Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Per Kongsted is active.

Publication


Featured researches published by Per Kongsted.


Clinical Cancer Research | 2016

Long-Lasting Complete Responses in Patients with Metastatic Melanoma after Adoptive Cell Therapy with Tumor-Infiltrating Lymphocytes and an Attenuated IL2 Regimen.

Rikke Andersen; Marco Donia; Eva Ellebaek; Troels Holz Borch; Per Kongsted; Trine Zeeberg Iversen; Lisbet Rosenkrantz Hölmich; Helle Westergren Hendel; Özcan Met; Mads Hald Andersen; Per thor Straten; Inge Marie Svane

Purpose: Adoptive cell transfer therapy (ACT) based on autologous tumor-infiltrating lymphocytes (TIL) has achieved impressive clinical results in several phase I and II trials performed outside of Europe. Although transient, the toxicities associated with high-dose (HD) bolus IL2 classically administered together with TILs are severe. To further scrutinize whether similar results can be achieved with lower doses of IL2, we have carried out a phase I/II trial of TIL transfer after classical lymphodepleting chemotherapy followed by an attenuated IL2 regimen. Experimental Design: Twenty-five patients with progressive treatment-refractory metastatic melanoma, good clinical performance, age < 70 years, and at least one resectable metastasis were eligible. TIL infusion was preceded by standard lymphodepleting chemotherapy and followed by attenuated doses of IL2 administered in an intravenous, continuous decrescendo regimen (ClinicalTrials.gov Identifier: NCT00937625). Results: Classical IL2-related toxicities were observed but patients were manageable in a general oncology ward without the need for intervention from the intensive care unit. RECIST 1.0 evaluation displayed three complete responses and seven partial responses (ORR 42%). Median overall survival was 21.8 months. Tumor regression was associated with a higher absolute number of infused tumor-reactive T cells. Moreover, induction and persistence of antimelanoma T-cell responses in the peripheral blood was strongly correlated to clinical response to treatment. Conclusions: TIL-ACT with a reduced IL2 decrescendo regimen results in long-lasting complete responses in patients with treatment-refractory melanoma. Larger randomized trials are needed to elucidate whether clinical efficacy is comparable with TIL-ACT followed by HD bolus IL2. Clin Cancer Res; 22(15); 3734–45. ©2016 AACR.


Urologic Oncology-seminars and Original Investigations | 2015

Low-dose prednisolone in first-line docetaxel for patients with metastatic castration-resistant prostate cancer: Is there a clinical benefit?

Per Kongsted; Inge Marie Svane; Henriette Lindberg; Gedske Daugaard; Lisa Sengeløv

BACKGROUND Randomized studies have shown improved survival with the combination of docetaxel (D) and prednisone in patients with metastatic castration-resistant prostate cancer (mCRPC). We retrospectively investigated whether coadministration of low-dose glucocorticoids has clinical benefits. MATERIAL AND METHODS Records from 358 patients with metastatic castration-resistant prostate cancer treated consecutively with either D 75mg/m(2) every 3 weeks (n = 124) (Rigshospitalet) or D and prednisolone (P) 10mg daily (n = 234) (Herlev Hospital) given as first-line chemotherapy were reviewed. Of these, 15 patients treated with glucocorticoids at initiation of D at Rigshospitalet were excluded. Common Terminology Criteria for Adverse Events (CTCAE) version 4.0 was used to register any grade of peripheral edema, grade ≥2 sensory neuropathy, and grade ≥3 nonhematological toxicity. Background clinical data, rates of toxicity, hospital admissions, dose reductions, and post-D treatments were analyzed by the Chi-squared test or Mann-Whitney U test. Progression-free survival and overall survival were calculated by the Kaplan-Meier method. RESULTS Patients treated with D alone had a higher incidence of peripheral edema (32% vs. 15%, P<0.001) and grade 3 nonhematological toxicity (56% vs. 43%, P = 0.022). Patients treated with D alone were also more frequently hospitalized (53% vs. 41%, P = 0.035), mainly owing to a higher incidence of febrile neutropenia in this group (25% vs. 10%, P<0.001). P did not influence progression-free survival (P = 0.692, log-rank test) or overall survival when adjusting for baseline levels of hemoglobin, alkaline phosphatase, lactate dehydrogenase, prostate-specific antigen, and Eastern Cooperative Oncology Group performance status (hazard ratioP = 0.98, 95% CI: 0.76-1.26, P = 0.89, Cox proportional hazard regression model). CONCLUSIONS Coadministration of low-dose P reduced the incidence of peripheral edema, grade 3 nonhematological toxicity, and the risk of being admitted owing to febrile neutropenia during treatment with D. Adjusted survival analysis did not indicate that P affected prognosis.


OncoImmunology | 2015

Tryptophan 2,3-dioxygenase (TDO)-reactive T cells differ in their functional characteristics in health and cancer

Hjortsø; Stine Kiaer Larsen; Per Kongsted; Özcan Met; Thomas Mørch Frøsig; Gitte Holmen Andersen; Shamaila Munir Ahmad; Inge Marie Svane; Jürgen C. Becker; Per thor Straten; Mads Hald Andersen

Tryptophan-2,3-dioxygenase (TDO) physiologically regulates systemic tryptophan levels in the liver. However, numerous studies have linked cancer with activation of local and systemic tryptophan metabolism. Indeed, similar to other heme dioxygenases TDO is constitutively expressed in many cancers. In the present study, we detected the presence of both CD8+ and CD4+ T-cell reactivity toward TDO in peripheral blood of patients with malignant melanoma (MM) or breast cancer (BC) as well as healthy subjects. However, TDO-reactive CD4+ T cells constituted distinct functional phenotypes in health and disease. In healthy subjects these cells predominately comprised interferon (IFN)γ and tumor necrosis factor (TNF)-α producing Th1 cells, while in cancer patients TDO-reactive CD4+ T-cells were more differentiated with release of not only IFNγ and TNFα, but also interleukin (IL)-17 and IL-10 in response to TDO-derived MHC-class II restricted peptides. Hence, in healthy donors (HD) a Th1 helper response was predominant, whereas in cancer patients CD4+ T-cell responses were skewed toward a regulatory T cell (Treg) response. Furthermore, MM patients hosting a TDO-specific IL-17 response showed a trend toward an improved overall survival (OS) compared to MM patients with IL-10 producing, TDO-reactive CD4+ T cells. For further characterization, we isolated and expanded both CD8+ and CD4+ TDO-reactive T cells in vitro. TDO-reactive CD8+ T cells were able to kill HLA-matched tumor cells of different origin. Interestingly, the processed and presented TDO-derived epitopes varied between different cancer cells. With respect to CD4+ TDO-reactive T cells, in vitro expanded T-cell cultures comprised a Th1 and/or a Treg phenotype. In summary, our data demonstrate that the immune modulating enzyme TDO is a target for CD8+ and CD4+ T cell responses both in healthy subjects as well as patients with cancer; notably, however, the functional phenotype of these T-cell responses differ depending on the respective conditions of the host.


Cytotherapy | 2017

Dendritic cell vaccination in combination with docetaxel for patients with metastatic castration-resistant prostate cancer: A randomized phase II study

Per Kongsted; Troels Holz Borch; Eva Ellebaek; Trine Zeeberg Iversen; Rikke Andersen; Özcan Met; Morten Hartvig Hansen; Henriette Lindberg; Lisa Sengeløv; Inge Marie Svane

BACKGROUND AIMS We investigated whether the addition of an autologous dendritic cell-based cancer vaccine (DCvac) induces an immune response in patients with metastatic castration-resistant prostate cancer treated with docetaxel. METHODS Forty-three patients were randomized 1:1 to receive up to 10 cycles of docetaxel alone, 75 mg/m2 every 3 weeks or in combination with DCvac. Monocytes were harvested following a leukapheresis procedure, matured ex vivo and subsequently transfected with messenger RNA encoding multiple tumor-associated antigens (TAAs). DCvac was administered intradermally twice through treatment cycles 1-4 and once through treatment cycles 5-10. Immune cell composition and antigen-specific responses were analyzed using flow cytometry, ELISpot and delayed type hypersensitivity (DTH) tests. Toxicity was graded according to Common Terminology Criteria for Adverse Events version 3.0. Progression-free survival (PFS) and disease-specific survival (DSS) was calculated using the Kaplan-Meier method. RESULTS Prostate-specific antigen responses were similar in patients treated with docetaxel alone and combination therapy (58% versus 38%; P = 0.21). PFS and DSS were comparable: 5.5 versus 5.7 months (P = 0.62, log rank) and 21.9 versus 25.1 months (P = 0.60, log rank). Nine (50%) and 14 (78%) patients treated with docetaxel and DCvac had a TAA-specific or vaccine-specific immune response in the ELISpot and DTH analysis, respectively. Vaccine induced toxicity was limited to local reactions. Decline in myeloid-derived suppressor cells at the third treatment cycle was found to be an independent predictor of DSS. CONCLUSIONS The addition of DCvac was safe. Immune responses were detected in approximately half of the patients investigated.


Journal for ImmunoTherapy of Cancer | 2014

Adoptive cell therapy with tumor infiltrating lymphocytes and intermediate dose IL-2 for metastatic melanoma

Rikke Andersen; Marco Donia; Troels Holz Borch; Eva Ellebæk Steensgaard; Trine Zeeberg Iversen; Per Kongsted; Mads Hald Andersen; Per thor Straten; Inge Marie Svane

Meeting abstracts Adoptive cell therapy (ACT) with tumor infiltrating lymphocytes (TILs) achieved impressive clinical results in several single institution Phase I/II clinical trials performed outside of Europe, and holds the promise to enter the mainstream of standard melanoma care in the near


Annals of Oncology | 2018

T cells isolated from patients with checkpoint inhibitor-resistant melanoma are functional and can mediate tumor regression

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.


Anti-Cancer Drugs | 2016

Cabazitaxel as second-line or third-line therapy in patients with metastatic castration-resistant prostate cancer.

Per Kongsted; Inge Marie Svane; Henriette Lindberg; Rasmus Bisbjerg; Gedske Daugaard; Lisa Sengeløv

To compare treatment outcomes in patients with metastatic castration-resistant prostate cancer treated with cabazitaxel (CA) as second-line or third-line therapy in the everyday clinical setting. Charts from 94 patients treated with CA as second-line (n=28) or third-line therapy (n=66) were evaluated. Common Terminology Criteria for Adverse Events were used to register grade 3–4 nonhematological toxicity during treatment with CA. Baseline metastatic castration-resistant prostate cancer-related prognostic factors, duration of therapy, and maximum prostate-specific antigen (PSA) percentage change were registered during treatment with CA and previous/subsequent novel androgen receptor targeting therapies. Progression-free survival (PFS) and overall survival (OS) were calculated using the Kaplan–Meier method. A median of 6 versus 5 treatment cycles was administered in patients treated with second-line and third-line CA (P=0.483). Events with grade 3–4 nonhematological toxicity were equally distributed in the two groups (32 vs. 35%, P=0.80). PSA responses were observed in 46 and 17% of patients treated with second-line and third-line CA (P=0.002). PFS (5.5 vs. 3.3 months, P=0.087, log rank) and OS (18.3 vs. 11.4 months, P=0.003, log rank) was longer in patients treated with second-line CA. OS measured from second-line abiraterone acetate/enzalutamide was similar (18.0 months) to second-line CA (P=0.883, log rank). Treatment-related toxicity was independent of CA being administered as second-line or third-line therapy. Although PFS and the frequency of PSA responders favored patients treated with second-line CA, one treatment sequence could not be considered superior to the other in this study.


Clinical Genitourinary Cancer | 2017

Clinical Impact of the Number of Treatment Cycles in First-Line Docetaxel for Patients With Metastatic Castration-Resistant Prostate Cancer.

Per Kongsted; Inge Marie Svane; Henriette Lindberg; Lisa Sengeløv

Micro‐Abstract In this retrospective study we compared survival outcome in a cohort of patients treated with 6 to 8 versus ≥ 9 cycles of docetaxel, administered as first‐line chemotherapy in metastatic castration‐resistant prostate cancer. Treatment with ≥ 9 cycles of docetaxel was found to be an independent predictor of overall survival suggesting that a higher number of treatment cycles might be beneficial in this group of patients. Background: We investigated the impact of the number of docetaxel cycles administered in patients with metastatic castration‐resistant prostate cancer (mCRPC) treated with first‐line chemotherapy. Patients and Methods: Charts from 421 consecutive patients who initiated standard treatment with docetaxel‐based chemotherapy (75 mg/m2 every 3 weeks) between 2007 and 2013 were reviewed. Patients who received < 6 cycles of docetaxel were excluded from the analysis. Remaining patients were divided into 2 groups on the basis of whether or not ≥ 9 cycles of docetaxel were administered (n = 108 and 184, respectively). Reasons for treatment discontinuation and postdocetaxel treatments were registered. Prostate‐specific antigen (PSA) responses were defined as a confirmed ≥ 50% decrease in baseline PSA levels. Overall survival (OS) was calculated from start of therapy using the Kaplan–Meier method. Cox proportional hazards were calculated to estimate the effect of clinical variables on OS. Results: OS was longer in patients treated with ≥ 9 cycles of docetaxel (21.9 months vs. 17.2 months; P < .0001, log rank). Survival also favored patients treated with ≥ 9 cycles of docetaxel when only patients ending docetaxel because of toxicity or treatment conclusion (22.3 vs. 19.4 months; P = .048, log rank) or patients who achieved a PSA response (22.3 vs. 18.7 months; P = .012, log rank) were evaluated. mCRPC‐related prognostic factors and patients who received ≥ 1 subsequent line of therapy post‐docetaxel were well balanced. Conclusion: On the basis of our retrospective findings, a superior OS was found in patients treated with ≥ 9 cycles of docetaxel when adjusting for known prognostic factors. Dose reductions might increase the number of docetaxel cycles administered.


Journal for ImmunoTherapy of Cancer | 2014

T cell therapy in combination with Vemurafenib in BRAF mutated metastatic melanoma patients

Troels Holz Borch; Rikke Andersen; Per Kongsted; Özcan Met; Mads Hald Andersen; Per thor Straten; Marco Donia; Inge Marie Svane

Background Adoptive T cell therapy (ACT) with tumor infiltrating lymphocytes (TIL) has proven to be a powerful treatment option for patients with metastatic melanoma, even when heavily pretreated, with response rates of approximately 50% and durable complete responses in about 15%. However, there is still a need for improving TIL efficacy and a promising strategy is combination with immunomodulating agents. One such is Vemurafenib (Vem), a selective BRAF inhibitor, which has been shown to induce objective responses in about 50% of treated melanoma patients expressing BRAF V600E/K , improving progression-free survival and overall survival compared to standard chemotherapy. In addition to the direct anti-cancer effect, Vem has been shown to increase T cell infiltration into tumors, up-regulate melanoma antigen expression and increase the frequency of TIL recognizing autologous melanoma cells. Combining TIL treatment and Vem is currently being investigated in other clinical trials testing different treatment schedules (ClinicalTrials.gov Identifier: NCT01659151 and NCT01585415). Methods A total of 12 patients will be included in this Phase II trial primarily to investigate safety when combining ACT and Vem. Secondarily, clinical responses will be evaluated according to RECIST and extensive immune monitoring will be performed. Patients will receive Vem orally 960 mg BID one week prior to excision of tumor material for T cell generation and continue this treatment until hospital admission. The patients will be hospitalized one week prior to TIL infusion in order to follow a preparative lymphodepleting regimen consisting of Cyclophosphamide 60 mg/kg for 2days and Fludarabine 25 mg/m 2 for 5 days. TIL infusion typically consists of 5-10 × 10 10 T cells and patients are subsequently treated with continuous interleukin-2 infusion following the decrescendo-regimen for 5 days. Patients will be evaluated 6 weeks after TIL infusion and continuously thereafter. Results


Clinical Genitourinary Cancer | 2016

Predictors of Chemotherapy-Induced Toxicity and Treatment Outcomes in Elderly Versus Younger Patients With Metastatic Castration-Resistant Prostate Cancer

Per Kongsted; Inge Marie Svane; Henriette Lindberg; Lisa Sengeløv

Collaboration


Dive into the Per Kongsted's collaboration.

Top Co-Authors

Avatar

Inge Marie Svane

Copenhagen University Hospital

View shared research outputs
Top Co-Authors

Avatar

Rikke Andersen

University of Copenhagen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Özcan Met

Copenhagen University Hospital

View shared research outputs
Top Co-Authors

Avatar

Lisa Sengeløv

Copenhagen University Hospital

View shared research outputs
Top Co-Authors

Avatar

Marco Donia

University of Copenhagen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mads Hald Andersen

Copenhagen University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge