Bioinformatics for cancer research

cancer_bioinformatics.png

Understand cancer, develop new drugs and personalize treatment with Genevia Technologies.

Our history in cancer bioinformatics is long: we have had the privilege to work with biologists seeking for a deeper understanding of cancer, companies developing new treatments to cancer, as well as oncologists wishing to optimize therapies to individual patients.

Below you will find highlights of our journey in cancer research. If you feel inspired and would like to benefit from our expertise, leave us a message and we will book you a call with our bioinformatician.

Leave us a short description of your bioinformatics needs and we will be in touch very soon!

Understanding cancer

What causes cancer? Which alterations in DNA, pathways and metabolic processes allow a tumor to grow, spread and evade treatment? How does tumorigenic reprogramming relate to normal cellular differentiation?

Our experience in cancer biology covers research into fundamental questions across cancer types and high-throughput molecular data types. Together with our collaborators and customers, we have studied heritable and somatic variants and their downstream molecular effects as well as the evolution and microenvironment of tumors, to name but a few aspects of cancer biology.

Whether you are setting out to characterize an understudied malignancy or dive deep into the molecular biology of a more common cancer, we have you covered, bioinformatically speaking.

Learn more

References and case studies

Selected publications from our customers

  • Chang, Y. T. et al. (2024). MHC-I upregulation safeguards neoplastic T cells in the skin against NK cell-mediated eradication in mycosis fungoides. Nature communications, 15(1), 752. https://doi.org/10.1038/s41467-024-45083-8
  • Häyrinen, M. et al. (2024). Tumor RNA sequencing identifies a group of Mycosis Fungoides patients with failure of skin-directed therapies. The Journal of investigative dermatology, S0022-202X(24)02084-0. Advance online publication. https://doi.org/10.1016/j.jid.2024.06.1292
  • Boyd, S., et al. (2024). NGS of brush cytology samples improves the detection of high-grade dysplasia and cholangiocarcinoma in patients with primary sclerosing cholangitis: A retrospective and prospective study. Hepatology communications, 8(4), e0415. https://doi.org/10.1097/HC9.0000000000000415
  • Chérouvrier Hansson, V. et al. (2024). Dichotomous Effects of Glypican-4 on Cancer Progression and Its Crosstalk with Oncogenes. International journal of molecular sciences, 25(7), 3945. https://doi.org/10.3390/ijms25073945
  • Häyrinen, M. J. et al. (2023). The Transcription Factor Twist1 Has a Significant Role in Mycosis Fungoides (MF) Cell Biology: An RNA Sequencing Study of 40 MF Cases. Cancers, 15(5), 1527. https://doi.org/10.3390/cancers15051527
  • Karihtala, P. et al. (2022). Comparison of the mutational profiles of neuroendocrine breast tumours, invasive ductal carcinomas and pancreatic neuroendocrine carcinomas. Oncogenesis, 11(1), 53. https://doi.org/10.1038/s41389-022-00427-1
  • Song, J. et al. (2022). The ubiquitin-ligase TRAF6 and TGFβ type I receptor form a complex with Aurora kinase B contributing to mitotic progression and cytokinesis in cancer cells. EBioMedicine, 82, 104155. https://doi.org/10.1016/j.ebiom.2022.104155
  • Yuan, O. et al. (2022). A somatic mutation in moesin drives progression into acute myeloid leukemia. Science advances, 8(16), eabm9987. https://doi.org/10.1126/sciadv.abm9987
  • Wahlström, G. et al. (2022). The variant rs77559646 associated with aggressive prostate cancer disrupts ANO7 mRNA splicing and protein expression. Human molecular genetics, ddac012. Advance online publication. https://doi.org/10.1093/hmg/ddac012
  • Tusup, M. et al. (2022). Epitranscriptomics modifier pentostatin indirectly triggers Toll-like receptor 3 and can enhance immune infiltration in tumors. Molecular therapy : the journal of the American Society of Gene Therapy, 30(3), 1163–1170. https://doi.org/10.1016/j.ymthe.2021.09.022
  • Kundu, S. et al. (2021). Common and mutation specific phenotypes of KRAS and BRAF mutations in colorectal cancer cells revealed by integrative -omics analysis. Journal of experimental & clinical cancer research : CR, 40(1), 225. https://doi.org/10.1186/s13046-021-02025-2
  • Tikkanen, T. et al. (2018). Seshat: A Web service for accurate annotation, validation, and analysis of TP53 variants generated by conventional and next-generation sequencing. Human mutation, 39(7), 925–933. https://doi.org/10.1002/humu.23543

Selected publications from our team

  • Kiviaho, A. et al. (2024). Androgen deprivation therapy-resistant club cells are linked to myeloid cell-driven immunosuppression in the prostate tumor microenvironment. bioRxiv 2024.03.25.586330; doi: https://doi.org/10.1101/2024.03.25.586330

  • Obacz, J. et al. (2024). IRE1 endoribonuclease signaling promotes myeloid cell infiltration in glioblastoma. Neuro-oncology, 26(5), 858–871. https://doi.org/10.1093/neuonc/noad256

  • Nurminen, A. et al. (2023). Cancer origin tracing and timing in two high-risk prostate cancers using multisample whole genome analysis: prospects for personalized medicine. Genome medicine, 15(1), 82. https://doi.org/10.1186/s13073-023-01242-y

  • Caronni, N. et al. (2023). IL-1β+ macrophages fuel pathogenic inflammation in pancreatic cancer. Nature, 623(7986), 415–422. https://doi.org/10.1038/s41586-023-06685-2

  • Beumers, L. et al. (2023). Clonal heterogeneity in ER+ breast cancer reveals the proteasome and PKC as potential therapeutic targets. NPJ breast cancer, 9(1), 97. https://doi.org/10.1038/s41523-023-00604-4

  • Ricordel, C. et al. (2023). Genomic characteristics and clinical significance of CD56+ circulating tumor cells in small cell lung cancer. Scientific reports, 13(1), 3626. https://doi.org/10.1038/s41598-023-30536-9

  • Simigdala, N. et al. (2023). Loss of Kmt2c in vivo leads to EMT, mitochondrial dysfunction and improved response to lapatinib in breast cancer. Cellular and molecular life sciences : CMLS, 80(4), 100. https://doi.org/10.1007/s00018-023-04734-7

  • Aakula, A. et al. (2023). RAS and PP2A activities converge on epigenetic gene regulation. Life science alliance, 6(5), e202301928. https://doi.org/10.26508/lsa.202301928

  • Dundr, P. et al. (2023). Uterine leiomyoma with RAD51B::NUDT3 fusion: a report of 2 cases. Virchows Archiv : an international journal of pathology, 10.1007/s00428-023-03603-9. Advance online publication. https://doi.org/10.1007/s00428-023-03603-9

  • Kontogianni, G. et al. (2023). A Comprehensive Analysis of Cutaneous Melanoma Patients in Greece Based on Multi-Omic Data. Cancers, 15(3), 815. https://doi.org/10.3390/cancers15030815

  • Rodriguez-Martinez, A. et al. (2022). Novel ZNF414 activity characterized by integrative analysis of ChIP-exo, ATAC-seq and RNA-seq data. Biochimica et biophysica acta. Gene regulatory mechanisms, 1865(3), 194811. Advance online publication. https://doi.org/10.1016/j.bbagrm.2022.194811

  • Kukkonen, K. et al. (2022). Nonmalignant AR-positive prostate epithelial cells and cancer cells respond differently to androgen. Endocrine-related cancer, 29(12), 717–733. https://doi.org/10.1530/ERC-22-0108

  • Machado-Lopez, A. et al. (2022). Integrative Genomic and Transcriptomic Profiling Reveals a Differential Molecular Signature in Uterine Leiomyoma versus Leiomyosarcoma. International journal of molecular sciences, 23(4), 2190. https://doi.org/10.3390/ijms23042190

  • Gao, Y. et al. (2022). Inactivation of AMPK Leads to Attenuation of Antigen Presentation and Immune Evasion in Lung Adenocarcinoma. Clinical cancer research : an official journal of the American Association for Cancer Research, 28(1), 227–237. https://doi.org/10.1158/1078-0432.CCR-21-2049

  • Andersson, A. et al. (2021). Spatial deconvolution of HER2-positive breast cancer delineates tumor-associated cell type interactions. Nature communications, 12(1), 6012. https://doi.org/10.1038/s41467-021-26271-2

  • Hall, Z. et al. (2021). Lipid Remodeling in Hepatocyte Proliferation and Hepatocellular Carcinoma. Hepatology (Baltimore, Md.), 73(3), 1028–1044. https://doi.org/10.1002/hep.31391

  • Taavitsainen, S. et al. (2021). Single-cell ATAC and RNA sequencing reveal pre-existing and persistent cells associated with prostate cancer relapse. Nature communications, 12(1), 5307. https://doi.org/10.1038/s41467-021-25624-1
  • Filppu, P. et al. (2021). CD109-GP130 interaction drives glioblastoma stem cell plasticity and chemoresistance through STAT3 activity. JCI insight, 6(9), e141486. https://doi.org/10.1172/jci.insight.141486
  • Vlachavas, E. I. et al. (2021). A Detailed Catalogue of Multi-Omics Methodologies for Identification of Putative Biomarkers and Causal Molecular Networks in Translational Cancer Research. International journal of molecular sciences, 22(6), 2822. https://doi.org/10.3390/ijms22062822
  • Woodcock, D. J. et al. (2020). Prostate cancer evolution from multilineage primary to single lineage metastases with implications for liquid biopsy. Nature communications, 11(1), 5070. https://doi.org/10.1038/s41467-020-18843-5

  • Dufva, O. et al. (2020). Immunogenomic Landscape of Hematological Malignancies. Cancer cell, 38(3), 380–399.e13. https://doi.org/10.1016/j.ccell.2020.06.002
  • Mehtonen, J. et al. (2020). Single cell characterization of B-lymphoid differentiation and leukemic cell states during chemotherapy in ETV6-RUNX1-positive pediatric leukemia identifies drug-targetable transcription factor activities. Genome medicine, 12(1), 99. https://doi.org/10.1186/s13073-020-00799-2
  • Yoosuf, N. et al. (2020). Identification and transfer of spatial transcriptomics signatures for cancer diagnosis. Breast cancer research : BCR, 22(1), 6. https://doi.org/10.1186/s13058-019-1242-9
  • Pölönen, P. et al. (2019). Hemap: An Interactive Online Resource for Characterizing Molecular Phenotypes across Hematologic Malignancies. Cancer research, 79(10), 2466–2479. https://doi.org/10.1158/0008-5472.CAN-18-2970
  • Vlachavas, E. I. et al. (2019). Radiogenomic Analysis of F-18-Fluorodeoxyglucose Positron Emission Tomography and Gene Expression Data Elucidates the Epidemiological Complexity of Colorectal Cancer Landscape. Computational and structural biotechnology journal, 17, 177–185. https://doi.org/10.1016/j.csbj.2019.01.007
  • Escobar, G. et al. (2018). Interferon gene therapy reprograms the leukemia microenvironment inducing protective immunity to multiple tumor antigens. Nature communications, 9(1), 2896. https://doi.org/10.1038/s41467-018-05315-0
  • Norelli, M. et al. (2018). Monocyte-derived IL-1 and IL-6 are differentially required for cytokine-release syndrome and neurotoxicity due to CAR T cells. Nature medicine, 24(6), 739–748. https://doi.org/10.1038/s41591-018-0036-4
  • Berglund, E. et al. (2018). Spatial maps of prostate cancer transcriptomes reveal an unexplored landscape of heterogeneity. Nature communications, 9(1), 2419. https://doi.org/10.1038/s41467-018-04724-5
  • de Bock, C. E. et al. (2018). HOXA9 Cooperates with Activated JAK/STAT Signaling to Drive Leukemia Development. Cancer discovery, 8(5), 616–631. https://doi.org/10.1158/2159-8290.CD-17-0583
  • Gao, Q. et al. (2018). Driver Fusions and Their Implications in the Development and Treatment of Human Cancers. Cell reports, 23(1), 227–238.e3. https://doi.org/10.1016/j.celrep.2018.03.050
  • Määttä, K. et al. (2016). Whole-exome sequencing of Finnish hereditary breast cancer families. European journal of human genetics : EJHG, 25(1), 85–93. https://doi.org/10.1038/ejhg.2016.141

cancer_genetics.pngsomatic_variation.pngcancer_molecular_mechanisms.pngtumor_heterogeneity_and_microenvironment.png

tumor_etiology_and_evolution.pnggenevia_technologies_tumor_evolution_analysis.png

Treating cancer

Developing a new cancer therapy is a long, costly and risky process. High-throughput measurements coupled with cutting-edge bioinformatics has a lot to offer along the way to both speed up the process and to increase the chances of success.

We can help in identifying targets for a given disease based on public and proprietary molecular and clinical data. Public data on molecular drug perturbation profiles, on the other hand, enables scanning for new applications for pharmaceuticals that are already on the market.

For both preclinical and clinical research on a new treatment, transcriptomic, epigenomic and proteomic measurements can be used to study the molecular mechanism of action. This allows for further optimizing the treatment and ruling out off-target effects.

Learn more

References and case studies

Selected publications from our team

  • Annala, M. et al. (2021). Cabazitaxel versus abiraterone or enzalutamide in poor prognosis metastatic castration-resistant prostate cancer: a multicentre, randomised, open-label, phase II trial. Annals of oncology : official journal of the European Society for Medical Oncology, 32(7), 896–905. https://doi.org/10.1016/j.annonc.2021.03.205

target_discovery.pngmechanism_of_action.png

Predicting outcomes

Being able to predict the onset and development of cancer enables better treatment through early and accurate diagnosis and personalized treatment.

We use survival analyses and machine learning approaches with clinical and molecular data to predict patient-specific risks. Such analyses result in biomarkers or multi-marker signatures with clinical potential.

Biomarker discovery projects have been some of the most fruitful and clinically promising ones we have participated in, as you may see from the references and publications below!

Learn more

References and case studies

Selected publications from our customers

  • Karihtala, P. et al. (2024). Mutational signatures and their association with cancer survival and gene expression in multiple cancer types. International journal of cancer, 10.1002/ijc.35148. Advance online publication. https://doi.org/10.1002/ijc.35148
  • Nissinen, L. et al. (2024). Clustering of RNA co-expression network identifies novel long non-coding RNA biomarkers in squamous cell carcinoma. Scientific reports, 14(1), 16864. https://doi.org/10.1038/s41598-024-67808-x
  • Karihtala, P. et al. (2023). Mutational signatures and their association with survival and gene expression in urological carcinomas. Neoplasia (New York, N.Y.), 44, 100933. Advance online publication. https://doi.org/10.1016/j.neo.2023.100933
  • Cheng, F. et al. (2023). Attenuation of cancer proliferation by suppression of glypican-1 and its pleiotropic effects in neoplastic behavior. Oncotarget, 14, 219–235. https://doi.org/10.18632/oncotarget.28388
  • Mezheyeuski, A. et al. (2023). An immune score reflecting pro- and anti-tumoural balance of tumour microenvironment has major prognostic impact and predicts immunotherapy response in solid cancers. EBioMedicine, 88, 104452. Advance online publication. https://doi.org/10.1016/j.ebiom.2023.104452
  • Karihtala, P. et al. (2022). Mutational Signatures Associate With Survival in Gastrointestinal Carcinomas. Cancer genomics & proteomics, 19(5), 556–569. https://doi.org/10.21873/cgp.20340
  • Pommergaard, H. C et al. (2022). Aldehyde dehydrogenase expression may be a prognostic biomarker and associated with liver cirrhosis in patients resected for hepatocellular carcinoma. Surgical oncology, 40, 101677. https://doi.org/10.1016/j.suronc.2021.101677
  • Tsakonas, G. et al. (2021). High Density of NRF2 Expression in Malignant Cells Is Associated with Increased Risk of CNS Metastasis in Early-Stage NSCLC. Cancers, 13(13), 3151. https://doi.org/10.3390/cancers13133151
  • Madonna, G. et al. (2021). Clinical Categorization Algorithm (CLICAL) and Machine Learning Approach (SRF-CLICAL) to Predict Clinical Benefit to Immunotherapy in Metastatic Melanoma Patients: Real-World Evidence from the Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy. Cancers, 13(16), 4164. https://doi.org/10.3390/cancers13164164
  • Ness, C. et al. (2021). Integrated differential DNA methylation and gene expression of formalin-fixed paraffin-embedded uveal melanoma specimens identifies genes associated with early metastasis and poor prognosis. Experimental eye research, 203, 108426. https://doi.org/10.1016/j.exer.2020.108426

  • Pommergaard, H. C. et al. (2021). Peroxisome proliferator-activated receptor activity correlates with poor survival in patients resected for hepatocellular carcinoma. Journal of hepato-biliary-pancreatic sciences, 28(4), 327–335. https://doi.org/10.1002/jhbp.745
  • Lehto, T. K. et al. (2021). Transcript analysis of commercial prostate cancer risk stratification panels in hard-to-predict grade group 2-4 prostate cancers. The Prostate, 81(7), 368–376. https://doi.org/10.1002/pros.24108
  • Simao, F. et al. (2021). SRF-CLICAL: an approach for patient risk stratification using random forest models. bioRxiv 2021.06.22.448514; doi: https://doi.org/10.1101/2021.06.22.448514

Selected publications from our team

  • Fonseca, N. M. et al. (2024). Prediction of plasma ctDNA fraction and prognostic implications of liquid biopsy in advanced prostate cancer. Nature communications, 15(1), 1828. https://doi.org/10.1038/s41467-024-45475-w
  • Kallio, H. M. et al. (2024). Sensitive circulating tumor DNA-based residual disease detection in epithelial ovarian cancer. Life science alliance, 7(6), e202402658. https://doi.org/10.26508/lsa.202402658
  • Cao, S., Wang et al. (2022). Estimation of tumor cell total mRNA expression in 15 cancer types predicts disease progression. Nature biotechnology, 10.1038/s41587-022-01342-x. Advance online publication. https://doi.org/10.1038/s41587-022-01342-x
  • Rautajoki, K. J. et al. (2022). PTPRD and CNTNAP2 as markers of tumor aggressiveness in oligodendrogliomas. Scientific reports, 12(1), 14083. https://doi.org/10.1038/s41598-022-14977-2
  • Vandekerkhove, G. et al. (2021). Plasma ctDNA is a tumor tissue surrogate and enables clinical-genomic stratification of metastatic bladder cancer. Nature communications, 12(1), 184. https://doi.org/10.1038/s41467-020-20493-6
  • Taavitsainen, S. et al. (2019). Evaluation of Commercial Circulating Tumor DNA Test in Metastatic Prostate Cancer. JCO precision oncology, 3, PO.19.00014. https://doi.org/10.1200/PO.19.00014
  • Kaikkonen, E. et al. (2018). ANO7 is associated with aggressive prostate cancer. International journal of cancer, 143(10), 2479–2487. https://doi.org/10.1002/ijc.31746

biomarker_discovery.png

Contact us

Leave your email address here with a brief description of your needs, and we will contact you to get things moving forward!

Antti Ylipää
Antti Ylipää CEO, co-founder Genevia Technologies Oy +358 40 747 7672