Julie Westerlin Kjeldsen
University of Copenhagen
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Julie Westerlin Kjeldsen.
Cancer Research | 2015
Marco Donia; Rikke Andersen; Julie Westerlin Kjeldsen; Paolo Fagone; Shamaila Munir; Ferdinando Nicoletti; Mads Hald Andersen; Per thor Straten; Inge Marie Svane
In the absence of a local inflammatory response, expression of MHC class II molecules is restricted mainly to hematopoietic cells and thymus epithelium. However, certain tumors, such as melanoma, may acquire aberrant constitutive expression of MHC class II. In a set of primary melanoma cell populations and correspondingly expanded autologous tumor-infiltrating lymphocytes (TIL), we show how MHC class II expression on melanoma cells associates with strong MHC class II-restricted CD4(+) T-cell responses that are specific for tumors. Notably, we found that tumor-specific CD4(+) T-cell responses were dominated by TNF production. TNF reduced CD8(+) T-cell activation in IFNγ-rich environments resembling a tumor site. Conversely, direct CD4(+) T-cell responses had no influence on either the proliferation or viability of melanoma cells. Taken together, our results illustrate a novel immune escape mechanism that can be activated by aberrant expression of MHC class II molecules, which by attracting tumor-specific CD4(+) T cells elicit a local inflammatory response dominated by TNF that, in turn, inhibits cytotoxic CD8(+) T-cell responses
Clinical Cancer Research | 2017
Marco Donia; Julie Westerlin Kjeldsen; Rikke Andersen; Marie Christine Wulff Westergaard; Valentina Bianchi; Mateusz Legut; Meriem Attaf; Barbara Szomolay; Sascha Ott; Garry Dolton; Rikke Birgitte Lyngaa; Sine Reker Hadrup; Andrew K. Sewell; Inge Marie Svane
Purpose: Infusion of highly heterogeneous populations of autologous tumor-infiltrating lymphocytes (TIL) can result in tumor regression of exceptional duration. Initial tumor regression has been associated with persistence of tumor-specific TILs 1 month after infusion, but mechanisms leading to long-lived memory responses are currently unknown. Here, we studied the dynamics of bulk tumor-reactive CD8+ T-cell populations in patients with metastatic melanoma following treatment with TILs. Experimental Design: We analyzed the function and phenotype of tumor-reactive CD8+ T cells contained in serial blood samples of 16 patients treated with TILs. Results: Polyfunctional tumor-reactive CD8+ T cells accumulated over time in the peripheral lymphocyte pool. Combinatorial analysis of multiple surface markers (CD57, CD27, CD45RO, PD-1, and LAG-3) showed a unique differentiation pattern of polyfunctional tumor-reactive CD8+ T cells, with highly specific PD-1 upregulation early after infusion. The differentiation and functional status appeared largely stable for up to 1 year after infusion. Despite some degree of clonal diversification occurring in vivo within the bulk tumor-reactive CD8+ T cells, further analyses showed that CD8+ T cells specific for defined tumor antigens had similar differentiation status. Conclusions: We demonstrated that tumor-reactive CD8+ T-cell subsets that persist after TIL therapy are mostly polyfunctional, display a stable partially differentiated phenotype, and express high levels of PD-1. These partially differentiated PD-1+ polyfunctional TILs have a high capacity for persistence and may be susceptible to PD-L1/PD-L2–mediated inhibition. Clin Cancer Res; 23(19); 5779–88. ©2017 AACR.
Cancer immunology research | 2018
Rikke Andersen; Marie Christine Wulff Westergaard; Julie Westerlin Kjeldsen; Anja Müller; Natasja Wulff Pedersen; Sine Reker Hadrup; Özcan Met; Barbara Seliger; Bjarne Kromann-Andersen; Thomas Hasselager; Marco Donia; Inge Marie Svane
TILs isolated from primary RCC specimens could recognize tumors. However, their immune responses were weaker than TILs from metastatic melanoma and displayed a mono-/oligo-functional pattern. The ability to select and expand poly-functional T cells may improve cell therapy for RCC. In vitro expansion of large numbers of highly potent tumor-reactive T cells appears a prerequisite for effective adoptive cell therapy (ACT) with autologous tumor-infiltrating lymphocytes (TIL) as shown in metastatic melanoma (MM). We therefore sought to determine whether renal cell carcinomas (RCC) are infiltrated with tumor-reactive T cells that could be efficiently employed for adoptive transfer immunotherapy. TILs and autologous tumor cell lines (TCL) were successfully generated from 22 (92%) and 17 (77%) of 24 consecutive primary RCC specimens and compared with those generated from metastatic melanoma. Immune recognition of autologous TCLs or fresh tumor digests was observed in CD8+ TILs from 82% of patients (18/22). Cytotoxicity assays confirmed the tumoricidal capacity of RCC-TILs. The overall expansion capacity of RCC-TILs was similar to MM-TILs. However, the magnitude, polyfunctionality, and ability to expand in classical expansion protocols of CD8+ T-cell responses was lower compared with MM-TILs. The RCC-TILs that did react to the tumor were functional, and antigen presentation and processing of RCC tumors was similar to MM-TILs. Direct recognition of tumors with cytokine-induced overexpression of human leukocyte antigen class II was observed from CD4+ T cells (6/12; 50%). Thus, TILs from primary RCC specimens could be isolated, expanded, and could recognize tumors. However, immune responses of expanded CD8+ RCC-TILs were typically weaker than MM-TILs and displayed a mono-/oligofunctional pattern. The ability to select, enrich, and expand tumor-reactive polyfunctional T cells may be critical in developing effective ACT with TILs for RCC. In summary, TILs isolated from primary RCC specimens could recognize tumors. However, their immune responses were weaker than MM-TILs and displayed a mono-/oligofunctional pattern. The ability to select and expand polyfunctional T cells may improve cell therapy for RCC. Cancer Immunol Res; 6(2); 222–35. ©2018 AACR.
Annals of Oncology | 2018
Rikke Andersen; Troels Holz Borch; Arianna Draghi; A Gokuldass; M A H Rana; Magnus Pedersen; M Nielsen; Per Kongsted; Julie Westerlin Kjeldsen; Marie Christine Wulff Westergaard; H D Radic; C A Chamberlain; Lisbet Rosenkrantz Hölmich; Helle Westergren Hendel; M S Larsen; Özcan Met; Inge Marie Svane; Marco Donia
Background Almost half of the patients with metastatic melanoma obtain only short-term or no benefit at all from checkpoint inhibitor (CPI) immunotherapy. In this study, we investigated whether the immune system of patients progressing following CPI treatment was able to generate functional tumor-specific immune responses. Materials and methods Tumor-infiltrating lymphocytes (TILs) were isolated and expanded from metastatic melanoma lesions which progressed during or after anti-programmed cell death protein 1 (PD)-1 and anti-Cytotoxic T-Lymphocyte Antigen 4 (CTLA-4) treatment. Tumor-specific immune responses were assessed with co-culture assays of TILs and autologous tumor cells. Results TILs from 23 metastases of individual patients could be assessed for T cells recognition of autologous tumor cells. All metastases were progressive on or following anti-PD-1 (23/23, 100%), and the majority also after anti-CTLA-4 (17/23, 74%). Functional antitumor immune responses were detected in 19/23 patients (83%). Both CD8+ (in 18/23 patients, 78%) and CD4+ (in 16/23 patients, 70%) TILs were able to recognize autologous tumors. A large fraction of CD8+ TILs (median 23%, range 1.0%-84%) recognized tumor cells. This is similar to the cohorts of unselected patient populations with metastatic melanoma presented in previous studies. The localization of intratumoral immune infiltrates was heterogeneous among samples. In a phase I/II clinical trial, TILs were administered with lymphodepleting chemotherapy, pegIFNα2b and interleukin-2 to 12 patients with CPI-resistant melanoma. Out of 12 patients who previously failed CPI therapy, treatment with TILs resulted in two partial responses, of which one is ongoing. Conclusions Tumor-reactive T cells appear to heavily infiltrate the tumor microenvironment of patients who failed previous CPI treatment. These patients can still respond to an infusion of unselected autologous TILs. Our results warrant further testing of novel immune re-activation strategies in melanoma patients who failed multiple CPI therapy.
OncoImmunology | 2016
Marco Donia; Julie Westerlin Kjeldsen; Inge Marie Svane
ABSTRACT TNF has been associated with both inhibition and promotion of tumor growth. We recently described a mechanism by which tumor cells attract TNF producing cells via expression of MHC class II molecules.
Frontiers in Immunology | 2018
Julie Westerlin Kjeldsen; Trine Zeeberg Iversen; Lotte Engell-Noerregaard; Anders Mellemgaard; Mads Hald Andersen; Inge Marie Svane
Background: Long-term follow-up on a clinical trial of 15 stage III-IV NSCLC patients treated with an Indoleamine 2,3-Dioxygenase (IDO) peptide vaccine (NCT01219348). Methods: Fifteen HLA-A2-positive patients with stable stage III-IV NSCLC after standard chemotherapy were treated with subcutaneous vaccinations (100 μg IDO5 peptide, sequence ALLEIASCL, formulated in 900 μl Montanide) biweekly for 2.5 months and thereafter monthly until progression or up to 5 years. Here we report long-term clinical follow-up, toxicity and immunity. Results: Three of 15 patients are still alive corresponding to a 6-year overall survival of 20 %. Two patients continued monthly vaccinations for 5 years (56 vaccines). One of the two patients developed a partial response (PR) of target lesions in the liver 15 months after the first vaccine and has remained in PR ever since. The other patient had a solitary distant metastasis in a lymph node in retroperitoneum at baseline which normalized during treatment. All following evaluation scans during the treatment have been tumor free. The vaccine was well tolerated for all 5 years with no long-term toxicities registered. The third long-term surviving patient discontinued vaccinations after 11 months due to disease progression. Flow cytometry analyses of PBMCs from the two long-term responders demonstrated stable CD8+ and CD4+ T-cell populations during treatment. In addition, presence of IDO-specific T-cells was detected by IFN-γ Elispot in both patients at several time points during treatment. Conclusion: IDO peptide vaccination was well tolerated for administration up to 5years. Two of 15 patients are long-term responders with ongoing clinical response 6 years after 1st vaccination.
Journal for ImmunoTherapy of Cancer | 2015
Marco Donia; Julie Westerlin Kjeldsen; Rikke Andersen; Marie Christine Wulff Westergaard; Inge Marie Svane
Infusion of highly heterogeneous populations of autologous tumor-infiltrating lymphocytes (TILs) can result in tumor regression of exceptional duration. Initial tumor regression has been associated to persistence of TILs shortly after infusion, but it is currently not clear whether a single infusion of TILs can establish long term memory antitumor responses.
Annals of Oncology | 2016
Rikke Fredslund Andersen; Marie Christine Wulff Westergaard; Julie Westerlin Kjeldsen; Özcan Met; B. Kromann-Andersen; T. Hasselager; Marco Donia; I.M. Svane
Annals of Oncology | 2015
Rikke Fredslund Andersen; Marie Christine Wulff Westergaard; Julie Westerlin Kjeldsen; Marco Donia; I.M. Svane
Annals of Oncology | 2017
Rikke Andersen; Troels Holz Borch; Arianna Draghi; A Gokuldass; M A H Rana; Magnus Pedersen; M Nielsen; Per Kongsted; Julie Westerlin Kjeldsen; Marie Christine Wulff Westergaard; Lisbet Rosenkrantz Hölmich; Helle Westergren Hendel; M S Larsen; Özcan Met; I.M. Svane; Marco Donia