Network


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

Hotspot


Dive into the research topics where Therese Törngren is active.

Publication


Featured researches published by Therese Törngren.


Clinical Cancer Research | 2012

Molecular Profiling Reveals Low- and High-Grade Forms of Primary Melanoma

Katja Harbst; Johan Staaf; Martin Lauss; Anna Karlsson; Anna Måsbäck; Iva Johansson; Pär-Ola Bendahl; Johan Vallon-Christersson; Therese Törngren; Henrik Ekedahl; Jürgen Geisler; Mattias Höglund; Markus Ringnér; Lotta Lundgren; Karin Jirström; Håkan Olsson; Christian Ingvar; Åke Borg; Hensin Tsao; Göran Jönsson

Purpose: For primary melanomas, tumor thickness, mitotic rate, and ulceration are well-laid cornerstones of prognostication. However, a molecular exposition of melanoma aggressiveness is critically missing. We recently uncovered a four-class structure in metastatic melanoma, which predicts outcome and informs biology. This raises the possibility that a molecular structure exists even in the early stages of melanoma and that molecular determinants could underlie histophenotype and eventual patient outcome. Experimental Design: We subjected 223 archival primary melanomas to a horizontally integrated analysis of RNA expression, oncogenic mutations at 238 lesions, histomorphometry, and survival data. Results: Our previously described four-class structure that was elucidated in metastatic lesions was evident within the expression space of primary melanomas. Because these subclasses converged into two larger prognostic and phenotypic groups, we used the metastatic lesions to develop a binary subtype-based signature capable of distinguishing between “high” and “low” grade forms of the disease. The two-grade signature was subsequently applied to the primary melanomas. Compared with low-grade tumors, high-grade primary melanomas were significantly associated with increased tumor thickness, mitotic rate, ulceration (all P < 0.01), and poorer relapse-free (HR = 4.94; 95% CI, 2.84–8.59), and overall (HR = 3.66; 95% CI, 2.40–5.58) survival. High-grade melanomas exhibited elevated levels of proliferation and BRCA1/DNA damage signaling genes, whereas low-grade lesions harbored higher expression of immune genes. Importantly, the molecular-grade signature was validated in two external gene expression data sets. Conclusions: We provide evidence for a molecular organization within melanomas, which is preserved across all stages of disease. Clin Cancer Res; 18(15); 4026–36. ©2012 AACR.


Human Mutation | 2010

Characterization of BRCA1 and BRCA2 deleterious mutations and variants of unknown clinical significance in unilateral and bilateral breast cancer: the WECARE study.

Åke Borg; Robert W. Haile; Kathleen E. Malone; Marinela Capanu; Ahn Diep; Therese Törngren; Sharon N. Teraoka; Colin B. Begg; Duncan C. Thomas; Patrick Concannon; Lene Mellemkjær; Leslie Bernstein; Lina Tellhed; Shanyan Xue; Eric R. Olson; Xiaolin Liang; Jessica Dolle; Anne Lise Børresen-Dale; Jonine L. Bernstein

BRCA1 and BRCA2 screening in women at high‐risk of breast cancer results in the identification of both unambiguously defined deleterious mutations and sequence variants of unknown clinical significance (VUS). We examined a population‐based sample of young women with contralateral breast cancer (CBC, n=705) or unilateral breast cancer (UBC, n=1398). We identified 470 unique sequence variants, of which 113 were deleterious mutations. The remaining 357 VUS comprised 185 unique missense changes, 60% were observed only once, while 3% occurred with a frequency of >10%. Deleterious mutations occurred three times more often in women with CBC (15.3%) than in women with UBC (5.2%), whereas combined, VUS were observed in similar frequencies in women with CBC and UBC. A protein alignment algorithm defined 16 rare VUS, occurring at highly conserved residues and/or conferring a considerable biochemical difference, the majority located in the BRCA2 DNA‐binding domain. We confirm a multiplicity of BRCA1 and BRCA2 VUS that occur at a wide range of allele frequencies. Although some VUS inflict chemical differences at conserved residues, suggesting a deleterious effect, the majority are not associated with an increased risk of CBC.


Human Mutation | 2008

Detection and precise mapping of germline rearrangements in BRCA1, BRCA2, MSH2, and MLH1 using zoom-in array comparative genomic hybridization (aCGH)†

Johan Staaf; Therese Törngren; Eva Rambech; Ulla Johansson; Camilla Persson; Gunilla Sellberg; Lina Tellhed; Mef Nilbert; Åke Borg

Disease‐predisposing germline mutations in cancer susceptibility genes may consist of large genomic rearrangements that are challenging to detect and characterize using standard PCR‐based mutation screening methods. Here, we describe a custom‐made zoom‐in microarray comparative genomic hybridization (CGH) platform of 60mer oligonucleotides. The 4 × 44 K array format provides high‐resolution coverage (200–300 bp) of 400–700 kb genomic regions surrounding six cancer susceptibility genes. We evaluate its performance to accurately detect and precisely map earlier described or novel large germline deletions or duplications occurring in BRCA1 (n=11), BRCA2 (n=2), MSH2 (n=7), or MLH1 (n=9). Additionally, we demonstrate its applicability for uncovering complex somatic rearrangements, exemplified by zoom‐in analysis of the PTEN and CDKN2A loci in breast cancer cells. The sizes of rearrangements ranged from several 100 kb, including large flanking regions, to <500‐bp deletions, including parts of single exons that would be missed by standard multiplex ligation‐dependent probe amplification (MLPA) methods. Zoom‐in CGH arrays accurately defined the borders of rearrangements, allowing convenient design of primers for sequence determination of the breakpoints. The array platform can be streamlined for a particular application, e.g., focusing on breast cancer susceptibility genes, with increased capacity using multiformat design, and represents a valuable new tool and complement for genetic screening in clinical diagnostics. Hum Mutat 29(4), 555–564, 2008.


Cancer Research | 2016

Multiregion whole-exome sequencing uncovers the genetic evolution and mutational heterogeneity of early-stage metastatic melanoma

Katja Harbst; Martin Lauss; Helena Cirenajwis; Karolin Isaksson; Frida Rosengren; Therese Törngren; Anders Kvist; Maria Johansson; Johan Vallon-Christersson; Bo Baldetorp; Åke Borg; Håkan Olsson; Christian Ingvar; Ana Carneiro; Göran Jönsson

Cancer genome sequencing has shed light on the underlying genetic aberrations that drive tumorigenesis. However, current sequencing-based strategies, which focus on a single tumor biopsy, fail to take into account intratumoral heterogeneity. To address this challenge and elucidate the evolutionary history of melanoma, we performed whole-exome and transcriptome sequencing of 41 multiple melanoma biopsies from eight individual tumors. This approach revealed heterogeneous somatic mutations in the range of 3%-38% in individual tumors. Known mutations in melanoma drivers BRAF and NRAS were always ubiquitous events. Using RNA sequencing, we found that the majority of mutations were not expressed or were expressed at very low levels, and preferential expression of a particular mutated allele did not occur frequently. In addition, we found that the proportion of ultraviolet B (UVB) radiation-induced C>T transitions differed significantly (P < 0.001) between early and late mutation acquisition, suggesting that different mutational processes operate during the evolution of metastatic melanoma. Finally, clinical history reports revealed that patients harboring a high degree of mutational heterogeneity were associated with more aggressive disease progression. In conclusion, our multiregion tumor-sequencing approach highlights the genetic evolution and non-UVB mutational signatures associated with melanoma development and progression, and may provide a more comprehensive perspective of patient outcome. Cancer Res; 76(16); 4765-74. ©2016 AACR.


Breast Cancer Research | 2013

COMPLEXO: identifying the missing heritability of breast cancer via next generation collaboration

Melissa C. Southey; Daniel J. Park; Tú Nguyen-Dumont; Ian G. Campbell; Ella R. Thompson; Alison H. Trainer; Georgia Chenevix-Trench; Jacques Simard; Martine Dumont; Penny Soucy; Mads Thomassen; Lars Jønson; Inge Søkilde Pedersen; Thomas V O Hansen; Heli Nevanlinna; Sofia Khan; Olga M. Sinilnikova; Sylvie Mazoyer; Fabienne Lesueur; Francesca Damiola; Rita K. Schmutzler; Alfons Meindl; Eric Hahnen; Michael R. Dufault; T. L. Chris Chan; Ava Kwong; Rosa B. Barkardottir; Paolo Radice; Paolo Peterlongo; Peter Devilee

Linkage analysis, positional cloning, candidate gene mutation scanning and genome-wide association study approaches have all contributed significantly to our understanding of the underlying genetic architecture of breast cancer. Taken together, these approaches have identified genetic variation that explains approximately 30% of the overall familial risk of breast cancer, implying that more, and likely rarer, genetic susceptibility alleles remain to be discovered.


Annals of Oncology | 2016

Targeted sequencing of BRCA1 and BRCA2 across a large unselected breast cancer cohort suggests that one-third of mutations are somatic

Christof Winter; Martin Nilsson; Eleonor Olsson; Anthony George; Yilun Chen; Anders Kvist; Therese Törngren; Johan Vallon-Christersson; Cecilia Hegardt; Jari Häkkinen; Göran Jönsson; Dorthe Grabau; Martin Malmberg; Ulf Kristoffersson; Martin Rehn; Sofia K. Gruvberger-Saal; Christer Larsson; Åke Borg; Niklas Loman; Lao H. Saal

We carried out targeted sequencing of BRCA1/2 in an unselected cohort of patients diagnosed with primary breast cancer within a population without strong founder mutations. Eleven percent of cases harbored a germline or somatic BRCA1/2 mutation, and the ratio of germline versus somatic mutation was 2 : 1. This has implications for treatment, genetic counseling, and interpretation of tumor-only testing.


Nature Communications | 2017

Mutational and putative neoantigen load predict clinical benefit of adoptive T cell therapy in melanoma

Martin Lauss; Marco Donia; Katja Harbst; Rikke Andersen; Shamik Mitra; Frida Rosengren; Maryem Salim; Johan Vallon-Christersson; Therese Törngren; Anders Kvist; Markus Ringnér; Inge Marie Svane; Göran Jönsson

Adoptive T-cell therapy (ACT) is a highly intensive immunotherapy regime that has yielded remarkable response rates and many durable responses in clinical trials in melanoma; however, 50–60% of the patients have no clinical benefit. Here, we searched for predictive biomarkers to ACT in melanoma. Whole exome- and transcriptome sequencing and neoantigen prediction were applied to pre-treatment samples from 27 patients recruited to a clinical phase I/II trial of ACT in stage IV melanoma. All patients had previously progressed on other immunotherapies. We report that clinical benefit is associated with significantly higher predicted neoantigen load. High mutation and predicted neoantigen load are significantly associated with improved progression-free and overall survival. Further, clinical benefit is associated with the expression of immune activation signatures including a high MHC-I antigen processing and presentation score. These results improve our understanding of mechanisms behind clinical benefit of ACT in melanoma.Adoptive T cell therapy (ACT) has yielded high response rates in melanoma, however 50–60% of patients experience no clinical benefit. Here, the authors identify predictive biomarkers, high non-synonymous mutation and high expressed neoantigen load, that associate with clinical benefit in ACT melanoma patients.


Oncotarget | 2015

Remarkable similarities of chromosomal rearrangements between primary human breast cancers and matched distant metastases as revealed by whole-genome sequencing.

Man-Hung Eric Tang; Malin Dahlgren; Christian Brueffer; Tamara Tjitrowirjo; Christof Winter; Yilun Chen; Eleonor Olsson; Kun Wang; Therese Törngren; Martin Sjöström; Dorthe Grabau; Pär-Ola Bendahl; Lisa Rydén; Emma Niméus; Lao H. Saal; Åke Borg; Sofia K. Gruvberger-Saal

To better understand and characterize chromosomal structural variation during breast cancer progression, we enumerated chromosomal rearrangements for 11 patients by performing low-coverage whole-genome sequencing of 11 primary breast tumors and their 13 matched distant metastases. The tumor genomes harbored a median of 85 (range 18-404) rearrangements per tumor, with a median of 82 (26-310) in primaries compared to 87 (18-404) in distant metastases. Concordance between paired tumors from the same patient was high with a median of 89% of rearrangements shared (range 61-100%), whereas little overlap was found when comparing all possible pairings of tumors from different patients (median 3%). The tumors exhibited diverse genomic patterns of rearrangements: some carried events distributed throughout the genome while others had events mostly within densely clustered chromothripsis-like foci at a few chromosomal locations. Irrespectively, the patterns were highly conserved between the primary tumor and metastases from the same patient. Rearrangements occurred more frequently in genic areas than expected by chance and among the genes affected there was significant enrichment for cancer-associated genes including disruption of TP53, RB1, PTEN, and ESR1, likely contributing to tumor development. Our findings are most consistent with chromosomal rearrangements being early events in breast cancer progression that remain stable during the development from primary tumor to distant metastasis.


Molecular Oncology | 2017

NF1-mutated melanoma tumors harbor distinct clinical and biological characteristics

Helena Cirenajwis; Martin Lauss; Henrik Ekedahl; Therese Törngren; Anders Kvist; Lao H. Saal; Håkan Olsson; Johan Staaf; Ana Carneiro; Christian Ingvar; Katja Harbst; Nicholas K. Hayward; Göran Jönsson

In general, melanoma can be considered as a UV‐driven disease with an aggressive metastatic course and high mutational load, with only few tumors (acral, mucosal, and uveal melanomas) not induced by sunlight and possessing a lower mutational load. The most commonly activated pathway in melanoma is the mitogen‐activated protein kinase (MAPK) pathway. However, the prognostic significance of mutational stratification is unclear and needs further investigation. Here, in silico we combined mutation data from 162 melanomas subjected to targeted deep sequencing with mutation data from three published studies. Tumors from 870 patients were grouped according to BRAF, RAS, NF1 mutation or triple‐wild‐type status and correlated with tumor and patient characteristics. We found that the NF1‐mutated subtype had a higher mutational burden and strongest UV mutation signature. Searching for co‐occurring mutated genes revealed the RASopathy genes PTPN11 and RASA2, as well as another RAS domain‐containing gene RASSF2 enriched in the NF1 subtype after adjustment for mutational burden. We found that a larger proportion of the NF1‐mutant tumors were from males and with older age at diagnosis. Importantly, we found an increased risk of death from melanoma (disease‐specific survival, DSS; HR, 1.9; 95% CI, 1.21–3.10; P = 0.046) and poor overall survival (OS; HR, 2.0; 95% CI, 1.28–2.98; P = 0.01) in the NF1 subtype, which remained significant after adjustment for age, gender, and lesion type (DSS P = 0.03, OS P = 0.06, respectively). Melanoma genomic subtypes display different biological and clinical characteristics. The poor outcome observed in the NF1 subtype highlights the need for improved characterization of this group.


Genetics in Medicine | 2017

Individuals with FANCM biallelic mutations do not develop Fanconi anemia, but show risk for breast cancer, chemotherapy toxicity and may display chromosome fragility

Irene Catucci; Ana Osorio; Brita Arver; Guido Neidhardt; Massimo Bogliolo; Federica Zanardi; Mirko Riboni; Simone Minardi; Roser Pujol; Jacopo Azzollini; Bernard Peissel; Siranoush Manoukian; Giovanna De Vecchi; Stefano Casola; Jan Hauke; Lisa Richters; Kerstin Rhiem; Rita K. Schmutzler; Karin Wallander; Therese Törngren; Åke Borg; Paolo Radice; Jordi Surrallés; Eric Hahnen; Hans Ehrencrona; Anders Kvist; Javier Benitez; Paolo Peterlongo

PurposeMonoallelic germ-line mutations in the BRCA1/FANCS, BRCA2/FANCD1 and PALB2/FANCN genes confer high risk of breast cancer. Biallelic mutations in these genes cause Fanconi anemia (FA), characterized by malformations, bone marrow failure, chromosome fragility, and cancer predisposition (BRCA2/FANCD1 and PALB2/FANCN), or an FA-like disease presenting a phenotype similar to FA but without bone marrow failure (BRCA1/FANCS). FANCM monoallelic mutations have been reported as moderate risk factors for breast cancer, but there are no reports of any clinical phenotype observed in carriers of biallelic mutations.MethodsBreast cancer probands were subjected to mutation analysis by sequencing gene panels or testing DNA damage response genes.ResultsFive cases homozygous for FANCM loss-of-function mutations were identified. They show a heterogeneous phenotype including cancer predisposition, toxicity to chemotherapy, early menopause, and possibly chromosome fragility. Phenotype severity might correlate with mutation position in the gene.ConclusionOur data indicate that biallelic FANCM mutations do not cause classical FA, providing proof that FANCM is not a canonical FA gene. Moreover, our observations support previous findings suggesting that FANCM is a breast cancer-predisposing gene. Mutation testing of FANCM might be considered for individuals with the above-described clinical features.

Collaboration


Dive into the Therese Törngren's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge