72 results on '"Russnes HG"'
Search Results
2. Abstract P3-03-19: Tumor cell detection and immune profiling of lymph nodes from breast cancer patients by mass cytometry
- Author
-
Russnes, HG, primary, Rye, IH, additional, Huse, K, additional, Schlichting, E, additional, Garred, O, additional, and Mykelbust, JH, additional
- Published
- 2019
- Full Text
- View/download PDF
3. Abstract P3-05-04: Intra-tumor heterogeneity as a predictor of therapy response in HER2 positive breast cancer
- Author
-
Rye, IH, primary, Helland, Å, additional, Sætersdal, A, additional, Naume, B, additional, Almendro, V, additional, Polyak, K, additional, Børessen-Dale, A-L, additional, and Russnes, HG, additional
- Published
- 2012
- Full Text
- View/download PDF
4. Extracellular matrix signature identifies breast cancer subgroups with different clinical outcome
- Author
-
Bergamaschi, A, primary, Tagliabue, E, additional, Sørlie, T, additional, Naume, B, additional, Triulzi, T, additional, Orlandi, R, additional, Russnes, HG, additional, Nesland, JM, additional, Tammi, R, additional, Auvinen, P, additional, Kosma, V‐M, additional, Ménard, S, additional, and Børresen‐Dale, A‐L, additional
- Published
- 2008
- Full Text
- View/download PDF
5. Circulating tumor cells in metastatic breast cancer patients treated with immune checkpoint inhibitors - a biomarker analysis of the ALICE and ICON trials.
- Author
-
Andresen NK, Røssevold AH, Borgen E, Schirmer CB, Gilje B, Garred Ø, Lømo J, Stensland M, Nordgård O, Falk RS, Mathiesen RR, Russnes HG, Kyte JA, and Naume B
- Abstract
Immune checkpoint inhibitors (ICIs) have been introduced in breast cancer (BC) treatment and better biomarkers are needed to predict benefit. Circulating tumor cells (CTCs) are prognostic in BC, but knowledge is limited on CTCs in the context of ICI therapy. In this study, serial sampling of CTCs (CellSearch system) was evaluated in 82 patients with metastatic BC enrolled in two randomized trials investigating ICI plus chemotherapy. Programmed death-ligand 1 (PD-L1) expression on CTCs was also measured. Patients with ≥ 2 CTCs per 7.5 mL at baseline had gene expression profiles in tumor suggestive of increased T-cell activity, including increased tumor inflammation signature (TIS) in both triple-negative (P = 0.010) and hormone receptor-positive (P = 0.024) disease. Patients with luminal A BC had higher CTC levels. The association between CTC status and outcome was most apparent 4 weeks into therapy. PD-L1 expression in CTCs was observed in 6/17 CTC-positive patients and was associated with inferior survival. In conclusion, our study indicates that CTC numbers may inform on tumor immune composition, as well as prognosis. These findings suggest a potential of using CTCs as an accessible biomarker source in BC patients treated with immunotherapy., (© 2024 The Author(s). Molecular Oncology published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)
- Published
- 2024
- Full Text
- View/download PDF
6. Acta Oncologica Nordic Precision Cancer Medicine Symposium 2023 - merging clinical research and standard healthcare.
- Author
-
Bjørgo E, Fagereng GL, Russnes HG, Smeland S, Taskén K, and Helland Å
- Subjects
- Humans, Biomedical Research, Scandinavian and Nordic Countries, Delivery of Health Care standards, Congresses as Topic, Precision Medicine methods, Neoplasms therapy, Neoplasms drug therapy, Medical Oncology methods
- Published
- 2024
- Full Text
- View/download PDF
7. Impact of Prosigna test on adjuvant treatment decision in lymph node-negative early breast cancer-a prospective national multicentre study (EMIT-1).
- Author
-
Ohnstad HO, Blix ES, Akslen LA, Gilje B, Raj SX, Skjerven H, Borgen E, Janssen EAM, Mortensen E, Brekke MB, Falk RS, Schlichting E, Boge B, Songe-Møller S, Olsson P, Heie A, Mannsåker B, Vestlid MA, Kursetgjerde T, Gravdehaug B, Suhrke P, Sanchez E, Bublevic J, Røe OD, Geitvik GA, Halset EH, Rypdal MC, Langerød A, Lømo J, Garred Ø, Porojnicu A, Engebraaten O, Geisler J, Lyngra M, Hansen MH, Søiland H, Nakken T, Asphaug L, Kristensen V, Sørlie T, Nygård JF, Kiserud CE, Reinertsen KV, Russnes HG, and Naume B
- Subjects
- Humans, Female, Middle Aged, Prospective Studies, Chemotherapy, Adjuvant methods, Aged, Adult, Lymph Nodes pathology, Aged, 80 and over, Breast Neoplasms pathology, Breast Neoplasms drug therapy, Breast Neoplasms therapy
- Abstract
Background: EMIT-1 is a national, observational, single-arm trial designed to assess the value of the Prosigna, Prediction Analysis of Microarray using the 50 gene classifier (PAM50)/Risk of Recurrence (ROR), test as a routine diagnostic tool, examining its impact on adjuvant treatment decisions, clinical outcomes, side-effects and cost-effectiveness. Here we present the impact on treatment decisions., Patients and Methods: Patients with hormone receptor-positive, human epidermal growth factor receptor 2-negative pT1-pT2 lymph node-negative early breast cancer (EBC) were included. The Prosigna test and standard histopathology assessments were carried out. Clinicians' treatment decisions were recorded before (pre-Prosigna) and after (post-Prosigna) the Prosigna test results were disclosed., Results: Of 2217 patients included, 2178 had conclusive Prosigna results. The pre-Prosigna treatment decisions were: no systemic treatment (NT) in 27% of patients, endocrine treatment alone (ET) in 38% and chemotherapy (CT) followed by ET (CT + ET) in 35%. Post-Prosigna treatment decisions were 25% NT, 51% ET and 24% CT + ET, respectively. Adjuvant treatment changed in 28% of patients, including 21% change in CT use. Among patients assigned to CT + ET pre-Prosigna, 45% were de-escalated to ET post-Prosigna. Of patients assigned to ET, 12% were escalated to CT + ET and 8% were de-escalated to NT; of those assigned to NT, 18% were escalated to ET/CT + ET. CT was more frequently recommended for patients aged ≤50 years. In the subgroup with pT1c-pT2 G2 and intermediate Ki67 (0.5-1.5× local laboratory median Ki67 score), the pre-Prosigna CT treatment decision varied widely across hospitals (3%-51%). Post-Prosigna, the variability of CT use was markedly reduced (8%-24%). The correlation between Ki67 and ROR score within this subgroup was poor (r = 0.25-0.39). The median ROR score increased by increasing histological grade, but the ROR score ranges were wide (for G1 0-79, G2 0-90, G3 16-94)., Conclusion: The Prosigna test result changed adjuvant treatment decisions in all EBC clinical risk groups, markedly decreased the CT use for patients categorized as higher clinical risk pre-Prosigna and reduced treatment decision discrepancies between hospitals., (Copyright © 2024 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
8. High-throughput molecular assays for inclusion in personalised oncology trials - State-of-the-art and beyond.
- Author
-
Edsjö A, Russnes HG, Lehtiö J, Tamborero D, Hovig E, Stenzinger A, and Rosenquist R
- Subjects
- Humans, High-Throughput Nucleotide Sequencing, Clinical Trials as Topic, Medical Oncology methods, Medical Oncology trends, Precision Medicine methods, Neoplasms genetics, Neoplasms therapy, Neoplasms diagnosis, Neoplasms drug therapy, Biomarkers, Tumor
- Abstract
In the last decades, the development of high-throughput molecular assays has revolutionised cancer diagnostics, paving the way for the concept of personalised cancer medicine. This progress has been driven by the introduction of such technologies through biomarker-driven oncology trials. In this review, strengths and limitations of various state-of-the-art sequencing technologies, including gene panel sequencing (DNA and RNA), whole-exome/whole-genome sequencing and whole-transcriptome sequencing, are explored, focusing on their ability to identify clinically relevant biomarkers with diagnostic, prognostic and/or predictive impact. This includes the need to assess complex biomarkers, for example microsatellite instability, tumour mutation burden and homologous recombination deficiency, to identify patients suitable for specific therapies, including immunotherapy. Furthermore, the crucial role of biomarker analysis and multidisciplinary molecular tumour boards in selecting patients for trial inclusion is discussed in relation to various trial concepts, including drug repurposing. Recognising that today's exploratory techniques will evolve into tomorrow's routine diagnostics and clinical study inclusion assays, the importance of emerging technologies for multimodal diagnostics, such as proteomics and in vivo drug sensitivity testing, is also discussed. In addition, key regulatory aspects and the importance of patient engagement in all phases of a clinical trial are described. Finally, we propose a set of recommendations for consideration when planning a new precision cancer medicine trial., (© 2024 The Authors. Journal of Internal Medicine published by John Wiley & Sons Ltd on behalf of Association for Publication of The Journal of Internal Medicine.)
- Published
- 2024
- Full Text
- View/download PDF
9. Desmoplastic non-infantile ganglioglioma mimicking diffuse leptomeningeal glioneuronal tumor: precision diagnostics and therapeutic implications.
- Author
-
Niehusmann P, Leske H, Nygaard V, Russnes HG, Zhao S, Latysheva A, Straume Wiig U, Stankuniene B, and Ulvmoen A
- Subjects
- Humans, Diagnosis, Differential, Magnetic Resonance Imaging, Brain Neoplasms pathology, Brain Neoplasms diagnosis, Brain Neoplasms therapy, Brain Neoplasms diagnostic imaging, Ganglioglioma pathology, Ganglioglioma diagnosis, Ganglioglioma therapy, Meningeal Neoplasms pathology, Meningeal Neoplasms diagnosis
- Published
- 2024
- Full Text
- View/download PDF
10. PCM4EU and PRIME-ROSE: Collaboration for implementation of precision cancer medicine in Europe.
- Author
-
Taskén K, F Haj Mohammad S, Fagereng GL, Sørum Falk R, Helland Å, Barjesteh van Waalwijk van Doorn-Khosrovani S, Steen Carlsson K, Ryll B, Jalkanen K, Edsjö A, Russnes HG, Lassen U, Hallersjö Hult E, Lugowska I, Blay JY, Verlingue L, Abel E, Lowery MA, Krebs MG, Staal Rohrberg K, Ojamaa K, Oliveira J, Verheul HMW, Voest EE, and Gelderblom H
- Subjects
- Humans, Europe, European Union, Drug Repositioning, Clinical Trials as Topic organization & administration, Precision Medicine methods, Neoplasms therapy
- Abstract
Background: In the two European Union (EU)-funded projects, PCM4EU (Personalized Cancer Medicine for all EU citizens) and PRIME-ROSE (Precision Cancer Medicine Repurposing System Using Pragmatic Clinical Trials), we aim to facilitate implementation of precision cancer medicine (PCM) in Europe by leveraging the experience from ongoing national initiatives that have already been particularly successful., Patients and Methods: PCM4EU and PRIME-ROSE gather 17 and 24 partners, respectively, from 19 European countries. The projects are based on a network of Drug Rediscovery Protocol (DRUP)-like clinical trials that are currently ongoing or soon to start in 11 different countries, and with more trials expected to be established soon. The main aims of both the projects are to improve implementation pathways from molecular diagnostics to treatment, and reimbursement of diagnostics and tumour-tailored therapies to provide examples of best practices for PCM in Europe., Results: PCM4EU and PRIME-ROSE were launched in January and July 2023, respectively. Educational materials, including a podcast series, are already available from the PCM4EU website (http://www.pcm4eu.eu). The first reports, including an overview of requirements for the reimbursement systems in participating countries and a guide on patient involvement, are expected to be published in 2024., Conclusion: PCM4EU and PRIME-ROSE were launched in January and July 2023, respectively. Educational materials, including a podcast series, are already available from the PCM4EU website (http://www.pcm4eu.eu). The first reports, including an overview of requirements for the reimbursement systems in participating countries and a guide on patient involvement, are expected to be published in 2024., Conclusion: European collaboration can facilitate the implementation of PCM and thereby provide affordable and equitable access to precision diagnostics and matched therapies for more patients.
- Published
- 2024
- Full Text
- View/download PDF
11. Ipilimumab and nivolumab combined with anthracycline-based chemotherapy in metastatic hormone receptor-positive breast cancer: a randomized phase 2b trial.
- Author
-
Andresen NK, Røssevold AH, Quaghebeur C, Gilje B, Boge B, Gombos A, Falk RS, Mathiesen RR, Julsrud L, Garred Ø, Russnes HG, Lereim RR, Chauhan SK, Lingjærde OC, Dunn C, Naume B, and Kyte JA
- Subjects
- Female, Humans, Anthracyclines, Antineoplastic Combined Chemotherapy Protocols adverse effects, B7-H1 Antigen, Cyclophosphamide, Ipilimumab pharmacology, Ipilimumab therapeutic use, Breast Neoplasms drug therapy, Breast Neoplasms chemically induced, Nivolumab pharmacology, Nivolumab therapeutic use
- Abstract
Background: Immune checkpoint inhibitors have shown minimal clinical activity in hormone receptor-positive metastatic breast cancer (HR
+ mBC). Doxorubicin and low-dose cyclophosphamide are reported to induce immune responses and counter regulatory T cells (Tregs). Here, we report the efficacy and safety of combined programmed cell death protein-1/cytotoxic T-lymphocyte-associated protein 4 blockade concomitant with or after immunomodulatory chemotherapy for HR+ mBC., Methods: Patients with HR+ mBC starting first-/second- line chemotherapy (chemo) were randomized 2:3 to chemotherapy (pegylated liposomal doxorubicin 20 mg/m2 every second week plus cyclophosphamide 50 mg by mouth/day in every other 2-week cycle) with or without concomitant ipilimumab (ipi; 1 mg/kg every sixth week) and nivolumab (nivo; 240 mg every second week). Patients in the chemo-only arm were offered cross-over to ipi/nivo without chemotherapy. Co-primary endpoints were safety in all patients starting therapy and progression-free survival (PFS) in the per-protocol (PP) population, defined as all patients evaluated for response and receiving at least two treatment cycles. Secondary endpoints included objective response rate, clinical benefit rate, Treg changes during therapy and assessment of programmed death-ligand 1 (PD-L1), mutational burden and immune gene signatures as biomarkers., Results: Eighty-two patients were randomized and received immune-chemo (N=49) or chemo-only (N=33), 16 patients continued to the ipi/nivo-only cross-over arm. Median follow-up was 41.4 months. Serious adverse events occurred in 63% in the immune-chemo arm, 39% in the chemo-only arm and 31% in the cross-over-arm. In the PP population (N=78) median PFS in the immune-chemo arm was 5.1 months, compared with 3.6 months in the chemo-only arm, with HR 0.94 (95% CI 0.59 to 1.51). Clinical benefit rates were 55% (26/47) and 48% (15/31) in the immune-chemo and chemo-only arms, respectively. In the cross-over-arm (ipi/nivo-only), objective responses were observed in 19% of patients (3/16) and clinical benefit in 25% (4/16). Treg levels in blood decreased after study chemotherapy. High-grade immune-related adverse events were associated with prolonged PFS. PD-L1 status and mutational burden were not associated with ipi/nivo benefit, whereas a numerical PFS advantage was observed for patients with a high Treg gene signature in tumor., Conclusion: The addition of ipi/nivo to chemotherapy increased toxicity without improving efficacy. Ipi/nivo administered sequentially to chemotherapy was tolerable and induced clinical responses., Trial Registration Number: ClinicalTrials.gov Identifier: NCT03409198., Competing Interests: Competing interests: JAK has in the last 5 years received research support from Bristol Myers Squibb, F. Hoffmann-La Roche, NanoString, and NEC OncoImmunity and has previously received advisory board/lecture honoraria from pharmaceutical companies, including Bristol Myers Squibb. CQ has received honoraria for advisory board from AstraZeneca. BG has received honoraria for advisory boards from Eli Lilly, Gilead, Daiichi Sankyo, Roche, and Pierre Fabre. LJ has received lecture honoraria from Pfizer, Novartis, and AstraZeneca. AG has received travel grants or honoraria for advisory boards from Lilly, Daiichi Sankyo, Seagen, Pfizer, and AstraZeneca. HGR has received research support from Illumina and NanoString. OCL has over the last 2 years received honoraria for work as statistical advisor for Novartis. All other authors declare no competing interests., (© Author(s) (or their employer(s)) 2024. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2024
- Full Text
- View/download PDF
12. [Raising the quality of cancer treatment].
- Author
-
Helland Å, Steinskog ESS, Blix ES, Flobak Å, Brabrand S, Puco K, Niehusmann P, Meltzer S, Oppedal IA, Haug Å, Torkildsen CF, Randen U, Gilje B, Lønning PE, Gjertsen BT, Hovland R, Russnes HG, Fagereng GL, Smeland S, and Tasken K
- Published
- 2024
- Full Text
- View/download PDF
13. Long-Smoldering T-prolymphocytic Leukemia: A Case Report and a Review of the Literature.
- Author
-
Gjelberg HK, Helgeland L, Liseth K, Micci F, Sandnes M, Russnes HG, and Reikvam H
- Subjects
- Humans, Alemtuzumab therapeutic use, Leukemia, Prolymphocytic diagnosis, Leukemia, Prolymphocytic genetics, Leukemia, Prolymphocytic therapy, Leukemia, Prolymphocytic, T-Cell diagnosis, Leukemia, Prolymphocytic, T-Cell genetics, Leukemia, Prolymphocytic, T-Cell therapy
- Abstract
T-prolymphocytic leukemia (T-PLL) is a rare malignancy of mature T-cells with distinct clinical, cytomorphological, and molecular genetic features. The disease typically presents at an advanced stage, with marked leukocytosis, B symptoms, hepatosplenomegaly, and bone marrow failure. It usually follows an aggressive course from presentation, and the prognosis is often considered dismal; the median overall survival is less than one year with conventional chemotherapy. This case report describes a patient with T-PLL who, after an unusually protracted inactive phase, ultimately progressed to a highly invasive, organ-involving disease. After initial treatments failed, a novel treatment approach resulted in a significant response.
- Published
- 2023
- Full Text
- View/download PDF
14. A convolutional neural network STIFMap reveals associations between stromal stiffness and EMT in breast cancer.
- Author
-
Stashko C, Hayward MK, Northey JJ, Pearson N, Ironside AJ, Lakins JN, Oria R, Goyette MA, Mayo L, Russnes HG, Hwang ES, Kutys ML, Polyak K, and Weaver VM
- Subjects
- Humans, Female, Mechanical Phenomena, Elasticity, Collagen, Neural Networks, Computer, Microscopy, Atomic Force methods, Breast Neoplasms pathology
- Abstract
Intratumor heterogeneity associates with poor patient outcome. Stromal stiffening also accompanies cancer. Whether cancers demonstrate stiffness heterogeneity, and if this is linked to tumor cell heterogeneity remains unclear. We developed a method to measure the stiffness heterogeneity in human breast tumors that quantifies the stromal stiffness each cell experiences and permits visual registration with biomarkers of tumor progression. We present Spatially Transformed Inferential Force Map (STIFMap) which exploits computer vision to precisely automate atomic force microscopy (AFM) indentation combined with a trained convolutional neural network to predict stromal elasticity with micron-resolution using collagen morphological features and ground truth AFM data. We registered high-elasticity regions within human breast tumors colocalizing with markers of mechanical activation and an epithelial-to-mesenchymal transition (EMT). The findings highlight the utility of STIFMap to assess mechanical heterogeneity of human tumors across length scales from single cells to whole tissues and implicates stromal stiffness in tumor cell heterogeneity., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
15. Atezolizumab plus anthracycline-based chemotherapy in metastatic triple-negative breast cancer: the randomized, double-blind phase 2b ALICE trial.
- Author
-
Røssevold AH, Andresen NK, Bjerre CA, Gilje B, Jakobsen EH, Raj SX, Falk RS, Russnes HG, Jahr T, Mathiesen RR, Lømo J, Garred Ø, Chauhan SK, Lereim RR, Dunn C, Naume B, and Kyte JA
- Subjects
- Humans, Antineoplastic Combined Chemotherapy Protocols adverse effects, B7-H1 Antigen therapeutic use, Cyclophosphamide adverse effects, Double-Blind Method, Anthracyclines, Triple Negative Breast Neoplasms drug therapy, Triple Negative Breast Neoplasms pathology
- Abstract
Immune checkpoint inhibitors have shown efficacy against metastatic triple-negative breast cancer (mTNBC) but only for PD-L1
positive disease. The randomized, placebo-controlled ALICE trial ( NCT03164993 , 24 May 2017) evaluated the addition of atezolizumab (anti-PD-L1) to immune-stimulating chemotherapy in mTNBC. Patients received pegylated liposomal doxorubicin (PLD) and low-dose cyclophosphamide in combination with atezolizumab (atezo-chemo; n = 40) or placebo (placebo-chemo; n = 28). Primary endpoints were descriptive assessment of progression-free survival in the per-protocol population (>3 atezolizumab and >2 PLD doses; n = 59) and safety in the full analysis set (FAS; all patients starting therapy; n = 68). Adverse events leading to drug discontinuation occurred in 18% of patients in the atezo-chemo arm (7/40) and in 7% of patients in the placebo-chemo arm (2/28). Improvement in progression-free survival was indicated in the atezo-chemo arm in the per-protocol population (median 4.3 months versus 3.5 months; hazard ratio (HR) = 0.57; 95% confidence interval (CI) 0.33-0.99; log-rank P = 0.047) and in the FAS (HR = 0.56; 95% CI 0.33-0.95; P = 0.033). A numerical advantage was observed for both the PD-L1positive (n = 27; HR = 0.65; 95% CI 0.27-1.54) and PD-L1negative subgroups (n = 31; HR = 0.57, 95% CI 0.27-1.21). The progression-free proportion after 15 months was 14.7% (5/34; 95% CI 6.4-30.1%) in the atezo-chemo arm versus 0% in the placebo-chemo arm. The addition of atezolizumab to PLD/cyclophosphamide was tolerable with an indication of clinical benefit, and the findings warrant further investigation of PD1/PD-L1 blockers in combination with immunomodulatory chemotherapy., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
16. Functional precision cancer medicine: drug sensitivity screening enabled by cell culture models.
- Author
-
Flobak Å, Skånland SS, Hovig E, Taskén K, and Russnes HG
- Subjects
- Artificial Intelligence, Biomarkers, Tumor, Cell Culture Techniques, Early Detection of Cancer, Humans, Neoplasms drug therapy, Precision Medicine
- Abstract
Functional precision medicine is a new, emerging area that can guide cancer treatment by capturing information from direct perturbations of tumor-derived, living cells, such as by drug sensitivity screening. Precision cancer medicine as currently implemented in clinical practice has been driven by genomics, and current molecular tumor boards rely extensively on genomic characterization to advise on therapeutic interventions. However, genomic biomarkers can only guide treatment decisions for a fraction of the patients. In this review we provide an overview of the current state of functional precision medicine, highlight advances for drug-sensitivity screening enabled by cell culture models, and discuss how artificial intelligence (AI) can be coupled to functional precision medicine to guide patient stratification., Competing Interests: Declaration of interests S.S.S. has received honoraria from AbbVie and AstraZeneca, and research support from BeiGene and TG Therapeutics. Å.F. has received honoraria from Bayer, Novartis, Pierre Fabre, Amgen, and Pfizer. The other authors declare no conflicts of interest., (Copyright © 2022 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
17. Cost-effectiveness of molecularly matched off-label therapies for end-stage cancer - the MetAction precision medicine study.
- Author
-
Ree AH, Mælandsmo GM, Flatmark K, Russnes HG, Gómez Castañeda M, and Aas E
- Subjects
- Cost-Benefit Analysis, Health Care Costs, Humans, Off-Label Use, Quality-Adjusted Life Years, Randomized Controlled Trials as Topic, Neoplasms drug therapy, Neoplasms genetics, Precision Medicine
- Abstract
Background: Precision cancer medicine (PCM), frequently used for the expensive and often modestly efficacious off-label treatment with medications matched to the tumour genome of end-stage cancer, challenges healthcare resources. We compared the health effects, costs and cost-effectiveness of our MetAction PCM study with corresponding data from comparator populations given best supportive care (BSC) in two external randomised controlled trials., Methods: We designed three partitioned survival models to evaluate the healthcare costs and quality-adjusted life years (QALYs) as the main outcomes. Cost-effectiveness was calculated as the incremental cost-effectiveness ratio (ICER) of PCM relative to BSC with an annual willingness-to-pay (WTP) threshold of EUR 56,384 (NOK 605,000). One-way and probabilistic sensitivity analyses addressed uncertainty., Results: We estimated total healthcare costs (relating to next-generation sequencing (NGS) equipment and personnel wages, molecularly matched medications to the patients with an actionable tumour target and follow-up of the responding patients) and the health outcomes for the MetAction patients versus costs (relating to estimated hospital admission) and outcomes for the BSC cases. The ICERs for incremental QALYs were twice or more as high as the WTP threshold and relatively insensitive to cost decrease of the NGS procedures, while reduction of medication prices would contribute significantly towards a cost-effective PCM strategy., Conclusions: The models suggested that the high ICERs of PCM were driven by costs of the NGS diagnostics and molecularly matched medications, with a likelihood for the strategy to be cost-effective defying WTP constraints. Reducing drug expenses to half the list price would likely result in an ICER at the WTP threshold. This can be an incentive for a public-private partnership for sharing drug costs in PCM, exemplified by ongoing European initiatives., Clinicaltrials.gov, Identifier: NCT02142036.
- Published
- 2022
- Full Text
- View/download PDF
18. Correction to: Improving public cancer care by implementing precision medicine in Norway: IMPRESS-Norway.
- Author
-
Helland Å, Russnes HG, Fagereng GL, Al-Shibli K, Andersson Y, Berg T, Bjørge L, Blix E, Bjerkehagen B, Brabrand S, Cameron MG, Dalhaug A, Dietzel D, Dønnem T, Enerly E, Flobak Å, Fluge S, Gilje B, Gjertsen BT, Grønberg BH, Grønås K, Guren T, Hamre H, Haug Å, Heinrich D, Hjortland GO, Hovig E, Hovland R, Iversen AC, Janssen E, Kyte JA, von der Lippe Gythfeldt H, Lothe R, Lund JÅ, Meza-Zepeda L, Munthe-Kaas MC, Nguyen OTD, Niehusmann P, Nilsen H, Puco K, Ree AH, Riste TB, Semb K, Steinskog ESS, Stensvold A, Suhrke P, Tennøe Ø, Tjønnfjord GE, Vassbotn LJ, Aas E, Aasebø K, Tasken K, and Smeland S
- Published
- 2022
- Full Text
- View/download PDF
19. Improving public cancer care by implementing precision medicine in Norway: IMPRESS-Norway.
- Author
-
Helland Å, Russnes HG, Fagereng GL, Al-Shibli K, Andersson Y, Berg T, Bjørge L, Blix E, Bjerkehagen B, Brabrand S, Cameron MG, Dalhaug A, Dietzel D, Dønnem T, Enerly E, Flobak Å, Fluge S, Gilje B, Gjertsen BT, Grønberg BH, Grønås K, Guren T, Hamre H, Haug Å, Heinrich D, Hjortland GO, Hovig E, Hovland R, Iversen AC, Janssen E, Kyte JA, von der Lippe Gythfeldt H, Lothe R, Lund JÅ, Meza-Zepeda L, Munthe-Kaas MC, Nguyen OTD, Niehusmann P, Nilsen H, Puco K, Ree AH, Riste TB, Semb K, Steinskog ESS, Stensvold A, Suhrke P, Tennøe Ø, Tjønnfjord GE, Vassbotn LJ, Aas E, Aasebø K, Tasken K, and Smeland S
- Subjects
- Humans, Medical Oncology, Precision Medicine, Prospective Studies, Antineoplastic Agents therapeutic use, Neoplasms diagnosis, Neoplasms genetics, Neoplasms therapy
- Abstract
Background: Matching treatment based on tumour molecular characteristics has revolutionized the treatment of some cancers and has given hope to many patients. Although personalized cancer care is an old concept, renewed attention has arisen due to recent advancements in cancer diagnostics including access to high-throughput sequencing of tumour tissue. Targeted therapies interfering with cancer specific pathways have been developed and approved for subgroups of patients. These drugs might just as well be efficient in other diagnostic subgroups, not investigated in pharma-led clinical studies, but their potential use on new indications is never explored due to limited number of patients., Methods: In this national, investigator-initiated, prospective, open-label, non-randomized combined basket- and umbrella-trial, patients are enrolled in multiple parallel cohorts. Each cohort is defined by the patient's tumour type, molecular profile of the tumour, and study drug. Treatment outcome in each cohort is monitored by using a Simon two-stage-like 'admissible' monitoring plan to identify evidence of clinical activity. All drugs available in IMPRESS-Norway have regulatory approval and are funded by pharmaceutical companies. Molecular diagnostics are funded by the public health care system., Discussion: Precision oncology means to stratify treatment based on specific patient characteristics and the molecular profile of the tumor. Use of targeted drugs is currently restricted to specific biomarker-defined subgroups of patients according to their market authorization. However, other cancer patients might also benefit of treatment with these drugs if the same biomarker is present. The emerging technologies in molecular diagnostics are now being implemented in Norway and it is publicly reimbursed, thus more cancer patients will have a more comprehensive genomic profiling of their tumour. Patients with actionable genomic alterations in their tumour may have the possibility to try precision cancer drugs through IMPRESS-Norway, if standard treatment is no longer an option, and the drugs are available in the study. This might benefit some patients. In addition, it is a good example of a public-private collaboration to establish a national infrastructure for precision oncology. Trial registrations EudraCT: 2020-004414-35, registered 02/19/2021; ClinicalTrial.gov: NCT04817956, registered 03/26/2021., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
20. ALK Inhibitors in Patients With ALK Fusion-Positive GI Cancers: An International Data Set and a Molecular Case Series.
- Author
-
Ambrosini M, Del Re M, Manca P, Hendifar A, Drilon A, Harada G, Ree AH, Klempner S, Mælandsmo GM, Flatmark K, Russnes HG, Cleary JM, Singh H, Sottotetti E, Martinetti A, Randon G, Sartore-Bianchi A, Capone I, Milione M, Di Bartolomeo M, and Pietrantonio F
- Subjects
- Crizotinib therapeutic use, Humans, Prospective Studies, Protein Kinase Inhibitors therapeutic use, Carcinoma, Non-Small-Cell Lung drug therapy, Gastrointestinal Neoplasms drug therapy, Lung Neoplasms drug therapy
- Abstract
Purpose: In GI cancers, anaplastic lymphoma kinase ( ALK ) rearrangements are extremely less frequent than in non-small-cell lung cancer but may be important to offer personalized strategies of treatment in selected patients. Data about the activity and efficacy of ALK inhibitors (ALKi) in GI cancers are scarce., Materials and Methods: We assembled a clinical and molecular international data set of pretreated patients with metastatic or nonresectable cancers of GI primary tumor origin with documented ALK rearrangement treated with at least one line of ALKi. Measurable disease as per RECIST 1.1 was required for response analysis., Results: Primary tumor sites were distributed as follows: 5 (38%) pancreas, 3 (23%) right colon, and 1 (8%) for each one of gastric, duodenal, rectal, left colon, and biliary tract sites. Seven patients (54%) were treated with alectinib, 5 (38%) with crizotinib, and 1 (8%) with entrectinib. After disease progression, five patients (38%) received a subsequent ALKi treatment line, and at the time of data cutoff date, treatment was still ongoing in two patients. Five of 12 evaluable patients (41%) achieved a partial response to first-line ALKi, five patients (41%) had stable disease, and 2 (17%) had progressive disease. No complete responses were registered. At a median follow-up of 39.6 months (interquartile range: 19.8-59.5), the median progression-free survival was 5.0 months (95% CI, 3.68 to no response) and the median overall survival was 9.3 months (95% CI, 5.46 to no response)., Conclusion: Treatment with ALKi provides remarkable responses and clinical benefit in pretreated patients with ALK fusion-positive GI malignancies. Despite the rarity, ALK rearrangements represent an important therapeutic target in individual pretreated patients with GI solid tumors. Further work providing prospective clinical validation of this target is needed., Competing Interests: Marzia Del ReConsulting or Advisory Role: Sanofi, Celgene, Janssen-Cilag, Ipsen, Roche Molecular DiagnosticsSpeakers' Bureau: Sanofi, Pfizer, Novartis, Janssen-Cilag, Ipsen, AstraZeneca, Astellas Pharma, Roche, MSD Oncology Paolo MancaPatents, Royalties, Other Intellectual Property: I have a patent for a method for the identification of gene panels optimal for TMB estimation (Inst) Andrew HendifarConsulting or Advisory Role: Novartis, Ipsen, Perthera, Celgene, AbbVie, EisaiResearch Funding: Ipsen, NGM Biopharmaceuticals (Inst)Travel, Accommodations, Expenses: HalozymeOther Relationship: FibroGen Alexander DrilonHonoraria: Pfizer, Loxo/Bayer/Lilly, IASLCConsulting or Advisory Role: Ignyta, Loxo, AstraZeneca, Pfizer, Blueprint Medicines, Genentech/Roche, BeiGene, Hengrui Therapeutics, Exelixis, Bayer, Tyra Biosciences, Takeda/Millennium, BerGenBio, MORE Health, Lilly, AbbVie, 14ner Oncology/Elevation Oncology, Monopteros Therapeutics, Novartis, EMD Serono/Merck, Repare Therapeutics, Melendi, Archer, Nuvalent Inc, Janssen, Amgen, Merus, Axis Pharma, Medscape, Liberum, Med Learning, PeerView, EPG Health, JNCCN/Harborside, Ology, Clinical Care Options, touchIME, Entos, Treeline, Prelude Therapeutics, Applied Pharmaceutical Science IncSpeakers' Bureau: Helsinn Therapeutics, Beigene, Remedica Ltd, TP Therapeutics, Verastem, Ignyta/Genentech/Roche, AstraZeneca, Liberum, Loxo/Bayer/Lilly, Lungevity, NIH, PER, OncLive/MJH Life Sciences, Clinical Care Options/NCCN, Lung Cancer Canada, AIOT, Chugai Pharma, Chugai Pharma, Sirio Libanes Hospital, Answers in CME, Faculty RTP, RV MoreResearch Funding: Foundation MedicinePatents, Royalties, Other Intellectual Property: Wolters Kluwer (Royalties for Pocket Oncology)Other Relationship: Merck, GlaxoSmithKline, Teva, Taiho Pharmaceutical, Pfizer, PharmaMar, Puma Biotechnology, Merus, Boehringer Ingelheim Guilherme HaradaSpeakers' Bureau: MSD, AstraZeneca, Pfizer Anne Hansen ReeHonoraria: MSD Oncology, BMSiResearch Funding: BMSi (Inst) Samuel KlempnerStock and Other Ownership Interests: TP Therapeutics, Nuvalent IncHonoraria: NateraConsulting or Advisory Role: Lilly, Astellas Pharma, Bristol Myers Squibb, Pieris Pharmaceuticals, Merck, Daiichi Sankyo/UCB Japan, Sanofi/Aventis, MersanaResearch Funding: Leap Therapeutics (Inst), BeiGene (Inst), Silverback Therapeutics (Inst)Other Relationship: NCCN Gunhild Mari MælandsmoPatents, Royalties, Other Intellectual Property: Patent application submitted for a nine-protein/gene panel predicting response to anti VEGF therapies in combination with chemotherapy (Inst) Hege G. RussnesHonoraria: AstraZeneca (Inst), Pfizer (Inst), InCyte (Inst), Merck (Inst), Roche (Inst)Research Funding: Foundation Medicine (Inst) James M. ClearyHonoraria: Blueprint Medicines, Syros PharmaceuticalsConsulting or Advisory Role: Bristol Myers SquibbResearch Funding: Merck, Tesaro, AstraZeneca, Esperas Pharma, AbbVie (Inst), Merus (Inst), Roche/Genentech (Inst), BMS (Inst)Travel, Accommodations, Expenses: Roche, Agios, Bristol Myers Squibb Andrea Sartore-BianchiConsulting or Advisory Role: Amgen, Bayer, Sanofi, Servier, Novartis Maria Di BartolomeoHonoraria: Lilly, MSD Oncology, Servier, BMSiConsulting or Advisory Role: Lilly, MSD OncologyResearch Funding: LillyTravel, Accommodations, Expenses: Roche, Sanofi Filippo PietrantonioHonoraria: Servier, Bayer, AstraZeneca/MedImmune, Lilly, Sanofi, MSD Oncology, AmgenConsulting or Advisory Role: Amgen, Servier, MSD Oncology, MerckResearch Funding: Bristol Myers Squibb (Inst), Astrazeneca (Inst)No other potential conflicts of interest were reported.
- Published
- 2022
- Full Text
- View/download PDF
21. Incidence of breast cancer subtypes in immigrant and non-immigrant women in Norway.
- Author
-
Hjerkind KV, Johansson ALV, Trewin CB, Russnes HG, and Ursin G
- Subjects
- Biomarkers, Tumor metabolism, Female, Humans, Incidence, Receptor, ErbB-2 metabolism, Receptors, Estrogen metabolism, Receptors, Progesterone metabolism, Breast Neoplasms pathology, Emigrants and Immigrants, Triple Negative Breast Neoplasms pathology
- Abstract
Background: Breast cancer incidence differs between non-immigrants and immigrants from low- and middle-income countries. This study investigates whether immigrants also have different subtype-specific incidences., Methods: We used national health registries in Norway and calculated subtype-specific incidence rate ratios (IRRs) for invasive breast cancer among women aged 20-75 and 20-49 years between 2005 and 2015. Immigrant groups were classified by country of birth broadly defined based on WHO regional groupings. Subtype was defined using estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor 2 (HER2) status as luminal A-like (ER+ PR+ HER2-), luminal B-like/HER2- (ER+ PR- HER2-), luminal B-like/HER2+ (ER+ PR any HER2+), HER2+ (ER-PR-HER2+) and triple-negative breast cancer (TNBC) (ER-PR-HER2-)., Results: Compared to non-immigrants, incidence of the luminal A-like subtype was lower in immigrants from Sub-Saharan Africa (IRR 0.43 95% CI 0.28-0.66), South East Asia (IRR 0.63 95% CI 0.51-0.79), South Asia (IRR 0.67 95% CI 0.52-0.86) and Eastern Europe (IRR 0.86 95% CI 0.76-0.99). Immigrants from South Asia had higher rates of HER2 + tumors (IRR 2.02 95% CI 1.26-3.23). The rates of TNBC tended to be similar regardless of region of birth, except that women from South East Asia had an IRR of 0.54 (95% CI 0.32-0.91)., Conclusions: Women from Eastern Europe, Sub-Saharan Africa and Asia had different subtype-specific incidences compared to women from high-income countries (including non-immigrants). These differences in tumor characteristics between immigrant groups should be taken into consideration when planning preventive or screening strategies., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
22. Ipilimumab in a real-world population: A prospective Phase IV trial with long-term follow-up.
- Author
-
Aamdal E, Jacobsen KD, Straume O, Kersten C, Herlofsen O, Karlsen J, Hussain I, Amundsen A, Dalhaug A, Nyakas M, Schuster C, Hagene KT, Holmsen K, Russnes HG, Skovlund E, Kaasa S, Aamdal S, Kyte JA, and Guren TK
- Subjects
- Adult, Aged, Aged, 80 and over, Female, Follow-Up Studies, Humans, Male, Melanoma pathology, Middle Aged, Prognosis, Prospective Studies, Skin Neoplasms secondary, Survival Rate, Antineoplastic Agents, Immunological therapeutic use, Ipilimumab therapeutic use, Melanoma drug therapy, Skin Neoplasms drug therapy
- Abstract
Ipilimumab was the first treatment that improved survival in advanced melanoma. Efficacy and toxicity in a real-world setting may differ from clinical trials, due to more liberal eligibility criteria and less intensive monitoring. Moreover, high costs and lack of biomarkers have raised cost-benefit concerns about ipilimumab in national healthcare systems and limited its use. Here, we report the prospective, interventional study, Ipi4 (NCT02068196), which aimed to investigate the toxicity and efficacy of ipilimumab in a real-world population with advanced melanoma. This national, multicentre, phase IV trial included 151 patients. Patients received ipilimumab 3 mg/kg intravenously and were followed for at least 5 years or until death. Treatment interruption or cessation occurred in 38%, most frequently due to disease progression (19%). Treatment-associated grade 3 to 4 toxicity was observed in 28% of patients, and immune-related toxicity in 56%. The overall response rate was 9%. Median overall survival was 12.1 months (95% CI: 8.3-15.9); and progression-free survival 2.7 months (95% CI: 2.6-2.8). After 5 years, 20% of patients were alive. In a landmark analysis from 6 months, improved survival was associated with objective response (HR 0.16, P = .001) and stable disease (HR 0.49, P = .005) compared to progressive disease. Poor performance status, elevated lactate dehydrogenase and C-reactive protein were identified as biomarkers. This prospective trial represents the longest reported follow-up of a real-world melanoma population treated with ipilimumab. Results indicate safety and efficacy comparable to phase III trials and suggest that the use of ipilimumab can be based on current cost-benefit estimates., (© 2021 The Authors. International Journal of Cancer published by John Wiley & Sons Ltd on behalf of UICC.)
- Published
- 2022
- Full Text
- View/download PDF
23. Breast cancer metastasis: immune profiling of lymph nodes reveals exhaustion of effector T cells and immunosuppression.
- Author
-
Rye IH, Huse K, Josefsson SE, Kildal W, Danielsen HE, Schlichting E, Garred Ø, Riis ML, Osbreac, Lingjaerde OC, Myklebust JH, and Russnes HG
- Subjects
- CD8-Positive T-Lymphocytes, Female, Humans, Immunosuppression Therapy, Lymph Nodes pathology, Melanoma, Skin Neoplasms, T-Lymphocyte Subsets pathology, Melanoma, Cutaneous Malignant, Breast Neoplasms pathology
- Abstract
Sentinel lymph nodes are the first nodes draining the lymph from a breast and could reveal early changes in the host immune system upon dissemination of breast cancer cells. To investigate this, we performed single-cell immune profiling of lymph nodes with and without metastatic cells. Whereas no significant changes were observed for B-cell and natural killer (NK)-cell subsets, metastatic lymph nodes had a significantly increased frequency of CD8 T cells and a skewing toward an effector/memory phenotype of CD4 and CD8 T cells, suggesting an ongoing immune response. Additionally, metastatic lymph nodes had an increased frequency of TIGIT (T-cell immunoreceptor with Ig and ITIM domains)-positive T cells with suppressed TCR signaling compared with non-metastatic nodes, indicating exhaustion of effector T cells, and an increased frequency of regulatory T cells (Tregs) with an activated phenotype. T-cell alterations correlated with the percentage of metastatic tumor cells, reflecting the presence of metastatic tumor cells driving T effector cells toward exhaustion and promoting immunosuppression by recruitment or increased differentiation toward Tregs. These results show that immune suppression occurs already in early stages of tumor progression., (© 2021 The Authors. Molecular Oncology published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)
- Published
- 2022
- Full Text
- View/download PDF
24. Whole genome copy number analyses reveal a highly aberrant genome in TP53 mutant lung adenocarcinoma tumors.
- Author
-
Bjaanæs MM, Nilsen G, Halvorsen AR, Russnes HG, Solberg S, Jørgensen L, Brustugun OT, Lingjærde OC, and Helland Å
- Subjects
- Adenocarcinoma of Lung pathology, Alleles, Carcinoma, Non-Small-Cell Lung pathology, Chromosomes, Human, Pair 7, Chromosomes, Human, Pair 9, Class I Phosphatidylinositol 3-Kinases genetics, DNA Copy Number Variations, Ex-Smokers, Female, Gene Expression, Genes, erbB-1 genetics, Genes, ras genetics, Humans, Lung Neoplasms pathology, Male, Non-Smokers, Polymorphism, Single Nucleotide, Signal Transduction genetics, Smokers, TOR Serine-Threonine Kinases genetics, Adenocarcinoma of Lung genetics, Carcinoma, Non-Small-Cell Lung genetics, Gene Dosage, Genes, p53, Lung Neoplasms genetics
- Abstract
Background: Genetic alterations are common in non-small cell lung cancer (NSCLC), and DNA mutations and translocations are targets for therapy. Copy number aberrations occur frequently in NSCLC tumors and may influence gene expression and further alter signaling pathways. In this study we aimed to characterize the genomic architecture of NSCLC tumors and to identify genomic differences between tumors stratified by histology and mutation status. Furthermore, we sought to integrate DNA copy number data with mRNA expression to find genes with expression putatively regulated by copy number aberrations and the oncogenic pathways associated with these affected genes., Methods: Copy number data were obtained from 190 resected early-stage NSCLC tumors and gene expression data were available from 113 of the adenocarcinomas. Clinical and histopathological data were known, and EGFR-, KRAS- and TP53 mutation status was determined. Allele-specific copy number profiles were calculated using ASCAT, and regional copy number aberration were subsequently obtained and analyzed jointly with the gene expression data., Results: The NSCLC tumors tissue displayed overall complex DNA copy number profiles with numerous recurrent aberrations. Despite histological differences, tissue samples from squamous cell carcinomas and adenocarcinomas had remarkably similar copy number patterns. The TP53-mutated lung adenocarcinomas displayed a highly aberrant genome, with significantly altered copy number profiles including gains, losses and focal complex events. The EGFR-mutant lung adenocarcinomas had specific arm-wise aberrations particularly at chromosome7p and 9q. A large number of genes displayed correlation between copy number and expression level, and the PI(3)K-mTOR pathway was highly enriched for such genes., Conclusions: The genomic architecture in NSCLC tumors is complex, and particularly TP53-mutated lung adenocarcinomas displayed highly aberrant copy number profiles. We suggest to always include TP53-mutation status when studying copy number aberrations in NSCLC tumors. Copy number may further impact gene expression and alter cellular signaling pathways., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
25. The impact of tumor epithelial and microenvironmental heterogeneity on treatment responses in HER2+ breast cancer.
- Author
-
Janiszewska M, Stein S, Metzger Filho O, Eng J, Kingston NL, Harper NW, Rye IH, Alečković M, Trinh A, Murphy KC, Marangoni E, Cristea S, Oakes B, Winer EP, Krop IE, Russnes HG, Spellman PT, Bucher E, Hu Z, Chin K, Gray JW, Michor F, and Polyak K
- Subjects
- Adult, Aged, Aged, 80 and over, Antibodies, Monoclonal, Humanized therapeutic use, Breast Neoplasms genetics, Breast Neoplasms metabolism, Class I Phosphatidylinositol 3-Kinases metabolism, DNA Copy Number Variations, Female, Fibroblasts metabolism, Humans, Middle Aged, Mutation, Neoplasm Transplantation, Receptor, ErbB-2 antagonists & inhibitors, Receptor, ErbB-2 metabolism, Trastuzumab therapeutic use, Tumor Microenvironment, Vesicular Transport Proteins metabolism, Antineoplastic Agents, Immunological therapeutic use, Breast Neoplasms drug therapy, Class I Phosphatidylinositol 3-Kinases genetics, Drug Resistance, Neoplasm genetics, Epithelial Cells metabolism, Macrophages metabolism, Receptor, ErbB-2 genetics
- Abstract
Despite the availability of multiple human epidermal growth factor receptor 2-targeted (HER2-targeted) treatments, therapeutic resistance in HER2+ breast cancer remains a clinical challenge. Intratumor heterogeneity for HER2 and resistance-conferring mutations in the PIK3CA gene (encoding PI3K catalytic subunit α) have been investigated in response and resistance to HER2-targeting agents, while the role of divergent cellular phenotypes and tumor epithelial-stromal cell interactions is less well understood. Here, we assessed the effect of intratumor cellular genetic heterogeneity for ERBB2 (encoding HER2) copy number and PIK3CA mutation on different types of neoadjuvant HER2-targeting therapies and clinical outcome in HER2+ breast cancer. We found that the frequency of cells lacking HER2 was a better predictor of response to HER2-targeted treatment than intratumor heterogeneity. We also compared the efficacy of different therapies in the same tumor using patient-derived xenograft models of heterogeneous HER2+ breast cancer and single-cell approaches. Stromal determinants were better predictors of response than tumor epithelial cells, and we identified alveolar epithelial and fibroblastic reticular cells as well as lymphatic vessel endothelial hyaluronan receptor 1-positive (Lyve1+) macrophages as putative drivers of therapeutic resistance. Our results demonstrate that both preexisting and acquired resistance to HER2-targeting agents involve multiple mechanisms including the tumor microenvironment. Furthermore, our data suggest that intratumor heterogeneity for HER2 should be incorporated into treatment design.
- Published
- 2021
- Full Text
- View/download PDF
26. Bone marrow NG2 + /Nestin + mesenchymal stem cells drive DTC dormancy via TGFβ2.
- Author
-
Nobre AR, Risson E, Singh DK, Di Martino JS, Cheung JF, Wang J, Johnson J, Russnes HG, Bravo-Cordero JJ, Birbrair A, Naume B, Azhar M, Frenette PS, and Aguirre-Ghiso JA
- Subjects
- Bone Marrow metabolism, Female, Humans, Neoplasm Recurrence, Local metabolism, Nestin metabolism, Tumor Microenvironment, Breast Neoplasms genetics, Mesenchymal Stem Cells metabolism
- Abstract
In the bone marrow (BM) microenvironment, where breast cancer (BC) disseminated tumour cells (DTCs) can remain dormant for decades, NG2
+ /Nestin+ mesenchymal stem cells (MSCs) promote hematopoietic stem cell quiescence. Here, we reveal that periarteriolar BM-resident NG2+ /Nestin+ MSCs can also instruct BC DTCs to enter dormancy. NG2+ /Nestin+ MSCs produce TGFβ2 and BMP7 and activate a quiescence pathway dependent on TGFBRIII and BMPRII, which via p38-kinase result in p27 induction. Genetic depletion of MSCs or conditional knock-out of TGFβ2 in MSCs using an NG2-CreER driver led to bone metastatic outgrowth of otherwise dormant p27+ /Ki67- DTCs. Also ER+ BC patients without systemic recurrence displayed higher frequency of TGFβ2 and BMP7 detection in the BM. Our results provide a direct proof that HSC dormancy niches control BC DTC dormancy and suggest that aging or extrinsic factors that affect the NG2+ /Nestin+ MSC niche homeostasis may result in a break from dormancy and BC bone relapse.- Published
- 2021
- Full Text
- View/download PDF
27. ICON: a randomized phase IIb study evaluating immunogenic chemotherapy combined with ipilimumab and nivolumab in patients with metastatic hormone receptor positive breast cancer.
- Author
-
Kyte JA, Andresen NK, Russnes HG, Fretland SØ, Falk RS, Lingjærde OC, and Naume B
- Subjects
- Antineoplastic Combined Chemotherapy Protocols therapeutic use, Hormones, Humans, Ipilimumab therapeutic use, Breast Neoplasms drug therapy, Nivolumab therapeutic use
- Abstract
Background: Immunotherapy with checkpoint inhibitors (CPI) targeting PD-1 or CTLA-4 has emerged as an important treatment modality for several cancer forms. In hormone receptor positive breast cancer (HR + BC), this therapeutic approach is largely unexplored. We have started a clinical trial, ICON (CA209-9FN), evaluating CPI combined with selected chemotherapy in patients with metastatic HR + BC. The tumor lymphocyte infiltration is predictive for the effect of chemotherapy in BC. In ICON, we use anthracycline, which are considered as "immunogenic" chemotherapy, and low-dose cyclophosphamide, which has been reported to counter immunosuppressive cells., Methods: ICON is a randomized exploratory phase IIb study evaluating the safety and efficacy of combining nivolumab (nivo; anti-PD-1) and ipilimumab (ipi; anti-CTLA-4) with chemotherapy in subjects with metastatic HR + BC. Primary objectives are aassessment of toxicity and progression-free survival. The trial will enrol 75 evaluable subjects, randomized 2:3 into two arms (A:B). Patients in Arm A receive only chemotherapy, i.e. pegylated liposomal doxorubicin (PLD 20 mg/m
2 intravenously every 2nd week) + cyclophosphamide (cyclo; 50 mg per day, first 2 weeks in each 4 week cycle). Patients in Arm B receive PLD + cyclo + ipilimumab (1 mg intravenously every 6th week) + nivolumab (240 mg intravenously every 2nd week). Patients in arm A will be offered ipi + nivo after disease progression., Discussion: ICON is among the first clinical trials combining chemotherapy with PD-1 and CTLA-4 blockade, and the first in BC. There is a strong preclinical rationale for exploring if anthracyclines, which are considered to induce immunogenic cell death, synergize with CPI, and for combining PD-1 and CTLA-4 blockade, as these checkpoints are important in different phases of the immune response. If the ICON trial suggests acceptable safety and provide a signal of clinical efficacy, further studies are warranted. The cross-over patients from Arm A receiving ipilimumab/nivolumab without concomitant chemotherapy represent the first BC cohort receiving this therapy. The ICON trial includes a series of translational sub-projects addressing clinically important knowledge gaps. These studies may uncover biomarkers or mechanisms of efficacy and resistance, thereby informing the development of novel combinatory regimes and of personalised biomarker-based therapy. Trial registration NCT03409198, Jan 24th 2018; https://clinicaltrials.gov/ct2/show/record/NCT03409198.- Published
- 2020
- Full Text
- View/download PDF
28. Molecularly matched therapy in the context of sensitivity, resistance, and safety; patient outcomes in end-stage cancer - the MetAction study.
- Author
-
Ree AH, Nygaard V, Boye K, Heinrich D, Dueland S, Bergheim IR, Johansen C, Beiske K, Negård A, Lund-Iversen M, Nygaard V, Hovig E, Nakken S, Nasser S, Julsrud L, Reisse CH, Ruud EA, Kristensen VN, Flørenes VA, Geitvik GA, Lingjærde OC, Børresen-Dale AL, Russnes HG, Mælandsmo GM, and Flatmark K
- Subjects
- Adult, Aged, Antineoplastic Agents therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Carcinoma secondary, Crizotinib therapeutic use, DNA, Neoplasm analysis, Drug Resistance, Neoplasm genetics, Female, Humans, Immune Checkpoint Inhibitors therapeutic use, Irinotecan administration & dosage, Male, Middle Aged, Mutation, Neoplasms pathology, Panitumumab administration & dosage, Precision Medicine, Progression-Free Survival, Response Evaluation Criteria in Solid Tumors, Sarcoma secondary, Sequence Analysis, DNA, Signal Transduction drug effects, Survival Rate, Vemurafenib administration & dosage, Young Adult, Carcinoma drug therapy, Carcinoma genetics, Neoplasms drug therapy, Neoplasms genetics, Sarcoma drug therapy, Sarcoma genetics
- Abstract
Background: In precision cancer medicine, the challenge is to prioritize DNA driver events, account for resistance markers, and procure sufficient information for treatment that maintains patient safety. The MetAction project, exploring how tumor molecular vulnerabilities predict therapy response, first established the required workflow for DNA sequencing and data interpretation (2014-2015). Here, we employed it to identify molecularly matched therapy and recorded outcome in end-stage cancer (2016-2019). Material and methods: Metastatic tissue from 26 patients (16 colorectal cancer cases) was sequenced by the Oncomine assay. The study tumor boards interpreted called variants with respect to sensitivity or resistance to matched therapy and recommended single-agent or combination treatment if considered tolerable. The primary endpoint was the rate of progression-free survival 1.3-fold longer than for the most recent systemic therapy. The objective response rate and overall survival were secondary endpoints. Results: Both common and rare actionable alterations were identified. Thirteen patients were found eligible for therapy following review of tumor sensitivity and resistance variants and patient tolerability. The interventions were inhibitors of ALK/ROS1-, BRAF-, EGFR-, FGFR-, mTOR-, PARP-, or PD-1-mediated signaling for 2-3 cases each. Among 10 patients who received treatment until radiologic evaluation, 6 (46% of the eligible cases) met the primary endpoint. Four colorectal cancer patients (15% of the total study cohort) had objective response. The only serious adverse event was a transient colitis, which appeared in 1 of the 2 patients given PD-1 inhibitor with complete response. Apart from those two, overall survival was similar for patients who did and did not receive study treatment. Conclusions: The systematic MetAction approach may point forward to a refined framework for how to interpret the complexity of sensitivity versus resistance and patient safety that resides in tumor sequence data, for the possibly improved outcome of precision cancer medicine in future studies. ClinicalTrials.gov, identifier: NCT02142036.
- Published
- 2020
- Full Text
- View/download PDF
29. Clinical trials in the era of precision cancer medicine - for the few or for the many?
- Author
-
Russnes HG
- Subjects
- Financial Support, Humans, Norway, Clinical Trials as Topic economics, Neoplasms genetics, Precision Medicine economics
- Published
- 2020
- Full Text
- View/download PDF
30. DNA copy number motifs are strong and independent predictors of survival in breast cancer.
- Author
-
Pladsen AV, Nilsen G, Rueda OM, Aure MR, Borgan Ø, Liestøl K, Vitelli V, Frigessi A, Langerød A, Mathelier A, Engebråten O, Kristensen V, Wedge DC, Van Loo P, Caldas C, Børresen-Dale AL, Russnes HG, and Lingjærde OC
- Subjects
- Algorithms, Breast Neoplasms mortality, Breast Neoplasms therapy, Clinical Decision-Making, Databases, Genetic, Female, Gene Expression Profiling, Humans, Middle Aged, Predictive Value of Tests, Prognosis, Risk Assessment, Risk Factors, Transcriptome, Biomarkers, Tumor genetics, Breast Neoplasms genetics, DNA Copy Number Variations, Gene Dosage, Genomic Instability
- Abstract
Somatic copy number alterations are a frequent sign of genome instability in cancer. A precise characterization of the genome architecture would reveal underlying instability mechanisms and provide an instrument for outcome prediction and treatment guidance. Here we show that the local spatial behavior of copy number profiles conveys important information about this architecture. Six filters were defined to characterize regional traits in copy number profiles, and the resulting Copy Aberration Regional Mapping Analysis (CARMA) algorithm was applied to tumors in four breast cancer cohorts (n = 2919). The derived motifs represent a layer of information that complements established molecular classifications of breast cancer. A score reflecting presence or absence of motifs provided a highly significant independent prognostic predictor. Results were consistent between cohorts. The nonsite-specific occurrence of the detected patterns suggests that CARMA captures underlying replication and repair defects and could have a future potential in treatment stratification.
- Published
- 2020
- Full Text
- View/download PDF
31. An independent poor-prognosis subtype of breast cancer defined by a distinct tumor immune microenvironment.
- Author
-
Tekpli X, Lien T, Røssevold AH, Nebdal D, Borgen E, Ohnstad HO, Kyte JA, Vallon-Christersson J, Fongaard M, Due EU, Svartdal LG, Sveli MAT, Garred Ø, Frigessi A, Sahlberg KK, Sørlie T, Russnes HG, Naume B, and Kristensen VN
- Subjects
- Breast Neoplasms drug therapy, Breast Neoplasms pathology, Cell Proliferation, Computer Simulation, Epithelial-Mesenchymal Transition, Female, Genes, Neoplasm, Genetic Heterogeneity, Humans, Logistic Models, Neoadjuvant Therapy, Neoplasm Recurrence, Local pathology, Phenotype, Prognosis, Proportional Hazards Models, Risk Factors, Breast Neoplasms classification, Breast Neoplasms immunology, Tumor Microenvironment
- Abstract
How mixtures of immune cells associate with cancer cell phenotype and affect pathogenesis is still unclear. In 15 breast cancer gene expression datasets, we invariably identify three clusters of patients with gradual levels of immune infiltration. The intermediate immune infiltration cluster (Cluster B) is associated with a worse prognosis independently of known clinicopathological features. Furthermore, immune clusters are associated with response to neoadjuvant chemotherapy. In silico dissection of the immune contexture of the clusters identified Cluster A as immune cold, Cluster C as immune hot while Cluster B has a pro-tumorigenic immune infiltration. Through phenotypical analysis, we find epithelial mesenchymal transition and proliferation associated with the immune clusters and mutually exclusive in breast cancers. Here, we describe immune clusters which improve the prognostic accuracy of immune contexture in breast cancer. Our discovery of a novel independent prognostic factor in breast cancer highlights a correlation between tumor phenotype and immune contexture.
- Published
- 2019
- Full Text
- View/download PDF
32. Responsiveness to PD-1 Blockade in End-Stage Colon Cancer with Gene Locus 9p24.1 Copy-Number Gain.
- Author
-
Ree AH, Nygaard V, Russnes HG, Heinrich D, Nygaard V, Johansen C, Bergheim IR, Hovig E, Beiske K, Negård A, Børresen-Dale AL, Flatmark K, and Mælandsmo GM
- Subjects
- Colonic Neoplasms pathology, DNA Copy Number Variations, Female, Genetic Loci, Humans, Liver Neoplasms genetics, Liver Neoplasms secondary, Middle Aged, Mutation, Treatment Outcome, Antibodies, Monoclonal, Humanized therapeutic use, Antineoplastic Agents, Immunological therapeutic use, Chromosomes, Human, Pair 9 genetics, Colonic Neoplasms drug therapy, Colonic Neoplasms genetics, Liver Neoplasms drug therapy, Programmed Cell Death 1 Receptor antagonists & inhibitors
- Abstract
Most patients whose large bowel cancer has spread to other organs do not respond to immune therapy. We detected a rare gene mutation, termed 9p24.1 copy-number gain (CNG), in an otherwise incurable colorectal cancer that provoked an immune therapy response. We identified this gene mutation by gene-panel sequencing of DNA from a liver metastasis biopsy from a patient who had disease refractory to standard therapies. Following immune checkpoint blockade (ICB) with pembrolizumab (anti-PD-1), the patient experienced conversion of the tumor phenotype from one with epithelial features to that of an inflamed microenvironment, detected by high-resolution RNA sequencing. Circulating tumor DNA disappeared over the first weeks of therapy. As assessed by standard radiographic measurement, the patient had a partial response that was durable. This patient's response may support the use of histology-agnostic ICB in solid tumors that carry the rare 9p24.1 CNG., (©2019 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
33. Breast cancer quantitative proteome and proteogenomic landscape.
- Author
-
Johansson HJ, Socciarelli F, Vacanti NM, Haugen MH, Zhu Y, Siavelis I, Fernandez-Woodbridge A, Aure MR, Sennblad B, Vesterlund M, Branca RM, Orre LM, Huss M, Fredlund E, Beraki E, Garred Ø, Boekel J, Sauer T, Zhao W, Nord S, Höglander EK, Jans DC, Brismar H, Haukaas TH, Bathen TF, Schlichting E, Naume B, Luders T, Borgen E, Kristensen VN, Russnes HG, Lingjærde OC, Mills GB, Sahlberg KK, Børresen-Dale AL, and Lehtiö J
- Subjects
- Breast pathology, Breast Neoplasms genetics, Breast Neoplasms immunology, Carcinoma, Ductal, Breast genetics, Carcinoma, Ductal, Breast immunology, DNA Copy Number Variations, Datasets as Topic, Female, Gene Expression Profiling, Humans, Oligonucleotide Array Sequence Analysis, Proteogenomics methods, Proteome genetics, Proteome immunology, RNA, Messenger metabolism, Breast Neoplasms pathology, Carcinoma, Ductal, Breast pathology, Protein Interaction Maps, Proteome metabolism
- Abstract
In the preceding decades, molecular characterization has revolutionized breast cancer (BC) research and therapeutic approaches. Presented herein, an unbiased analysis of breast tumor proteomes, inclusive of 9995 proteins quantified across all tumors, for the first time recapitulates BC subtypes. Additionally, poor-prognosis basal-like and luminal B tumors are further subdivided by immune component infiltration, suggesting the current classification is incomplete. Proteome-based networks distinguish functional protein modules for breast tumor groups, with co-expression of EGFR and MET marking ductal carcinoma in situ regions of normal-like tumors and lending to a more accurate classification of this poorly defined subtype. Genes included within prognostic mRNA panels have significantly higher than average mRNA-protein correlations, and gene copy number alterations are dampened at the protein-level; underscoring the value of proteome quantification for prognostication and phenotypic classification. Furthermore, protein products mapping to non-coding genomic regions are identified; highlighting a potential new class of tumor-specific immunotherapeutic targets.
- Published
- 2019
- Full Text
- View/download PDF
34. Microvessel density in breast cancer: the impact of field area on prognostic informativeness.
- Author
-
Kraby MR, Opdahl S, Russnes HG, and Bofin AM
- Subjects
- Aged, Breast Neoplasms chemistry, Breast Neoplasms pathology, Female, Humans, Immunohistochemistry, Ki-67 Antigen analysis, Microvessels chemistry, Middle Aged, Predictive Value of Tests, Prognosis, Registries, von Willebrand Factor analysis, Breast Neoplasms blood supply, Microvessels pathology, Neovascularization, Pathologic
- Abstract
Aims: Tumour microvessel density (MVD) is assessed by counting vessels in the most vascularised tumour region, the vascular hot spot. Current uncertainty regarding the prognostic role of MVD in breast cancer could, in part, be explained by variations in field area size for MVD assessment. We aimed to identify the field area size that provides the most accurate prognostic information in breast carcinoma., Methods: MVD was assessed in 212 tumours. von Willebrand factor positively stained vessels were counted in 10 consecutive visual fields in vascular hotspots. The 10 visual fields in the original counting sequence (MVD-Consecutive) were sorted from highest to lowest vessel count (MVD-Decreasing), and randomly (MVD-Random). After adding counts from one visual field at a time, mean MVD was calculated for each cumulative field area. The prognostic informativeness of each field area and sorting strategy were compared., Results: Median MVD decreased with increasing field size for MVD-Decreasing and MVD-Consecutive. A 0.35 mm
2 total field area comprising only the highest vessel counts provided the most accurate prognostic information (MVD-Decreasing, HR for breast cancer death 1.06 per 10 vessels/mm2 increase, 95% CI 1.03 to 1.10). MVD-Decreasing gave more accurate prognostic information than MVD-Consecutive and MVD-Random, with decreasing prognostic informativeness with increasing field area., Conclusions: Median MVD and its prognostic informativeness decreased with increasing field area. Assessing MVD in a carefully selected small field area of 0.35 mm2 provides the most accurate prognostic information. This could facilitate the implementation of MVD assessment in breast cancer., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2019. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2019
- Full Text
- View/download PDF
35. Author Correction: Pan-cancer analysis of homozygous deletions in primary tumours uncovers rare tumour suppressors.
- Author
-
Cheng J, Demeulemeester J, Wedge DC, Vollan HKM, Pitt JJ, Russnes HG, Pandey BP, Nilsen G, Nord S, Bignell GR, White KP, Børresen-Dale AL, Campbell PJ, Kristensen VN, Stratton MR, Lingjærde OC, Moreau Y, and Van Loo P
- Abstract
The original version of this Article omitted a declaration from the competing interests statement, which should have included the following: 'K.P.W. is President of Tempus Lab, Inc., Chicago, IL, USA'. This has now been corrected in both the PDF and HTML versions of the Article.
- Published
- 2019
- Full Text
- View/download PDF
36. Author Correction: Pan-cancer analysis of homozygous deletions in primary tumours uncovers rare tumour suppressors.
- Author
-
Cheng J, Demeulemeester J, Wedge DC, Vollan HKM, Pitt JJ, Russnes HG, Pandey BP, Nilsen G, Nord S, Bignell GR, White KP, Børresen-Dale AL, Campbell PJ, Kristensen VN, Stratton MR, Lingjærde OC, Moreau Y, and Van Loo P
- Abstract
The original version of this Article contained an error in the author affiliations. The affiliation of Kevin P. White with Tempus Labs, Inc., Chicago, IL, USA was inadvertently omitted.This has now been corrected in both the PDF and HTML versions of the Article.
- Published
- 2018
- Full Text
- View/download PDF
37. Mutational mechanisms of amplifications revealed by analysis of clustered rearrangements in breast cancers.
- Author
-
Głodzik D, Purdie C, Rye IH, Simpson PT, Staaf J, Span PN, Russnes HG, and Nik-Zainal S
- Subjects
- Adult, Age Distribution, Aged, Aged, 80 and over, Algorithms, Breast pathology, Breast Neoplasms pathology, Carrier Proteins genetics, Chromosomes, Human, Pair 11 genetics, Chromosomes, Human, Pair 8 genetics, Cyclin D1 genetics, Datasets as Topic, Female, Genomics methods, Humans, Middle Aged, Oncogenes genetics, Receptor, Fibroblast Growth Factor, Type 1 genetics, Whole Genome Sequencing, Breast Neoplasms genetics, Gene Amplification, Genetic Loci genetics, Translocation, Genetic
- Abstract
Background: Complex clusters of rearrangements are a challenge in interpretation of cancer genomes. Some clusters of rearrangements demarcate clear amplifications of driver oncogenes but others are less well understood. A detailed analysis of rearrangements within these complex clusters could reveal new insights into selection and underlying mutational mechanisms., Patients and Methods: Here, we systematically investigate rearrangements that are densely clustered in individual tumours in a cohort of 560 breast cancers. Applying an agnostic approach, we identify 21 hotspots where clustered rearrangements recur across cancers., Results: Some hotspots coincide with known oncogene loci including CCND1, ERBB2, ZNF217, chr8:ZNF703/FGFR1, IGF1R, and MYC. Others contain cancer genes not typically associated with breast cancer: MCL1, PTP4A1, and MYB. Intriguingly, we identify clustered rearrangements that physically connect distant hotspots. In particular, we observe simultaneous amplification of chr8:ZNF703/FGFR1 and chr11:CCND1 where deep analysis reveals that a chr8-chr11 translocation is likely to be an early, critical, initiating event., Conclusions: We present an overview of complex rearrangements in breast cancer, highlighting a potential new way for detecting drivers and revealing novel mechanistic insights into the formation of two common amplicons.
- Published
- 2018
- Full Text
- View/download PDF
38. Intratumor heterogeneity defines treatment-resistant HER2+ breast tumors.
- Author
-
Rye IH, Trinh A, Saetersdal AB, Nebdal D, Lingjaerde OC, Almendro V, Polyak K, Børresen-Dale AL, Helland Å, Markowetz F, and Russnes HG
- Subjects
- Aged, Disease-Free Survival, Female, Humans, Middle Aged, Survival Rate, Breast Neoplasms genetics, Breast Neoplasms metabolism, Breast Neoplasms mortality, Breast Neoplasms therapy, Drug Resistance, Neoplasm, Gene Dosage, Neoadjuvant Therapy, Receptor, ErbB-2 biosynthesis, Receptor, ErbB-2 genetics
- Abstract
Targeted therapy for patients with HER2-positive (HER2+) breast cancer has improved overall survival, but many patients still suffer relapse and death from the disease. Intratumor heterogeneity of both estrogen receptor (ER) and HER2 expression has been proposed to play a key role in treatment failure, but little work has been done to comprehensively study this heterogeneity at the single-cell level. In this study, we explored the clinical impact of intratumor heterogeneity of ER protein expression, HER2 protein expression, and HER2 gene copy number alterations. Using combined immunofluorescence and in situ hybridization on tissue sections followed by a validated computational approach, we analyzed more than 13 000 single tumor cells across 37 HER2+ breast tumors. The samples were taken both before and after neoadjuvant chemotherapy plus HER2-targeted treatment, enabling us to study tumor evolution as well. We found that intratumor heterogeneity for HER2 copy number varied substantially between patient samples. Highly heterogeneous tumors were associated with significantly shorter disease-free survival and fewer long-term survivors. Patients for which HER2 characteristics did not change during treatment had a significantly worse outcome. This work shows the impact of intratumor heterogeneity in molecular diagnostics for treatment selection in HER2+ breast cancer patients and the power of computational scoring methods to evaluate in situ molecular markers in tissue biopsies., (© 2018 The Authors. Published by FEBS Press and John Wiley & Sons Ltd.)
- Published
- 2018
- Full Text
- View/download PDF
39. Number of Risky Lifestyle Behaviors and Breast Cancer Risk.
- Author
-
Ellingjord-Dale M, Vos L, Vik Hjerkind K, Hjartåker A, Russnes HG, Tretli S, Hofvind S, Dos-Santos-Silva I, and Ursin G
- Abstract
Background: Lifestyle factors are associated with overall breast cancer risk, but less is known about their associations, alone or jointly, with risk of specific breast cancer subtypes., Methods: We conducted a case-control subjects study nested within a cohort of women who participated in the Norwegian Breast Cancer Screening Program during 2006-2014 to examine associations between risky lifestyle factors and breast cancer risk. In all, 4402 breast cancer cases subjects with information on risk factors and hormone receptor status were identified. Conditional logistic regression was used to estimate odds ratios (ORs), with 95% confidence intervals (CIs), in relation to five risky lifestyle factors: body mass index (BMI) of 25 kg/m² or greater, three or more glasses of alcoholic beverages per week, ever smoking, fewer than four hours of physical activity per week, and ever use of menopausal hormone therapy. Analyses were adjusted for education, age at menarche, number of pregnancies, and menopausal status. All statistical tests were two-sided., Results: Compared with women with no risky lifestyle behaviors, those with five had 85% (OR = 1.85, 95% CI = 1.42 to 2.42, P
trend < .0001) increased risk of breast cancer overall. This association was limited to luminal A-like (OR = 2.20, 95% CI = 1.55 to 3.12, Ptrend < .0001) and luminal B-like human epidermal growth factor receptor 2 (HER2)-positive (OR = 1.66, 95% CI = 0.61 to 4.54, Ptrend < .004) subtypes. Number of risky lifestyle factors was not associated with increased risk of luminal B-like HER2-negative, HER2-positive, or triple-negative subtypes ( Ptrend > .18 for all)., Conclusions: Number of risky lifestyle factors was positively associated with increased risk for luminal A-like and luminal B-like HER2-positive breast cancer.- Published
- 2018
- Full Text
- View/download PDF
40. AKT1 low Quiescent Cancer Cells Promote Solid Tumor Growth.
- Author
-
Alves CP, Dey-Guha I, Kabraji S, Yeh AC, Talele NP, Solé X, Chowdhury J, Mino-Kenudson M, Loda M, Sgroi D, Borresen-Dale AL, Russnes HG, Ross KN, and Ramaswamy S
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation physiology, Cell Transformation, Neoplastic, Female, HCT116 Cells, Heterografts, Humans, MCF-7 Cells, Mice, Neoplasms pathology, Neoplasms enzymology, Proto-Oncogene Proteins c-akt metabolism
- Abstract
Human tumor growth depends on rapidly dividing cancer cells driving population expansion. Even advanced tumors, however, contain slowly proliferating cancer cells for reasons that remain unclear. Here, we selectively disrupt the ability of rapidly proliferating cancer cells to spawn AKT1
low daughter cells that are rare, slowly proliferating, tumor-initiating, and chemotherapy-resistant, using β1-integrin activation and the AKT1-E17K-mutant oncoprotein as experimental tools in vivo Surprisingly, we find that selective depletion of AKT1low slow proliferators actually reduces the growth of a molecularly diverse panel of human cancer cell xenograft models without globally altering cell proliferation or survival in vivo Moreover, we find that unusual cancer patients with AKT1-E17K-mutant solid tumors also fail to produce AKT1low quiescent cancer cells and that this correlates with significantly prolonged survival after adjuvant treatment compared with other patients. These findings support a model whereby human solid tumor growth depends on not only rapidly proliferating cancer cells but also on the continuous production of AKT1low slow proliferators. Mol Cancer Ther; 17(1); 254-63. ©2017 AACR ., (©2017 American Association for Cancer Research.)- Published
- 2018
- Full Text
- View/download PDF
41. Alcohol, Physical Activity, Smoking, and Breast Cancer Subtypes in a Large, Nested Case-Control Study from the Norwegian Breast Cancer Screening Program.
- Author
-
Ellingjord-Dale M, Vos L, Hjerkind KV, Hjartåker A, Russnes HG, Tretli S, Hofvind S, Dos-Santos-Silva I, and Ursin G
- Subjects
- Aged, Alcohol Drinking adverse effects, Breast Neoplasms diagnostic imaging, Breast Neoplasms pathology, Case-Control Studies, Early Detection of Cancer statistics & numerical data, Female, Humans, Mammography, Mass Screening statistics & numerical data, Middle Aged, Norway epidemiology, Receptor, ErbB-2 metabolism, Receptors, Estrogen metabolism, Receptors, Progesterone metabolism, Risk Factors, Smoking adverse effects, Alcohol Drinking epidemiology, Breast Neoplasms epidemiology, Exercise, Registries statistics & numerical data, Smoking epidemiology
- Abstract
Background: To what extent alcohol, smoking, and physical activity are associated with the various subtypes of breast cancer is not clear. We took advantage of a large population-based screening cohort to determine whether these risk factors also increase the risk of the poor prognosis subtypes. Methods: We conducted a matched case-control study nested within the Norwegian Breast Cancer Screening Program during 2006-2014. A total of 4,402 breast cancer cases with risk factor and receptor data were identified. Five controls were matched to each case on year of birth and year of screening. Conditional logistic regression was used to estimate ORs of breast cancer subtypes adjusted for potential confounders. Results: There were 2,761 luminal A-like, 709 luminal B-like HER2-negative, 367 luminal B-like HER2-positive, 204 HER2-positive, and 361 triple-negative cancers. Current alcohol consumption was associated with breast cancer risk overall [OR 1.26; 95% confidence interval (CI), 1.09-1.45] comparing 6+ glasses a week to never drinkers. However, this risk increase was found only for luminal A-like breast cancer. Smoking 20+ cigarettes a day was associated with an OR of 1.41 (95% CI, 1.06-1.89) overall, with significant trends for luminal A-like and luminal B-like HER2-negative cancer. Current physical activity (4+ hours/week compared with none) was associated with 15% decreased risk of luminal A-like cancer, but not clearly with other subtypes. Conclusions: In this large study, alcohol, smoking, and physical activity were predominantly associated with luminal A-like breast cancer. Impact: Alcohol, smoking, and physical activity were associated with luminal A-like breast cancer subtype. Cancer Epidemiol Biomarkers Prev; 26(12); 1736-44. ©2017 AACR ., (©2017 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
42. Prognostic value of PAM50 and risk of recurrence score in patients with early-stage breast cancer with long-term follow-up.
- Author
-
Ohnstad HO, Borgen E, Falk RS, Lien TG, Aaserud M, Sveli MAT, Kyte JA, Kristensen VN, Geitvik GA, Schlichting E, Wist EA, Sørlie T, Russnes HG, and Naume B
- Subjects
- Adult, Aged, Breast Neoplasms metabolism, Breast Neoplasms pathology, Female, Follow-Up Studies, Humans, Kaplan-Meier Estimate, Middle Aged, Neoplasm Grading, Neoplasm Metastasis, Neoplasm Recurrence, Local, Neoplasm Staging methods, Patient Outcome Assessment, Prognosis, Risk Assessment, Biomarkers, Tumor, Breast Neoplasms diagnosis, Breast Neoplasms mortality
- Abstract
Background: The aim of this study was to investigate the prognostic value of the PAM50 intrinsic subtypes and risk of recurrence (ROR) score in patients with early breast cancer and long-term follow-up. A special focus was placed on hormone receptor-positive/human epidermal growth factor receptor 2-negative (HR+/HER2-) pN0 patients not treated with chemotherapy., Methods: Patients with early breast cancer (n = 653) enrolled in the observational Oslo1 study (1995-1998) were followed for distant recurrence and breast cancer death. Clinicopathological parameters were collected from hospital records. The primary tumors were analyzed using the Prosigna® PAM50 assay to determine the prognostic value of the intrinsic subtypes and ROR score in comparison with pathological characteristics. The primary endpoints were distant disease-free survival (DDFS) and breast cancer-specific survival (BCSS)., Results: Of 653 tumors, 52.2% were classified as luminal A, 26.5% as luminal B, 10.6% as HER2-enriched, and 10.7% as basal-like. Among the HR+/HER2- patients (n = 476), 37.8% were categorized as low risk by ROR score, 22.7% as intermediate risk, and 39.5% as high risk. Median follow-up durations for BCSS and DDFS were 16.6 and 7.1 years, respectively. Multivariate analysis showed that intrinsic subtypes (all patients) and ROR risk classification (HR+/HER2- patients) yielded strong prognostic information. Among the HR+/HER2- pN0 patients with no adjuvant treatment (n = 231), 53.7% of patients had a low ROR, and their prognosis at 15 years was excellent (15-year BCSS 96.3%). Patients with intermediate risk had reduced survival compared with those with low risk (p = 0.005). In contrast, no difference in survival between the low- and intermediate-risk groups was seen for HR+/HER2- pN0 patients who received tamoxifen only. Ki-67 protein, grade, and ROR score were analyzed in the unselected, untreated pT1pN0 HR+/HER2- population (n = 171). In multivariate analysis, ROR score outperformed both Ki-67 and grade. Furthermore, 55% of patients who according to the PREDICT tool ( http://www.predict.nhs.uk/ ) would be considered chemotherapy candidates were ROR low risk (33%) or luminal A ROR intermediate risk (22%)., Conclusions: The PAM50 intrinsic subtype classification and ROR score improve classification of patients with breast cancer into prognostic groups, allowing for a more precise identification of future recurrence risk and providing an improved basis for adjuvant treatment decisions. Node-negative patients with low ROR scores had an excellent outcome at 15 years even in the absence of adjuvant therapy.
- Published
- 2017
- Full Text
- View/download PDF
43. Pan-cancer analysis of homozygous deletions in primary tumours uncovers rare tumour suppressors.
- Author
-
Cheng J, Demeulemeester J, Wedge DC, Vollan HKM, Pitt JJ, Russnes HG, Pandey BP, Nilsen G, Nord S, Bignell GR, White KP, Børresen-Dale AL, Campbell PJ, Kristensen VN, Stratton MR, Lingjærde OC, Moreau Y, and Van Loo P
- Subjects
- Alleles, Chromosome Fragile Sites genetics, Gene Dosage, Genome, Human, Homozygote, Humans, Ploidies, Telomere metabolism, Gene Deletion, Genes, Tumor Suppressor, Neoplasms genetics
- Abstract
Homozygous deletions are rare in cancers and often target tumour suppressor genes. Here, we build a compendium of 2218 primary tumours across 12 human cancer types and systematically screen for homozygous deletions, aiming to identify rare tumour suppressors. Our analysis defines 96 genomic regions recurrently targeted by homozygous deletions. These recurrent homozygous deletions occur either over tumour suppressors or over fragile sites, regions of increased genomic instability. We construct a statistical model that separates fragile sites from regions showing signatures of positive selection for homozygous deletions and identify candidate tumour suppressors within those regions. We find 16 established tumour suppressors and propose 27 candidate tumour suppressors. Several of these genes (including MGMT, RAD17, and USP44) show prior evidence of a tumour suppressive function. Other candidate tumour suppressors, such as MAFTRR, KIAA1551, and IGF2BP2, are novel. Our study demonstrates how rare tumour suppressors can be identified through copy number meta-analysis.
- Published
- 2017
- Full Text
- View/download PDF
44. Immune Escape in Breast Cancer During In Situ to Invasive Carcinoma Transition.
- Author
-
Gil Del Alcazar CR, Huh SJ, Ekram MB, Trinh A, Liu LL, Beca F, Zi X, Kwak M, Bergholtz H, Su Y, Ding L, Russnes HG, Richardson AL, Babski K, Min Hui Kim E, McDonnell CH 3rd, Wagner J, Rowberry R, Freeman GJ, Dillon D, Sorlie T, Coussens LM, Garber JE, Fan R, Bobolis K, Allred DC, Jeong J, Park SY, Michor F, and Polyak K
- Subjects
- B7-H1 Antigen genetics, Biomarkers, Tumor genetics, Breast Neoplasms genetics, CD3 Complex genetics, Carcinoma, Ductal, Breast genetics, Carcinoma, Intraductal, Noninfiltrating genetics, Disease Progression, Female, Gene Expression Regulation, Neoplastic, Humans, Leukocyte Common Antigens genetics, Receptor, ErbB-2 genetics, Tumor Microenvironment, Breast Neoplasms immunology, Carcinoma, Ductal, Breast immunology, Carcinoma, Intraductal, Noninfiltrating immunology, Gene Expression Profiling methods, T-Lymphocytes immunology
- Abstract
To investigate immune escape during breast tumor progression, we analyzed the composition of leukocytes in normal breast tissues, ductal carcinoma in situ (DCIS), and invasive ductal carcinomas (IDC). We found significant tissue and tumor subtype-specific differences in multiple cell types including T cells and neutrophils. Gene expression profiling of CD45
+ CD3+ T cells demonstrated a decrease in CD8+ signatures in IDCs. Immunofluorescence analysis showed fewer activated GZMB+ CD8+ T cells in IDC than in DCIS, including in matched DCIS and recurrent IDC. T-cell receptor clonotype diversity was significantly higher in DCIS than in IDCs. Immune checkpoint protein TIGIT-expressing T cells were more frequent in DCIS, whereas high PD-L1 expression and amplification of CD274 (encoding PD-L1) was only detected in triple-negative IDCs. Coamplification of a 17q12 chemokine cluster with ERBB2 subdivided HER2+ breast tumors into immunologically and clinically distinct subtypes. Our results show coevolution of cancer cells and the immune microenvironment during tumor progression. Significance: The design of effective cancer immunotherapies requires the understanding of mechanisms underlying immune escape during tumor progression. Here we demonstrate a switch to a less active tumor immune environment during the in situ to invasive breast carcinoma transition, and identify immune regulators and genomic alterations that shape tumor evolution. Cancer Discov; 7(10); 1098-115. ©2017 AACR. See related commentary by Speiser and Verdeil, p. 1062 This article is highlighted in the In This Issue feature, p. 1047 ., (©2017 American Association for Cancer Research.)- Published
- 2017
- Full Text
- View/download PDF
45. Breast Cancer Molecular Stratification: From Intrinsic Subtypes to Integrative Clusters.
- Author
-
Russnes HG, Lingjærde OC, Børresen-Dale AL, and Caldas C
- Subjects
- Breast Neoplasms genetics, DNA Copy Number Variations genetics, Female, Gene Dosage, Gene Expression Regulation, Neoplastic, Humans, Multilevel Analysis, Mutation genetics, Breast Neoplasms classification, Breast Neoplasms pathology
- Abstract
Breast carcinomas can be stratified into different entities based on clinical behavior, histologic features, and/or by biological properties. A classification of breast cancer should be based on underlying biology, which we know must be determined by the somatic genomic landscape of mutations. Moreover, because the latest generations of anticancer agents are founded on biological mechanisms, a detailed molecular stratification is a requirement for appropriate clinical management. Such stratification, based on genomic drivers, will be important for selecting patients for clinical trials. It will also facilitate the discovery of novel drivers, the study of tumor evolution, and the identification of mechanisms of treatment resistance. Assays for risk stratification have focused mainly on response prediction to existing treatment regimens. Molecular stratification based on gene expression profiling revealed that breast cancers could be classified in so-called intrinsic subtypes (luminal A and B, HER2-enriched, basal-like, and normal-like), which mostly corresponded to hormone receptor and HER2 status, and further stratified luminal tumors based on proliferation. The realization that a significant proportion of the gene expression landscape is determined by the somatic copy number alterations that drive expression in cis led to the newer classification of breast cancers into integrative clusters. This stratification of breast cancers into integrative clusters reveals prototypical patterns of single-nucleotide variants and is associated with distinct clinical courses and response to therapy., (Copyright © 2017 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
46. Implementing precision cancer medicine in the public health services of Norway: the diagnostic infrastructure and a cost estimate.
- Author
-
Ree AH, Russnes HG, Heinrich D, Dueland S, Boye K, Nygaard V, Silwal-Pandit L, Østrup O, Hovig E, Nygaard V, Rødland EA, Nakken S, Øien JT, Johansen C, Bergheim IR, Skarpeteig V, Sathermugathevan M, Sauer T, Lund-Iversen M, Beiske K, Nasser S, Julsrud L, Reisse CH, Ruud EA, Flørenes VA, Hagene KT, Aas E, Lurås H, Johnsen-Soriano S, Geitvik GA, Lingjærde OC, Børresen-Dale AL, Mælandsmo GM, and Flatmark K
- Abstract
Objective: Through the conduct of an individual-based intervention study, the main purpose of this project was to build and evaluate the required infrastructure that may enable routine practice of precision cancer medicine in the public health services of Norway, including modelling of costs., Methods: An eligible patient had end-stage metastatic disease from a solid tumour. Metastatic tissue was analysed by DNA sequencing, using a 50-gene panel and a study-generated pipeline for analysis of sequence data, supplemented with fluorescence in situ hybridisation to cover relevant biomarkers. Cost estimations compared best supportive care, biomarker-agnostic treatment with a molecularly targeted agent and biomarker-based treatment with such a drug. These included costs for medication, outpatient clinic visits, admission from adverse events and the biomarker-based procedures., Results: The diagnostic procedures, which comprised sampling of metastatic tissue, mutation analysis and data interpretation at the Molecular Tumor Board before integration with clinical data at the Clinical Tumor Board, were completed in median 18 (8-39) days for the 22 study patients. The 23 invasive procedures (12 from liver, 6 from lung, 5 from other sites) caused a single adverse event (pneumothorax). Per patient, 0-5 mutations were detected in metastatic tumours; however, no actionable target case was identified for the current single-agent therapy approach. Based on the cost modelling, the biomarker-based approach was 2.5-fold more costly than best supportive care and 2.5-fold less costly than the biomarker-agnostic option., Conclusions: The first project phase established a comprehensive diagnostic infrastructure for precision cancer medicine, which enabled expedite and safe mutation profiling of metastatic tumours and data interpretation at multidisciplinary tumour boards for patients with end-stage cancer. Furthermore, it prepared for protocol amendments, recently approved by the designated authorities for the second study phase, allowing more comprehensive mutation analysis and opportunities to define therapy targets., Competing Interests: Competing interests: None declared.
- Published
- 2017
- Full Text
- View/download PDF
47. Quantitative multigene FISH on breast carcinomas identifies der(1;16)(q10;p10) as an early event in luminal A tumors.
- Author
-
Rye IH, Lundin P, Månér S, Fjelldal R, Naume B, Wigler M, Hicks J, Børresen-Dale AL, Zetterberg A, and Russnes HG
- Subjects
- Chromosome Breakage, Chromosomes, Human, Pair 1, Chromosomes, Human, Pair 16, Female, Humans, Breast Neoplasms genetics, In Situ Hybridization, Fluorescence methods, Translocation, Genetic
- Abstract
In situ detection of genomic alterations in cancer provides information at the single cell level, making it possible to investigate genomic changes in cells in a tissue context. Such topological information is important when studying intratumor heterogeneity as well as alterations related to different steps in tumor progression. We developed a quantitative multigene fluorescence in situ hybridization (QM FISH) method to detect multiple genomic regions in single cells in complex tissues. As a "proof of principle" we applied the method to breast cancer samples to identify partners in whole arm (WA) translocations. WA gain of chromosome arm 1q and loss of chromosome arm 16q are among the most frequent genomic events in breast cancer. By designing five specific FISH probes based on breakpoint information from comparative genomic hybridization array (aCGH) profiles, we visualized chromosomal translocations in clinical samples at the single cell level. By analyzing aCGH data from 295 patients with breast carcinoma with known molecular subtype, we found concurrent WA gain of 1q and loss of 16q to be more frequent in luminal A tumors compared to other molecular subtypes. QM FISH applied to a subset of samples (n = 26) identified a derivative chromosome der(1;16)(q10;p10), a result of a centromere-close translocation between chromosome arms 1q and 16p. In addition, we observed that the distribution of cells with the translocation varied from sample to sample, some had a homogenous cell population while others displayed intratumor heterogeneity with cell-to-cell variation. Finally, for one tumor with both preinvasive and invasive components, the fraction of cells with translocation was lower and more heterogeneous in the preinvasive tumor cells compared to the cells in the invasive component., (© 2014 The Authors Genes, Chromosomes & Cancer Published by Wiley Periodicals, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
48. Glycan-related gene expression signatures in breast cancer subtypes; relation to survival.
- Author
-
Potapenko IO, Lüders T, Russnes HG, Helland Å, Sørlie T, Kristensen VN, Nord S, Lingjærde OC, Børresen-Dale AL, and Haakensen VD
- Subjects
- Carcinoma, Intraductal, Noninfiltrating genetics, Cluster Analysis, Female, Glycosylation, Humans, Polysaccharides metabolism, Survival Analysis, Breast Neoplasms classification, Breast Neoplasms genetics, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Polysaccharides genetics
- Abstract
Alterations in glycan structures are early signs of malignancy and have recently been proposed to be in part a driving force behind malignant transformation. Here, we explore whether differences in expression of genes related to the process of glycosylation exist between breast carcinoma subtypes - and look for their association to clinical parameters. Five expression datasets of 454 invasive breast carcinomas, 31 ductal carcinomas in situ (DCIS), and 79 non-malignant breast tissue samples were analysed. Results were validated in 1960 breast carcinomas. 419 genes encoding glycosylation-related proteins were selected. The DCIS samples appeared expression-wise similar to carcinomas, showing altered gene expression related to glycosaminoglycans (GAGs) and N-glycans when compared to non-malignant samples. In-situ lesions with different aggressiveness potentials demonstrated changes in glycosaminoglycan sulfation and adhesion proteins. Subtype-specific expression patterns revealed down-regulation of genes encoding glycan-binding proteins in the luminal A and B subtypes. Clustering basal-like samples using a consensus list of genes differentially expressed across discovery datasets produced two clusters with significantly differing prognosis in the validation dataset. Finally, our analyses suggest that glycolipids may play an important role in carcinogenesis of breast tumors - as demonstrated by association of B3GNT5 and UGCG genes to patient survival. In conclusion, most glycan-specific changes occur early in the carcinogenic process. We have identified glycan-related alterations specific to breast cancer subtypes including a prognostic signature for two basal-like subgroups. Future research in this area may potentially lead to markers for better prognostication and treatment stratification of breast cancer patients., (Copyright © 2015 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
49. eEF2K--a new target in breast cancers with combined inactivation of p53 and PTEN.
- Author
-
Russnes HG and Caldas C
- Subjects
- Animals, Female, Humans, Elongation Factor 2 Kinase metabolism, Gene Deletion, PTEN Phosphohydrolase genetics, Triple Negative Breast Neoplasms enzymology, Tumor Suppressor Protein p53 genetics
- Abstract
Extensive efforts have now characterized the somatic molecular alterations in human breast cancer (Cancer Genome Atlas Network, 2012; Stephens et al, 2012) and have led to a re-definition of the disease as a constellation of 10 distinct driver-based subtypes (IntClust subtypes) (Curtis et al, 2012). The pursuit of druggable targets for each of these subtypes is now pressing. This is elegantly illustrated by the work of Liu et al (2014).
- Published
- 2014
- Full Text
- View/download PDF
50. GoIFISH: a system for the quantification of single cell heterogeneity from IFISH images.
- Author
-
Trinh A, Rye IH, Almendro V, Helland A, Russnes HG, and Markowetz F
- Subjects
- Breast Neoplasms genetics, Female, Fluorescent Antibody Technique methods, Genetic Heterogeneity, Humans, In Situ Hybridization, Fluorescence methods, Image Processing, Computer-Assisted methods, Single-Cell Analysis methods, Software
- Abstract
Molecular analysis has revealed extensive intra-tumor heterogeneity in human cancer samples, but cannot identify cell-to-cell variations within the tissue microenvironment. In contrast, in situ analysis can identify genetic aberrations in phenotypically defined cell subpopulations while preserving tissue-context specificity. GoIFISHGoIFISH is a widely applicable, user-friendly system tailored for the objective and semi-automated visualization, detection and quantification of genomic alterations and protein expression obtained from fluorescence in situ analysis. In a sample set of HER2-positive breast cancers GoIFISHGoIFISH is highly robust in visual analysis and its accuracy compares favorably to other leading image analysis methods. GoIFISHGoIFISH is freely available at www.sourceforge.net/projects/goifish/.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.