignant progression series. Dimension reduction analysis reveals type I collagen as a dominant feature, inducing expression of both markers in pre-stasis finite lifespan HMECs, and transformed non-malignant and malignant immortal cell lines. Basement membrane-associated matrix proteins, laminin-111 and type IV collagen, suppress AXL and cKIT expression in pre-stasis and non-malignant cells. However, AXL and cKIT are not suppressed by laminin-111 in malignant cells. General linear models identified key factors, osteopontin, IL-8, and type VIα3 collagen, which significantly upregulated AXL and cKIT, as well as a plasticity-related gene expression program that is often observed in stem cells and in epithelial-to-mesenchymal-transition. These factors are co-located with AXL-expressing cells in situ in normal and breast cancer tissues, and associated with resistance to paclitaxel. A greater diversity of microenvironments induced AXL and cKIT expression consistent with plasticity and drug-tolerant phenotypes in tumorigenic cells compared to normal or immortal cells, suggesting a reduced perception of microenvironment specificity in malignant cells. Microenvironment-imposed reprogramming could explain why resistant cells are seemingly persistent and rapidly adaptable to multiple classes of drugs. These results support the notion that specific microenvironments drive drug-tolerant cellular phenotypes and suggest a novel interventional avenue for preventing acquired therapy resistance.
Archive | 2017
Kjersti Davidsen; Gry S. Haaland; Maria Lie; James B. Lorens; Agnete Engelsen
In spite of the advances in cancer treatment over several decades, resistance to antitumor therapy continues to confound current treatment strategies. Recent insights into the epigenetic heterogeneity of cancer have emphasized a need to address the underlying mechanisms driving tumor cell plasticity. Epithelial-to-mesenchymal transition (EMT)-related transdifferentiation programs are prevalent in aggressive tumors displaying a drug-resistant, invasive, and immune-evasive phenotype. Novel therapeutically actionable targets are needed in order to disable tumor plasticity mechanisms. The Axl receptor tyrosine kinase has a remarkably broad association with aggressive and therapy-resistant cancers, and the understanding that Axl is not a traditional oncogenic driver as first envisioned, but rather involved in regulating tumor cell plasticity related to the EMT program has provided a framework to understand the role of Axl-mediated signal transduction in cancer. Accordingly, a growing number of studies have demonstrated that Axl signaling is required to maintain tumor plasticity and resistance to cytotoxic and targeted anticancer agents. Novel Axl-targeting agents are emerging, facilitating clinical translation of novel combination approaches dedicated to reverse the plasticity-mediated resistance mechanisms and potentiate current anticancer treatments. In this chapter, we describe the unique roles of the Axl receptor tyrosine kinase in tumor cell plasticity and therapeutic resistance and provide an update on Axl-targeting agents entering clinical trials.
Cancer Research | 2017
Katarzyna Wnuk-Lipinska; Kjersti Davidsen; Magnus Blø; Agnete Engelsen; Jing Kang; Linn Hodneland; Maria Lie; Sébastien Bougnaud; Kristina Y. Aguilera; Lavina Ahmed; Agata Rybicka; Eline Milde Nævdal; Paulina Deyna; Anna Boniecka; Straume Oddbjørn; Salem Chouaib; Rolf A. Brekken; Gro Gausdal; James B. Lorens
The AXL receptor tyrosine kinase is associated with poor overall survival in a wide spectrum of cancers including lung and breast adenocarcinomas. AXL signaling is an important regulator of tumor plasticity related to epithelial-to-mesenchymal transition (EMT) and stem cell traits that drive metastasis and drug resistance. Signaling via AXL is also a key suppressor of the anti-tumor innate immune response, and AXL is expressed on several cells associated with the tumor immune microenvironment including natural killer (NK) cells and tumor-associated macrophages. Hence AXL resides uniquely at the nexus between tumor and microenvironmental anti-tumor immune suppression mechanisms. We report that BGB324, a selective clinical-stage small molecule Axl kinase inhibitor, enhances the effect of immune checkpoint blockade in aggressive adenocarcinoma models with limited immunogenicity by targeting both tumor intrinsic and microenvironmental immune suppression. Immune therapy with anti-CTLA4/PD1 in the 4T1 model increased AXL and EMT-marker expression correlating with a lack of response. Combination with BGB324 resulted in durable primary tumor clearance versus anti-CTLA4/PD1 alone. In a separate study, BGB324 + anti-CTLA4 treatment resulted in significant long-term primary tumor clearance while no response was observed with anti-CTLA4 treatment alone. The extensive metastasis to the lung, liver and spleen characteristic of the 4T1 model was not detected in animals responding to the combination treatment. Importantly, responding animals rejected orthotopic 4T1 tumor cell re-challenge, demonstrating sustained tumor immunity. In the LL2 Lewis Lung model, BGB324 in combination with anti-PD1/PDL1 significantly prevented tumor growth compared to treatment with anti-PD1/PDL1. Tumors from mice treated with BGB324 in combination with immune checkpoint inhibitors displayed reduced EMT traits, altered cytokine expression, enhanced tumor infiltration of effector cells and decreased number of mMDSC. Also, BGB324 significantly reduced IL10 secretion by isolated human macrophages and enhanced human NK-cell mediated NSCLC tumor cell lysis. Collectively these results support a prominent role for AXL in resistance to immune therapy and support clinical translation of combining BGB324 with immune checkpoint inhibitors to improve cancer treatment. Citation Format: Katarzyna Wnuk-Lipinska, Kjersti Davidsen, Magnus Blo, Agnete Engelsen, Jing Kang, Linn Hodneland, Maria Lie, Sebastien Bougnaud, Kristina Aguilera, Lavina Ahmed, Agata Rybicka, Eline Milde Naevdal, Paulina Deyna, Anna Boniecka, Straume Oddbjorn, Salem Chouaib, Rolf Brekken, Gro Gausdal, James B. Lorens. BGB324, a selective small molecule inhibitor of receptor tyrosine kinase AXL, abrogates tumor intrinsic and microenvironmental immune suppression and enhances immune checkpoint inhibitor efficacy in lung and mammary adenocarcinoma models [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 626. doi:10.1158/1538-7445.AM2017-626
Cancer Research | 2016
Gro Gausdal; Kjersti Davidsen; Katarzyna Wnuk-Lipinska; Kathleen Wiertel; Jing Kang; Agnete Engelsen; Sébastien Bougnaud; Monica Hellesøy; Magnus Blø; Lavina Ahmed; Linn Hodneland; Sergej Kiprijanov; Oddbjørn Straume; Rolf A. Brekken; James B. Lorens
Signaling via the AXL receptor tyrosine kinase is a key suppressor of the anti-tumor innate immune response. AXL is expressed on several cells associated with the tumor immune microenvironment including natural killer cells, dendritic cells and tumor-associated macrophages. AXL is also an important regulator of tumor plasticity related to epithelial-to-mesenchymal transition (EMT) that drives tumor immune evasion and resistance to cytotoxic T cell-mediated cell killing. Hence AXL signaling contributes uniquely to both tumor cell intrinsic and microenvironmental anti-tumor immune suppression mechanisms. We therefore evaluated whether blocking AXL signaling with BGB324, a selective clinical-stage small molecule Axl kinase inhibitor, enhances the effect of immune checkpoint blockade in syngeneic cancer mouse models that display limited immunogenicity. We conducted studies in the aggressive mammary adenocarcinoma (4T1) syngeneic (Balb/C) mouse model. We found that AXL expression increased in 4T1 tumors treated with anti-CTLA-4/anti-PD-1 and correlated with lack of response to immune therapy. Combination with BGB324 (50 mg/kg bid) significantly enhanced responsiveness to anti-CTLA-4/anti-PD-1 treatment (10 mg/kg of each, 4 doses) in Balb/C mice bearing established 4T1 tumors. The combination of BGB324 + anti-CTLA-4/anti-PD-1 resulted in durable primary tumor clearance in 23% of treated mice versus 5.6% obtained with anti-CTLA-4/anti-PD-1 alone (p = 0.0157). In a separate study, BGB324 + anti-CTLA-4 treated resulted in 22% long-term primary tumor clearance while no response was observed with anti-CTLA4 treatment alone. The extensive metastasis to the lung, liver and spleen characteristic of this model were concomitantly abrogated in the animals responding to the combination treatment. In addition, BGB324 + anti-CTLA-4/anti-PD-1 treated tumors displayed enhanced infiltration of cytotoxic T lymphocytes. Importantly, responding animals rejected orthotopic 4T1 tumor cell re-challenge, demonstrating sustained tumor immunity. In conclusion, targeting AXL signaling represents a unique opportunity to address multiple tumor immune suppression mechanisms. Our results support combining the clinical-stage AXL inhibitor, BGB324, with immune checkpoint inhibitors to improve treatment of human cancers. Citation Format: Gro Gausdal, Kjersti Davidsen, Katarzyna Wnuk-Lipinska, Kathleen Wiertel, Jing Kang, Agnete Engelsen, Sebastien Bougnaud, Monica Hellesoy, Magnus Blo, Lavina Ahmed, Linn Hodneland, Sergej Kiprijanov, Oddbjorn Straume, Rolf A. Brekken, James B. Lorens. BGB324, a selective small molecule inhibitor of the receptor tyrosine kinase AXL, enhances immune checkpoint inhibitor efficacy. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 566.
The Journal of Steroid Biochemistry and Molecular Biology | 2018
Olivera Bozickovic; Linn Skartveit; Agnete Engelsen; Thomas Helland; Kristin Jonsdottir; Marianne Hauglid Flågeng; Ingvild S. Fenne; Emiel A.M. Janssen; James B. Lorens; Lise Bjørkhaug; Jørn V. Sagen; Gunnar Mellgren
Steroid receptor coactivator 2 (SRC-2) is a nuclear receptor coactivator, important for the regulation of estrogen receptor alpha (ERα)-mediated transcriptional activity in breast cancer cells. However, the transcriptional role of SRC-2 in breast cancer is still ambiguous. Here we aimed to unravel a more precise transcriptional role of SRC-2 and uncover unique target genes in MCF-7 breast cancer cells, as opposed to the known oncogene SRC-3. Gene expression analyses of cells depleted of either SRC-2 or SRC-3 showed that they transcriptionally regulate mostly separate gene sets. However, individual unique gene sets were implicated in some of the same major gene ontology biological processes, such as cellular structure and development. This finding was supported by three-dimensional cell cultures, demonstrating that depletion of SRC-2 and SRC-3 changed the morphology of the cells into epithelial-like hollow acinar structures, indicating that both SRC proteins are involved in maintaining the hybrid E/M phenotype. In clinical ER-positive, HER2-negative breast cancer samples the expression of SRC-2 was negatively correlated with the expression of MCF-7-related luminal, cell cycle and cellular morphogenesis genes. Finally, elucidating SRC-2 unique transcriptional effects, we identified Lyn kinase (an EMT biomarker) to be upregulated exclusively after SRC-2 depletion. In conclusion, we show that both SRC-2 and SRC-3 are essential for the EMT in breast cancer cells, controlling different transcriptional niches.
Cancer Research | 2017
James B. Lorens; Kw Lipinska; Kjersti Davidsen; Magnus Blø; Linn Hodneland; Agnete Engelsen; Jing Kang; Maria Lie; Sébastien Bougnaud; Kristina Y. Aguilera; Lavina Ahmed; Agata Rybicka; Eline Milde Nævdal; P Deyna; Anna Boniecka; Oddbjørn Straume; Salem Chouaib; Rolf A. Brekken; Gro Gausdal
The AXL receptor tyrosine kinase is associated with poor overall survival in breast cancer. Axl signaling is an important regulator of tumor plasticity related to epithelial-to-mesenchymal transition (EMT) and stem cell traits that drive metastasis and drug resistance. Signaling via AXL is also a key suppressor of the anti-tumor innate immune response. AXL is expressed on several cells associated with the tumor immune microenvironment including natural killer cells, dendritic cells and tumor-associated macrophages. AXL is required for tumor immune evasion in mammary adenocarcinoma models and EMT-mediated resistance to cytotoxic T cell and natural killer (NK)-cell mediated cell killing. Hence AXL signaling contributes uniquely to both tumor cell intrinsic and microenvironmental anti-tumor immune suppression mechanisms in breast cancer. We evaluated whether blocking AXL signaling with BGB324, a selective clinical-stage small molecule Axl kinase inhibitor, enhances the effect of immune checkpoint blockade in the aggressive mammary adenocarcinoma (4T1) syngeneic (Balb/C) mouse modelthat display limited immunogenicity. Immune therapy with anti-CTLA-4/anti-PD-1 increased AXL and EMT-marker expression in 4T1 tumors, and correlated with lack of response to immune therapy. Combination treatment with BGB324 (50 mg/kg bid) significantly enhanced responsiveness to anti-CTLA-4/anti-PD-1 treatment (10 mg/kg of each, 4 doses) in Balb/C mice bearing established 4T1 tumors. The combination of BGB324 + anti-CTLA-4/anti-PD-1 resulted in durable primary tumor clearance in 23 % of treated mice versus 5.6% obtained with anti-CTLA-4/anti-PD-1 alone (p=0.0157). In a separate study, BGB324 + anti-CTLA-4 treated resulted in 22% long-term primary tumor clearance while no response was observed with anti-CTLA4 treatment alone. The extensive metastasis to the lung, liver and spleen characteristic of this model were concomitantly abrogated in the animals responding to the combination treatment. In addition, BGB324 + anti-CTLA-4/anti-PD-1 treated tumors displayed enhanced infiltration of cytotoxic T lymphocytes (CTLs). Enhanced presence of CTLs was also detected in spleens from animals responding to treatment. BGB324 + anti-CTLA-4/anti-PD-1 treatment increased the number of NK cells, macrophages and polymorphonuclear neutrophils, but decreased the number of mMDSC. Importantly, responding animals rejected orthotopic 4T1 tumor cell re-challenge, demonstrating sustained tumor immunity. Together with recent results in other tumor types that support a prominent role for AXL in resistance to immune therapy and encouraging results from ongoing clinical trials with BGB324, support combining BGB324 with immune checkpoint inhibitors to improve treatment of breast cancer. Citation Format: Lorens JB, Lipinska KW, Davidsen K, Blo M, Hodneland L, Engelsen A, Kang J, Lie MK, Bougnaud S, Aguilera K, Ahmed L, Rybicka A, Naevdal EM, Deyna P, Boniecka A, Straume O, Chouaib S, Brekken RA, Gausdal G. BGB324, a selective small molecule inhibitor of the receptor tyrosine kinase AXL, enhances immune checkpoint inhibitor efficacy in mammary adenocarcinoma [abstract]. In: Proceedings of the 2016 San Antonio Breast Cancer Symposium; 2016 Dec 6-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2017;77(4 Suppl):Abstract nr P2-04-08.
MedChemComm | 2017
Bibiana I. Ferreira; Maria Lie; Agnete Engelsen; Susana Machado; Wolfgang Link; James B. Lorens
Journal of Clinical Oncology | 2018
Murray Yule; Kjersti Davidsen; Magnus Bloe; Linn Hodneland; Agnete Engelsen; Maria Lie; Sébastien Bougnaud; Stacey D'Mello; Kristina Y. Aguilera; Lavina Ahmed; Agata Rybika; Eline Milde Naeval; Anna Boniecka; Jean Paul Thiery; Salem Chouaib; Rolf A. Brekken; Gro Gausdal; James B. Lorens