Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
Filter
  • American Association for Cancer Research (AACR)  (5)
  • 2020-2024  (5)
Type of Medium
Publisher
  • American Association for Cancer Research (AACR)  (5)
Language
Years
  • 2020-2024  (5)
Year
Subjects(RVK)
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 14 ( 2021-07-15), p. 3862-3875
    Abstract: Lung cancers driven by mutant forms of EGFR invariably develop resistance to kinase inhibitors, often due to secondary mutations. Here we describe an unconventional mechanism of resistance to dacomitinib, a newly approved covalent EGFR kinase inhibitor, and uncover a previously unknown step of resistance acquisition. Dacomitinib-resistant (DR) derivatives of lung cancer cells were established by means of gradually increasing dacomitinib concentrations. These DR cells acquired no secondary mutations in the kinase or other domains of EGFR. Along with resistance to other EGFR inhibitors, DR cells acquired features characteristic to epithelial–mesenchymal transition, including an expanded population of aldehyde dehydrogenase–positive cells and upregulation of AXL, a receptor previously implicated in drug resistance. Unexpectedly, when implanted in animals, DR cells reverted to a dacomitinib-sensitive state. Nevertheless, cell lines derived from regressing tumors displayed renewed resistance when cultured in vitro. Three-dimensional and cocultures along with additional analyses indicated lack of involvement of hypoxia, fibroblasts, and immune cells in phenotype reversal, implying that other host-dependent mechanisms might nullify nonmutational modes of resistance. Thus, similar to the phenotypic resistance of bacteria treated with antibiotics, the reversible resisters described here likely evolve from drug-tolerant persisters and give rise to the irreversible, secondary mutation–driven nonreversible resister state. Significance: This study reports that stepwise acquisition of kinase inhibitor resistance in lung cancers driven by mutant EGFR comprises a nonmutational, reversible resister state.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2020
    In:  Cancer Research Vol. 80, No. 16_Supplement ( 2020-08-15), p. 480-480
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 480-480
    Abstract: INTRODUCTION: Cancer cell-targeted photoimmunotherapy (PIT) is a platform technology under development for the treatment of various cancers. PIT is a drug and device combination that utilizes an antibody conjugated to the IR700 (IRDye 700DX®) light-excitable dye, which upon antigen binding and illumination with non-thermal red light, leads to necrotic cell death. The objective of these studies was to characterize the molecular pharmacology and the underlying mechanism of action of PIT. METHODS: In vitro PIT studies using human and mouse cancer cells were performed to evaluate conjugate (antibody + dye) binding, dose response and light dosimetry response. Mouse xenograft experiments assessed necrotic markers in tumor tissue and surrounding normal tissue post-PIT treatment. Oxidative damage to lipids was detected with a reporter dye. Mass spectrometry analysis was performed to assess protein oxidation, crosslinking and axial ligand dissociation from the dye. RESULTS: Binding of the conjugate to the target cancer cells was required for PIT treatment to induce cell death as measured by antibody competition studies, however, internalization of the conjugate was not required for cytotoxic effects. Administration of non-thermal red light to the target cells bound with the conjugate led to morphologic changes and membrane disruption within minutes, and necrotic cell death was observed in a conjugate- and light-dose dependent manner. Requirement of conjugate binding to the cancer antigen provided high level of tumor specificity as demonstrated by lack of damage to normal surrounding tissue after PIT treatment. Light excitation of the conjugate generated 1O2 with high quantum yield. In vitro PIT treatment led to cell membrane lipid peroxidation, mediated by 1O2 as shown by abrogation with sodium azide, a selective 1O2 quencher. In addition, protein oxidation, cross-linking and aggregation of the conjugate was observed. In low oxygen concentrations and in the presence of ascorbate, photo-induced dissociation of axial ligands from the dye was detected. CONCLUSIONS: These studies suggest the molecular mechanism of action of PIT includes generation of 1O2 that leads to lipid peroxidation and cell membrane disruption. Additional mechanisms may include protein oxidation and non-oxygen dependent mechanisms such as axial ligand loss of the IR700 dye. These results describe a biophysical process that damages and disrupts the cell membrane integrity resulting in necrotic cell death and immunogenic cell death. Citation Format: Roger Heim, Lew Makings, Torsten Wiemann, Anthony Manibusan, Miguel Garcia-Guzman. Molecular mechanism of action of photoimmunotherapy with antibody-IR700 dye conjugates [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 480.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2021
    In:  Cancer Research Vol. 81, No. 4_Supplement ( 2021-02-15), p. PS16-36-PS16-36
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 4_Supplement ( 2021-02-15), p. PS16-36-PS16-36
    Abstract: Introduction: The main cause of death for cancer patients is the development of metastasis. These arise mainly due to irresponsiveness of cancer cells to the administered therapy, which then fails to eliminate all cancer cells present in the patient. To overcome this problem, it is essential to understand which mechanisms are involved in the lack of treatment response. We are investigating how the tumour microenvironment (TME) affects the response of cancer cells to chemotherapy (CTX) and how it can be modulated to improve the outcome of patients to therapy. Materials and Methods: Co-culture of chemotherapy-treated breast cancer cell lines with primary fibroblasts isolated from breast cancer patients was performed to investigate if fibroblasts affect the response of tumour cells to commonly used agents, such as epirubicin and paclitaxel. Recovery of cells was assessed using colony formation assays (CFA) and cell cycle profiling by EdU and the FUCCI system. To further explore the complex crosstalk between cancer cells and fibroblasts in the context of CTX, gene expression analysis of both cell types was done using next generation sequencing. Validation and evaluation of the biological impact of the identified pathways was done using RT-qPCR, western-blot and perturbation experiments. Lastly, publicly available datasets for breast cancer were used to investigate the clinical relevance of our findings. Results and Discussion: We show that cancer cells utilize paracrine signalling with stromal fibroblasts to drive their recovery after treatment withdrawal. Cell cycle analysis and RNA-sequencing revealed an increase in cell cycle re-entry of CTX-treated cancer cells in co-culture with fibroblasts. In addition, we have successfully shown that treated cancer cells upregulate an important secreted factor that modulates fibroblasts into a pro-tumorigenic state. Moreover, analysis of human breast carcinomas supported the proposed role of the identified factor since its expression is inversely correlated with recurrence free survival (RFS). Moreover, expression of the gene signature identified in stromal fibroblasts in co-culture with CTX-treated cancer cells was equally associated with higher recurrence rates and a worse outcome in breast cancer patients. Conclusion: CTX-induced secretory profile of cancer cells orchestrates the reprogramming of stromal fibroblasts into a pro-tumorigenic state, which drives the expansion of cancer cells. Our study unravels a novel paracrine communication between cancer cells and stromal fibroblasts that ultimately results in the escape of malignant cells to treatment, highlighting the importance of the TME in drug response. Targeting of this axis could potentially improve the outcome of breast cancer patients to CTX treatment. Citation Format: Ana Maia, Zuguang Gu, André Koch, Mireia Berdiel-Acer, Rainer Will, Matthias Schlesner, Stefan Wiemann. Paracrine signalling with stromal fibroblasts drives recovery of cancer cells after chemotherapy treatment [abstract]. In: Proceedings of the 2020 San Antonio Breast Cancer Virtual Symposium; 2020 Dec 8-11; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2021;81(4 Suppl):Abstract nr PS16-36.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 4_Supplement ( 2022-02-15), p. P1-08-15-P1-08-15
    Abstract: Introduction. Neoadjuvant chemotherapy (NACT) is the standard of care in aggressive breast cancer, including triple negative breast cancer (TNBC). Cytotoxic drugs specifically target proliferating cells, however, patient outcome is variable. Tumor physiology and response to therapy are orchestrated by an intricate interplay between cancer, stromal and immune cells collectively forming the tumor microenvironment. We have recently uncovered a feedback mechanism of tumor cells and fibroblasts, involving IFNB1 signaling, that supports tumor cells in the recovery from chemotherapy-induced stress. Here, we wanted to assess whether targets of IFNB1 signaling in fibroblasts and tumor cells would qualify as predictive markers of pathological complete response (pCR) after neoadjuvant therapy (NACT) as prognostic markers for the course of the disease. Methods and Patients. RNA-sequencing data from in vitro experiments found the GO-term GO:0051607 ‘defense response to virus’ significantly enriched. Twentyfour genes intersected between differentially expressed genes and the genes of this GO-term. We selected three of the encoded proteins a) interferon induced with helicase C domain 1 IFIH1, b) interferon alpha-inducible protein ISG15, c) 2'-5'-oligoadenylate synthetase OAS1 to test their expression in human specimens of TNBCs after NACT by immunohistochemistry (IHC). None of the respective genes correlated with recurrence free survival when tested in treatment-naïve tumor biopsies (KM Plotter). A prospective consecutively enrolled cohort (2000 - 2021) was available with an overall pCR rate of 46%. pCR was defined by no invasive cancer cell in breast or axilla (ypT0 N0). The median follow-up for iDFS was 36.2 months (6-154) and for OS 39.3 months (6-214).Primary objective was the correlation between IFIH1, ISG15 and OAS1 protein expression in the residual tumor by non-pCR or in the tumor bed by pCR. Second objective was the association of IFIH1, ISG15 and OAS1 protein expression to invasive disease-free survival (iDFS) and overall survival (OS). Results. To date, IHC staining has been established for IFIH1, ISG15 and OAS1. In representative stainings of FFPE tissue samples with pCR we did not detect any positive signal for IFIH1 and ISG15 in stromal cells like fibroblast or lymphocytes. Slight to strong protein expression was detected by non-pCR in cancer cells, stromal cells and tumour infiltrating lymphocytes. In contrast OAS1 was expressed especially strong in lymphocytes by pCR or non-pCR. Cancer cell showed moderate OAS1 expression. IHC analysis and of the entire cohort is in progress including the analyses of the association of these markers to pCR, non-pCR and iDFS/OS. Conclusion. Using samples from our consecutive, multicentre enrolled cohort, an association between the expression of markers of an IFNB1-triggered antiviral response and pCR and survival was demonstrated in patients of the TNBC subgroup. Analysis of the entire cohort is necessary to potentially demonstrate applicability of an interferon-response as predictor of survival. Citation Format: Marcus Bauer, Martina Vetter, Ana Maia, Efstathios Vlachavas, Brigitta Michels, Mireia Berdiel-Acer, Kathleen Schüler, Alessandra Morselli, Manio Skarlatou, Christoph Thomssen, Stefan Wiemann. Communication between tumor cells and fibroblasts as a prognostic factor of NACT in TNBC [abstract]. In: Proceedings of the 2021 San Antonio Breast Cancer Symposium; 2021 Dec 7-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2022;82(4 Suppl):Abstract nr P1-08-15.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Discovery, American Association for Cancer Research (AACR), Vol. 12, No. 11 ( 2022-11-02), p. 2666-2683
    Abstract: Anticancer therapies have been limited by the emergence of mutations and other adaptations. In bacteria, antibiotics activate the SOS response, which mobilizes error-prone factors that allow for continuous replication at the cost of mutagenesis. We investigated whether the treatment of lung cancer with EGFR inhibitors (EGFRi) similarly engages hypermutators. In cycling drug-tolerant persister (DTP) cells and in EGFRi-treated patients presenting residual disease, we observed upregulation of GAS6, whereas ablation of GAS6's receptor, AXL, eradicated resistance. Reciprocally, AXL overexpression enhanced DTP survival and accelerated the emergence of T790M, an EGFR mutation typical to resistant cells. Mechanistically, AXL induces low-fidelity DNA polymerases and activates their organizer, RAD18, by promoting neddylation. Metabolomics uncovered another hypermutator, AXL-driven activation of MYC, and increased purine synthesis that is unbalanced by pyrimidines. Aligning anti-AXL combination treatments with the transition from DTPs to resistant cells cured patient-derived xenografts. Hence, similar to bacteria, tumors tolerate therapy by engaging pharmacologically targetable endogenous mutators. Significance: EGFR-mutant lung cancers treated with kinase inhibitors often evolve resistance due to secondary mutations. We report that in similarity to the bacterial SOS response stimulated by antibiotics, endogenous mutators are activated in drug-treated cells, and this heralds tolerance. Blocking the process prevented resistance in xenograft models, which offers new treatment strategies. This article is highlighted in the In This Issue feature, p. 2483
    Type of Medium: Online Resource
    ISSN: 2159-8274 , 2159-8290
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2607892-2
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages