Network


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

Hotspot


Dive into the research topics where Gunnar Kvalheim is active.

Publication


Featured researches published by Gunnar Kvalheim.


Journal of Clinical Oncology | 2003

High-Dose Therapy Improves Progression-Free Survival and Survival in Relapsed Follicular Non-Hodgkin's Lymphoma: Results From the Randomized European CUP Trial

Harry C. Schouten; Wendi Qian; Stein Kvaloy; Adolfo Porcellini; Hans Hagberg; Hans Erik Johnsen; Jeanette K. Doorduijn; Matthew R. Sydes; Gunnar Kvalheim

PURPOSE To determine, in a randomized clinical trial, whether high-dose therapy (HDT) followed by autologous stem-cell transplantation is more effective than standard treatment with regard to progression-free survival (PFS) and overall survival (OS) in patients with relapsed follicular non-Hodgkins lymphoma; and to assess the additional value of B-cell purging of the stem-cell graft with regards to PFS and OS. PATIENTS AND METHODS Patients received three cycles of chemotherapy. Responding patients with limited bone marrow infiltration were eligible for random assignment to three further cycles of chemotherapy (C), unpurged HDT (U), or purged HDT (P). RESULTS Between August 1993 and April 1997, 140 patients were registered from 36 centers internationally, and 89 were randomly assigned. Reasons for not randomizing included patient refusal, early progression, or death on induction therapy. With a 69-month median follow-up, the log-rank P value for PFS and OS were.0037 and.079, respectively. For PFS, the hazard ratios (95% CIs) for U versus C, P versus C, and P versus U were 0.33 (0.16 to 0.70), 0.38 (0.19 to 0.79), and 1.02 (0.51 to 2.05), respectively. The hazard ratio (95% CI) for C versus U + P was 0.30 (0.15 to 0.61). Hazard ratios (95% CIs) for OS were 0.43 (0.18 to 1.06), 0.43 (0.18 to 1.02), and 0.72 (0.32 to 1.63). For C versus U + P, the hazard ratio (95% CI) was 0.40 (0.18 to 0.89). Kaplan-Meier estimates (95% CIs) of 2-year PFS for C, U, and P were 26% (8% to 44%), 58% (37% to 79%), and 55% (34% to 75%), respectively. OS at 4 years for C, U, and P are 46% (25% to 67%), 71% (52% to 91%), and 77% (60% to 95%) respectively. CONCLUSION HDT significantly improves PFS and OS. There is no clear evidence of benefit through purging.


Cancer | 2005

Snail, Slug, and Smad-interacting protein 1 as novel parameters of disease aggressiveness in metastatic ovarian and breast carcinoma†‡

Sivan Elloul; Mari Bukholt Elstrand; Jahn M. Nesland; Claes G. Tropé; Gunnar Kvalheim; Iris Goldberg; Reuven Reich; Ben Davidson

It was demonstrated previously that the Snail family of transcription factors and Smad‐interacting protein 1 (Sip1) regulate E‐cadherin and matrix metalloproteinase 2 (MMP‐2) expression, cellular morphology, and invasion in carcinoma. For the current study, the authors analyzed the relation between the expression of Snail, Slug, and Sip1; the expression of MMP‐2 and E‐cadherin; and clinical parameters in patients with metastatic ovarian and breast carcinoma.


Journal of Clinical Oncology | 2003

Detection of Isolated Tumor Cells in Bone Marrow Is an Independent Prognostic Factor in Breast Cancer

Elin Borgen; Rolf Kåresen; Gunnar Kvalheim; J. M. Nesland; Hanne Qvist; Ellen Schlichting; Torill Sauer; Jan Janbu; T. Harbitz; Bjørn Naume

PURPOSE This study was performed to disclose the clinical impact of isolated tumor cell (ITC) detection in bone marrow (BM) in breast cancer. PATIENTS AND METHODS BM aspirates were collected from 817 patients at primary surgery. Tumor cells in BM were detected by immunocytochemistry using anticytokeratin antibodies (AE1/AE3). Analyses of the primary tumor included histologic grading, vascular invasion, and immunohistochemical detection of c-erbB-2, cathepsin D, p53, and estrogen receptor (ER)/progesterone receptor (PgR) expression. These analyses were compared with clinical outcome. The median follow-up was 49 months. RESULTS ITC were detected in 13.2% of the patients. The detection rate rose with increasing tumor size (P =.011) and lymph node involvement (P <.001). Systemic relapse and death from breast cancer occurred in 31.7% and 26.9% of the BM-positive patients versus 13.7% and 10.9% of BM-negative patients, respectively (P <.001). Analyzing node-positive and node-negative patients separately, ITC positivity was associated with poor prognosis in the node-positive group and in node-negative patients not receiving adjuvant therapy (T1N0). In multivariate analysis, ITC in BM was an independent prognostic factor together with node, tumor, and ER/PgR status, histologic grade, and vascular invasion. In separate analysis of the T1N0 patients, histologic grade was independently associated with both distant disease-free survival (DDFS) and breast cancer-specific survival (BCSS), ITC detection was associated with BCSS, and vascular invasion was associated with DDFS. CONCLUSION ITC in BM is an independent predictor of DDFS and BCSS. An unfavorable prognosis was observed for node-positive patients and for node-negative patients not receiving systemic therapy. A combination of several independent prognostic factors can classify subgroups of patients into excellent and high-risk prognosis groups.


Clinical Cancer Research | 2004

Isolated tumor cells in bone marrow three years after diagnosis in disease-free breast cancer patients predict unfavorable clinical outcome

Elin Borgen; Rolf Kåresen; Hanne Qvist; Jan Janbu; Gunnar Kvalheim; Jahn M. Nesland; Bjørn Naume

Purpose: The aim of the study was to explore the value of analyzing bone marrow (BM) for the presence of isolated tumor cell(s) (ITCs) in disease-free breast cancer patients 3 years after diagnosis. Experimental Design: ITCs in BM at operation was found to be an independent prognostic factor in 817 breast cancer patients. Among these, 356 disease-free patients were analyzed with a second BM after 3 years follow-up (median 40 months, SD 3 months, range 29–52). ITC was detected by immunocytochemistry with anticytokeratine antibodies (AE1/AE3). Results: The population consisted of 70% T1 and 72% node-negative patients. ITC in BM was detected in 15%. At a median of 25 months since the second BM aspiration (66 months since diagnosis), 32 patients had developed relapse, 12 local and 20 systemic. Of the patients with ITC in BM, 21% relapsed compared with 7% of the ITC-negative patients (P < 0.001). Ten patients died of breast cancer. Survival analyses showed that ITC in BM predicted reduced distant disease-free survival (DDFS) and breast cancer specific survival (BCSS; P < 0.001, log-rank test). Uni-and multivariate analyses of the prognostic value of N, T, estrogen receptor/progesterone receptor, and BM status, histological grade, vascular invasion, p53-, c-erb-B2-, and cathepsin D expression were performed. BM status was the only independent prognostic factor for both DDFS and BCSS, whereas c-erbB-2 and N status were independent for BCSS and vascular invasion and T status for DDFS. Conclusions: ITC in BM 3 years after diagnosis in disease-free breast cancer patients is an independent prognostic factor. Detection of residual disease by BM analysis at follow-up may unravel insufficient adjuvant treatment. The clinical implications should be further explored.


The Journal of Pathology | 1998

Immunocytochemical detection of isolated epithelial cells in bone marrow: non‐specific staining and contribution by plasma cells directly reactive to alkaline phosphatase

Elin Borgen; Klaus Beiske; Sissel Trachsel; Jahn M. Nesland; Gunnar Kvalheim; Tove Karin Herstad; Ellen Schlichting; Hanne Qvist; Bjørn Naume

Detection of isolated tumour cells (TCs) in bone marrow (BM) from epithelial cancer patients by immunocytochemical (ICC) analysis seems to predict future relapse, but the reported percentages of positive BMs among patients with localized cancer show large variations and the number of detected TCs is low. This emphasizes the importance of thoroughly testing the methods in use. This study was performed to clarify to what extent positive staining of haematopoietic cells (HCs) interferes with the ICC detection of epithelial cells in BM. BM mononuclear cells (MNCs) from normal donors and stage I–II breast cancer patients were stained with anti‐cytokeratin (CK) and isotype control monoclonal antibodies (MAbs) followed by alkaline phosphatase (AP)‐based visualization of immunolabelled cells. In the ICC staining of normal donors by the anti‐CK MAbs AE1/AE3 or A45‐B/B3, rare immunoreactive cells were detected in 7/20 and 8/19 BMs, respectively. Morphological examination recognized all these cells as typical HCs. In the breast cancer patients (n=257), anti‐CK‐positive cells were detected in 26·6 per cent, excluding cells with HC morphology. Using the same morphological criteria, isotype control‐positive cells were detected in 5·4 per cent of patients. Some of the false‐positive events were further analysed and cells with strong reactivity against the AP enzyme alone were detected. Double ICC staining recognized the majority of these AP directly‐reactive cells as CD45‐negative and human Ig kappa/lambda‐positive, in accordance with the phenotype of mature plasma cells. Morphological evaluation and adequate controls are important to ensure the diagnostic specificity of micrometastases in BM. It is recommended that the number of BM MNCs included in negative controls should equal the number of cells in the diagnostic specimens.


Cancer Gene Therapy | 2006

Phase I/II trial of melanoma therapy with dendritic cells transfected with autologous tumor-mRNA

Jon Amund Kyte; L. J. Mu; Steinar Aamdal; Gunnar Kvalheim; Svein Dueland; M. Hauser; Hans Petter Gullestad; T. Ryder; Kari Lislerud; H. Hammerstad; Gustav Gaudernack

We have developed an individualized melanoma vaccine based on transfection of autologous dendritic cells (DCs) with autologous tumor-mRNA. Dendritic cells loaded with complete tumor-mRNA may generate an immune response against a broad repertoire of antigens, including unique patient-specific antigens. The purpose of the present phase I/II trial was to evaluate the feasibility and safety of the vaccine, and the ability of the DCs to elicit T-cell responses in melanoma patients. Further, we compared intradermal (i.d.) and intranodal (i.n.) vaccine administration. Twenty-two patients with advanced malignant melanoma were included, each receiving four weekly vaccines. Monocyte-derived DCs were transfected with tumor-mRNA by electroporation, matured and cryopreserved. We obtained successful vaccine production for all patients elected. No serious adverse effects were observed. A vaccine-specific immune response was demonstrated in 9/19 patients evaluable by T-cell assays (T-cell proliferation/interferon-γ ELISPOT) and in 8/18 patients evaluable by delayed-type hypersensitivity (DTH) reaction. The response was demonstrated in 7/10 patients vaccinated intradermally and in 3/12 patients vaccinated intranodally. We conclude that immuno-gene-therapy with the described DC-vaccine is feasible and safe, and that the vaccine can elicit in vivo T-cell responses against antigens encoded by the transfected tumor-mRNA. The response rates do not suggest an advantage in applying i.n. vaccination.


Cancer Cell | 2014

Myelodysplastic Syndromes Are Propagated by Rare and Distinct Human Cancer Stem Cells In Vivo.

Petter S. Woll; Una Kjällquist; Onima Chowdhury; Helen Doolittle; David C. Wedge; Supat Thongjuea; Mtakai Ngara; Kristina Anderson; Qiaolin Deng; Adam Mead; L Stenson; Alice Giustacchini; Eleni Giannoulatou; Stephen Taylor; Mohsen Karimi; Christian Scharenberg; Teresa Mortera-Blanco; Iain C Macaulay; Sally Ann Clark; Ingunn Dybedal; Dag Josefsen; Pierre Fenaux; Peter Hokland; Mette Holm; Mario Cazzola; Luca Malcovati; Sudhir Tauro; David G. Bowen; Jacqueline Boultwood; Andrea Pellagatti

Evidence for distinct human cancer stem cells (CSCs) remains contentious and the degree to which different cancer cells contribute to propagating malignancies in patients remains unexplored. In low- to intermediate-risk myelodysplastic syndromes (MDS), we establish the existence of rare multipotent MDS stem cells (MDS-SCs), and their hierarchical relationship to lineage-restricted MDS progenitors. All identified somatically acquired genetic lesions were backtracked to distinct MDS-SCs, establishing their distinct MDS-propagating function in vivo. In isolated del(5q)-MDS, acquisition of del(5q) preceded diverse recurrent driver mutations. Sequential analysis in del(5q)-MDS revealed genetic evolution in MDS-SCs and MDS-progenitors prior to leukemic transformation. These findings provide definitive evidence for rare human MDS-SCs in vivo, with extensive implications for the targeting of the cells required and sufficient for MDS-propagation.


International Journal of Cancer | 2006

Comparison of the clinical significance of occult tumor cells in blood and bone marrow in breast cancer

Elin Borgen; Cecilie Schirmer; Rolf Kåresen; Gunnar Kvalheim; Jahn M. Nesland; Bjørn Naume

Immunocytochemical (ICC) detection of disseminated tumor cells (DTC) in bone marrow (BM) in early breast cancer is an independent prognostic factor. The significance of circulating tumor cells (CTC) in peripheral blood (PB) needs further exploration and comparison to DTC detection. PB and BM were prospectively collected from 341 breast cancer patients median 40 months after operation. PB samples were analyzed for tumor cells by a negative immunomagnetic technique (10 × 106 cells/test). BM aspirates were analyzed by standard ICC (2 × 106 cells/test). CTC were present in 10% of the patients and DTC in 14%. Thirty‐seven relapses and 14 breast cancer deaths have occurred at median 66 months after diagnosis. Both CTC‐status and DTC‐status were significantly associated with disease free survival (DFS) (event rate: CTC‐positive 26.5% vs. CTC‐negative 9.1%; DTC‐positive 29.2% vs. DTC‐negative 7.8%) (p < 0.001/p < 0.001, log rank) and breast cancer specific survival (event rate: CTC‐positive 17.6% vs. CTC‐negative 2.6%; DTC‐positive 12.5% vs. DTC‐negative 2.7%) (p < 0.001/p < 0.001). The presence of both CTC and DTC (n = 8) resulted in an especially poor prognosis (p < 0.001). In node negative patients, DTC‐status, but not CTC‐status, predicted differences in DFS (p = 0.006 vs. p = 0.503). Excluding 23 patients with breast cancer‐related events prior to the sample collections, CTC detection was not significantly associated with DFS/distant‐DFS (p = 0.158/0.193), in contrast to DTC detection (p < 0.001/<0.001). Presence of CTC and absence of DTC did not affect DFS (p = 0.516). Applied to early stage disease, CTC analysis of increased volumes of PB appears less sensitive and prognostic than standard DTC analysis. Currently, this does not support an exchange of BM with PB for analysis of occult tumor cells.


Bone Marrow Transplantation | 2001

High-dose therapy with autologous stem cell transplantation in patients with peripheral T cell lymphomas

Anne Kirsti Blystad; Gunilla Enblad; Stein Kvaløy; Å Berglund; Jan Delabie; Harald Holte; K. Carlson; Gunnar Kvalheim; Mats Bengtsson; Hans Hagberg

Peripheral T cell lymphomas (PTCL) have a poorer prognosis after conventional treatment than do high-grade B cell lymphomas. The place for high-dose therapy (HDT) with autologous stem cell support in these patients is still not clear. Forty patients, 10 women and 30 men, median age 41.5 years (range 16–61) with PTCL were treated with HDT and autologous stem cell support at The Norwegian Radium Hospital, Oslo, Norway and The University Hospital, Uppsala, Sweden, between February 1990 and September 1999. The histologic subtypes were: PTCL unspecified, 20 patients; intestinal, two patients; angioimmunoblastic (AILD), two patients; angiocentric, two patients and anaplastic large cell lymphoma (ALCL), 14 patients. All patients had chemosensitive disease and had received anthracycline-containing regimens prior to transplantation. At the time of HDT, 17 patients were in first PR or CR and 23 were in second or third PR or CR. Conditioning regimens were BEAM in 15 patients, BEAC in 14 patients, cyclophosphamide and total body irradiation (TBI) in eight patients, BEAC, without etoposide and TBI in one patient and mitoxantrone and melphalan in two patients. There were three (7.5%) treatment-related deaths. The estimated overall survival (OS) at 3 years was 58%, the event-free survival (EFS) 48% and the relapse-free survival (RFS) 56%, with a median follow-up of 36 months (range 7–100) for surviving patients. The patients with ALCL tended to have a better prognosis compared to those with other PTCL subtypes, OS 79% vs 44%, respectively. In conclusion, patients with chemosensitive PTCL who are failing to achieve CR with first-line chemotherapy or are in relapse can successfully be treated with HDT and autologous stem cell support. Bone Marrow Transplantation (2001) 27, 711–716.


Molecular Oncology | 2007

Presence of bone marrow micrometastasis is associated with different recurrence risk within molecular subtypes of breast cancer

Bjørn Naume; Xi Zhao; Marit Synnestvedt; Elin Borgen; Hege G. Russnes; Ole Christian Lingjærde; Maria Strømberg; Gunnar Kvalheim; Rolf Kåresen; Jahn M. Nesland; Anne Lise Børresen-Dale; Therese Sørlie

Expression profiles of primary breast tumors were investigated in relation to disseminated tumor cells (DTCs) in bone marrow (BM) in order to increase our understanding of the dissemination process. Tumors were classified into five pre‐defined molecular subtypes, and presence of DTC identified (at median 85 months follow‐up) a subgroup of luminal A patients with particular poor outcome (p=0.008). This was not apparent for other tumor subtypes. Gene expression profiles associated with DTC and with systemic relapse for luminal A patients were identified. This study suggests that DTC in BM differentially distinguishes clinical outcome in patients with luminal A type tumors and that DTC‐associated gene expression analysis may identify genes of potential importance in tumor dissemination.

Collaboration


Dive into the Gunnar Kvalheim's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Harald Holte

Oslo University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Steinar Aamdal

Oslo University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Iris Bigalke

Oslo University Hospital

View shared research outputs
Top Co-Authors

Avatar

Bjørn Naume

Oslo University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge