1
|
Engvall K, Uvdal H, Björn N, Åvall-Lundqvist E, Gréen H. Prediction models of persistent taxane-induced peripheral neuropathy among breast cancer survivors using whole-exome sequencing. NPJ Precis Oncol 2024; 8:102. [PMID: 38755266 PMCID: PMC11099113 DOI: 10.1038/s41698-024-00594-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 05/03/2024] [Indexed: 05/18/2024] Open
Abstract
Persistent taxane-induced peripheral neuropathy (TIPN) is highly prevalent among early-stage breast cancer survivors (ESBCS) and has detrimental effect on quality of life. We leveraged logistic regression models to develop and validate polygenic prediction models to estimate the risk of persistent PN symptoms in a training cohort and validation cohort taking clinical risk factors into account. Based on 337 whole-exome sequenced ESBCS two of five prediction models for individual PN symptoms obtained AUC results above 60% when validated. Using the model for numbness in feet (35 SNVs) in the test cohort, 73% survivors were correctly predicted. For tingling in feet (55 SNVs) 70% were correctly predicted. Both models included SNVs from the ADAMTS20, APT6V0A2, CCDC88C, CYP2C8, EPHA5, NR1H3, PSKH2/APTV0D2, and SCN10A genes. For cramps in feet, difficulty climbing stairs and difficulty opening a jar the validation was unsuccessful. Polygenic prediction models including clinical risk factors can estimate the risk of persistent taxane-induced numbness in feet and tingling in feet in ESBCS.
Collapse
Affiliation(s)
- Kristina Engvall
- Department of Oncology, Jönköping, Region Jönköping County, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.
| | - Hanna Uvdal
- Division of Clinical Chemistry and Pharmacology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Niclas Björn
- Division of Clinical Chemistry and Pharmacology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Elisabeth Åvall-Lundqvist
- Department of Oncology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Henrik Gréen
- Division of Clinical Chemistry and Pharmacology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Department of Forensic Genetics and Forensic Toxicology, National Board of Forensic Medicine, Linköping, Sweden
| |
Collapse
|
2
|
Björn N, Jakobsen I, Udagawa C, Brandén E, Koyi H, Lewensohn R, De Petris L, Zembutsu H, Gréen H. The association of four genetic variants with myelosuppression in gemcitabine-treated Japanese is not evident in gemcitabine/carboplatin-treated Swedes. Basic Clin Pharmacol Toxicol 2022; 130:513-521. [PMID: 35132780 PMCID: PMC9303231 DOI: 10.1111/bcpt.13712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 11/26/2022]
Abstract
Gemcitabine/carboplatin‐induced myelosuppressive adverse drug reactions (ADRs) are clinical problems leading to patient suffering and dose alterations. There is a need for personalised medicine to improve treatment effects and patients' well‐being. We tested four genetic variants, rs11141915, rs1901440, rs12046844 and rs11719165, previously suggested as potential biomarkers for gemcitabine‐induced leukopenia/neutropenia in Japanese patients, in 213 Swedish gemcitabine/carboplatin‐treated non‐small cell lung cancer (NSCLC) patients. DNA was genotyped using TaqMan probes and real‐time PCR. The relationships between the risk alleles and low toxicity (non‐ADR: Common Terminology Criteria for Adverse Events [CTCAE] grades 0) or high toxicity (ADR: CTCAE grades 3–4) of platelets, leukocytes and neutrophils were evaluated using Fisher's exact test. The risk alleles did not correlate with myelosuppression, and the strongest borderline significance (not withstanding adjustment for multiple testing) was for rs1901440 (neutropenia, p = 0.043) and rs11719165 (leukopenia, p = 0.049) where the risk alleles trended towards lower toxicity, contrasting with previous study findings. Risk alleles and higher risk scores were more common among our patients. We conclude that the genetic variants do not apply to Swedish patients treated with gemcitabine/carboplatin. However, they can still be important in other populations and cohorts, especially in a gemcitabine monotherapy setting, where the causal genetic variation might influence myelosuppressive ADRs.
Collapse
Affiliation(s)
- Niclas Björn
- Division of Clinical Chemistry and Pharmacology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Ingrid Jakobsen
- Division of Clinical Chemistry and Pharmacology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.,Department of Laboratory Medicine, Örebro University Hospital, Örebro, Sweden
| | - Chihiro Udagawa
- Department of Genetic Medicine and Services, National Cancer Center Hospital, Tokyo, Japan
| | - Eva Brandén
- Department of Respiratory Medicine, Gävle Hospital, Gävle, Sweden.,Centre for Research and Development, Uppsala University/Region Gävleborg, Gävle, Sweden
| | - Hirsh Koyi
- Department of Respiratory Medicine, Gävle Hospital, Gävle, Sweden.,Centre for Research and Development, Uppsala University/Region Gävleborg, Gävle, Sweden
| | - Rolf Lewensohn
- Thoracic Oncology Unit, Tema Cancer, Karolinska University Hospital, and Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Luigi De Petris
- Thoracic Oncology Unit, Tema Cancer, Karolinska University Hospital, and Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Hitoshi Zembutsu
- Department of Clinical Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Henrik Gréen
- Division of Clinical Chemistry and Pharmacology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.,Department of Forensic Genetics and Forensic Toxicology, National Board of Forensic Medicine, Linköping, Sweden
| |
Collapse
|
3
|
Scheurer ME, Zhou R, Gilbert MR, Bondy ML, Sulman EP, Yuan Y, Liu Y, Vera E, Wendland MM, Youssef EF, Stieber VW, Komaki RR, Flickinger JC, Kenyon LC, Robins HI, Hunter GK, Crocker IR, Chao ST, Pugh SL, Armstrong TS. Germline polymorphisms in MGMT associated with temozolomide-related myelotoxicity risk in patients with glioblastoma treated on NRG Oncology/RTOG 0825. Neurooncol Adv 2022; 4:vdac152. [PMID: 36299794 PMCID: PMC9587696 DOI: 10.1093/noajnl/vdac152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background We sought to identify clinical and genetic predictors of temozolomide-related myelotoxicity among patients receiving therapy for glioblastoma. Methods Patients (n = 591) receiving therapy on NRG Oncology/RTOG 0825 were included in the analysis. Cases were patients with severe myelotoxicity (grade 3 and higher leukopenia, neutropenia, and/or thrombocytopenia); controls were patients without such toxicity. A risk-prediction model was built and cross-validated by logistic regression using only clinical variables and extended using polymorphisms associated with myelotoxicity. Results 23% of patients developed myelotoxicity (n = 134). This toxicity was first reported during the concurrent phase of therapy for 56 patients; 30 stopped treatment due to toxicity. Among those who continued therapy (n = 26), 11 experienced myelotoxicity again. The final multivariable clinical factor model included treatment arm, gender, and anticonvulsant status and had low prediction accuracy (area under the curve [AUC] = 0.672). The final extended risk prediction model including four polymorphisms in MGMT had better prediction (AUC = 0.827). Receiving combination chemotherapy (OR, 1.82; 95% CI, 1.02-3.27) and being female (OR, 4.45; 95% CI, 2.45-8.08) significantly increased myelotoxicity risk. For each additional minor allele in the polymorphisms, the risk increased by 64% (OR, 1.64; 95% CI, 1.43-1.89). Conclusions Myelotoxicity during concurrent chemoradiation with temozolomide is an uncommon but serious event, often leading to treatment cessation. Successful prediction of toxicity may lead to more cost-effective individualized monitoring of at-risk subjects. The addition of genetic factors greatly enhanced our ability to predict toxicity among a group of similarly treated glioblastoma patients.
Collapse
Affiliation(s)
- Michael E Scheurer
- Baylor College of Medicine, Departments of Pediatrics and Medicine, Houston, Texas, USA
| | - Renke Zhou
- Baylor College of Medicine, Departments of Pediatrics and Medicine, Houston, Texas, USA
| | - Mark R Gilbert
- National Institutes of Health Clinical Center, Bethesda, MD, USA
| | - Melissa L Bondy
- Baylor College of Medicine, Departments of Pediatrics and Medicine, Houston, Texas, USA
| | - Erik P Sulman
- M D Anderson Cancer Center, Brain and Spine Center, Houston, TX, USA
- Laura and Isaac Perlmutter Cancer Center at NYU Langone, New York, NY, USA
| | - Ying Yuan
- M D Anderson Cancer Center, Brain and Spine Center, Houston, TX, USA
| | - Yanhong Liu
- Baylor College of Medicine, Departments of Pediatrics and Medicine, Houston, Texas, USA
| | - Elizabeth Vera
- National Institutes of Health Clinical Center, Bethesda, MD, USA
- M D Anderson Cancer Center, Brain and Spine Center, Houston, TX, USA
| | - Merideth M Wendland
- National Cancer Institute, Bethesda, MD, USA
- Texas Oncology Cancer Center Sugar Land, Sugar Land, TX, USA
| | | | | | - Ritsuko R Komaki
- M D Anderson Cancer Center, Brain and Spine Center, Houston, TX, USA
| | | | | | - H Ian Robins
- University of Wisconsin Hospital, Madison, WI, USA
| | | | - Ian R Crocker
- Emory University, Winship Cancer Institute, Atlanta, GA, USA
| | | | - Stephanie L Pugh
- NRG Oncology Statistics and Data Management Center, Philadelphia, PA, USA
| | | |
Collapse
|
4
|
Saraiva SM, Gutiérrez-Lovera C, Martínez-Val J, Lores S, Bouzo BL, Díez-Villares S, Alijas S, Pensado-López A, Vázquez-Ríos AJ, Sánchez L, de la Fuente M. Edelfosine nanoemulsions inhibit tumor growth of triple negative breast cancer in zebrafish xenograft model. Sci Rep 2021; 11:9873. [PMID: 33972572 PMCID: PMC8110995 DOI: 10.1038/s41598-021-87968-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 03/29/2021] [Indexed: 12/18/2022] Open
Abstract
Triple negative breast cancer (TNBC) is known for being very aggressive, heterogeneous and highly metastatic. The standard of care treatment is still chemotherapy, with adjacent toxicity and low efficacy, highlighting the need for alternative and more effective therapeutic strategies. Edelfosine, an alkyl-lysophospholipid, has proved to be a promising therapy for several cancer types, upon delivery in lipid nanoparticles. Therefore, the objective of this work was to explore the potential of edelfosine for the treatment of TNBC. Edelfosine nanoemulsions (ET-NEs) composed by edelfosine, Miglyol 812 and phosphatidylcholine as excipients, due to their good safety profile, presented an average size of about 120 nm and a neutral zeta potential, and were stable in biorelevant media. The ability of ET-NEs to interrupt tumor growth in TNBC was demonstrated both in vitro, using a highly aggressive and invasive TNBC cell line, and in vivo, using zebrafish embryos. Importantly, ET-NEs were able to penetrate through the skin barrier of MDA-MB 231 xenografted zebrafish embryos, into the yolk sac, leading to an effective decrease of highly aggressive and invasive tumoral cells' proliferation. Altogether the results demonstrate the potential of ET-NEs for the development of new therapeutic approaches for TNBC.
Collapse
Affiliation(s)
- Sofia M Saraiva
- Nano-Oncology and Translational Therapeutics Unit, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital of Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Spain
- Cancer Network Research (CIBERONC), Madrid, Spain
| | - Carlha Gutiérrez-Lovera
- Department of Zoology, Genetics and Physical Anthropology, Campus of Lugo, University of Santiago de Compostela, Lugo, Spain
| | - Jeannette Martínez-Val
- Department of Zoology, Genetics and Physical Anthropology, Campus of Lugo, University of Santiago de Compostela, Lugo, Spain
| | - Sainza Lores
- Nano-Oncology and Translational Therapeutics Unit, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital of Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Spain
| | - Belén L Bouzo
- Nano-Oncology and Translational Therapeutics Unit, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital of Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Spain
| | - Sandra Díez-Villares
- Nano-Oncology and Translational Therapeutics Unit, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital of Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Spain
- Cancer Network Research (CIBERONC), Madrid, Spain
| | - Sandra Alijas
- Nano-Oncology and Translational Therapeutics Unit, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital of Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Spain
| | - Alba Pensado-López
- Department of Zoology, Genetics and Physical Anthropology, Campus of Lugo, University of Santiago de Compostela, Lugo, Spain
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Abi Judit Vázquez-Ríos
- Nano-Oncology and Translational Therapeutics Unit, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital of Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Spain
- Cancer Network Research (CIBERONC), Madrid, Spain
| | - Laura Sánchez
- Department of Zoology, Genetics and Physical Anthropology, Campus of Lugo, University of Santiago de Compostela, Lugo, Spain
| | - María de la Fuente
- Nano-Oncology and Translational Therapeutics Unit, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital of Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Spain.
- Cancer Network Research (CIBERONC), Madrid, Spain.
| |
Collapse
|
5
|
Mini E, Nobili S. "Pharmacogenetics of Cancer" - Cancer Drug Resistance special issue. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2020; 3:225-231. [PMID: 35582607 PMCID: PMC9090591 DOI: 10.20517/cdr.2020.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 02/05/2020] [Indexed: 11/21/2022]
Affiliation(s)
- Enrico Mini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence 50139, Italy
- Cancer Pharmacology Working Group of the Italian Society of Pharmacology, Milan 20129, Italy
| | - Stefania Nobili
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence 50139, Italy
- Cancer Pharmacology Working Group of the Italian Society of Pharmacology, Milan 20129, Italy
| |
Collapse
|
6
|
Cecchin E, De Mattia E, Ecca F, Toffoli G. Host genetic profiling to increase drug safety in colorectal cancer from discovery to implementation. Drug Resist Updat 2018; 39:18-40. [PMID: 30075835 DOI: 10.1016/j.drup.2018.07.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/11/2018] [Accepted: 07/06/2018] [Indexed: 02/07/2023]
Abstract
Adverse events affect the pharmacological treatment of approximately 90% of colorectal cancer (CRC) patients at any stage of the disease. Chemotherapy including fluoropyrimidines, irinotecan, and oxaliplatin is the cornerstone of the pharmacological treatment of CRC. The introduction of novel targeted agents, as anti-EGFR (i.e. cetuximab, panitumumab) and antiangiogenic (i.e. bevacizumab, ziv-aflibercept, regorafenib, and ramucirumab) molecules, into the oncologist's toolbox has led to significant improvements in the life expectancy of advanced CRC patients, but with a substantial increase in toxicity burden. In this respect, pharmacogenomics has largely been applied to the personalization of CRC chemotherapy, focusing mainly on the study of inhered polymorphisms in genes encoding phase I and II enzymes, ATP-binding cassette (ABC)/solute carrier (SLC) membrane transporters, proteins involved in DNA repair, folate pathway and immune response. These research efforts have led to the identification of some validated genetic markers of chemotherapy toxicity, for fluoropyrimidines and irinotecan. No validated genetic determinants of oxaliplatin-specific toxicity, as peripheral neuropathy, has thus far been established. The contribution of host genetic markers in predicting the toxicity associated with novel targeted agents' administration is still controversial due to the heterogeneity of published data. Pharmacogenomics guidelines have been published by some international scientific consortia such as the Clinical Pharmacogenomics Implementation Consortium (CPIC) and the Dutch Pharmacogenetics Working Group (DPWG) strongly suggesting a pre-treatment dose adjustment of irinotecan based on UGT1A1*28 genotype and of fluoropyrimidines based on some DPYD genetic variants, to increase treatment safety. However, these recommendations are still poorly applied at the patient's bedside. Several ongoing projects in the U.S. and Europe are currently evaluating how pharmacogenomics can be implemented successfully in daily clinical practice. The majority of drug-related adverse events are still unexplained, and a great deal of ongoing research is aimed at improving knowledge of the role of pharmacogenomics in increasing treatment safety. In this review, the issue of pre-treatment identification of CRC patients at risk of toxicity via the analysis of patients' genetic profiles is addressed. Available pharmacogenomics guidelines with ongoing efforts to implement them in clinical practice and new exploratory markers for clinical validation are described.
Collapse
Affiliation(s)
- Erika Cecchin
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico - National Cancer Institute, 33081 Aviano, Italy
| | - Elena De Mattia
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico - National Cancer Institute, 33081 Aviano, Italy
| | - Fabrizio Ecca
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico - National Cancer Institute, 33081 Aviano, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico - National Cancer Institute, 33081 Aviano, Italy.
| |
Collapse
|
7
|
Danielsen HE, Hveem TS, Domingo E, Pradhan M, Kleppe A, Syvertsen RA, Kostolomov I, Nesheim JA, Askautrud HA, Nesbakken A, Lothe RA, Svindland A, Shepherd N, Novelli M, Johnstone E, Tomlinson I, Kerr R, Kerr DJ. Prognostic markers for colorectal cancer: estimating ploidy and stroma. Ann Oncol 2018; 29:616-623. [PMID: 29293881 PMCID: PMC5889021 DOI: 10.1093/annonc/mdx794] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Background We report here the prognostic value of ploidy and digital tumour-stromal morphometric analyses using material from 2624 patients with early stage colorectal cancer (CRC). Patients and methods DNA content (ploidy) and stroma-tumour fraction were estimated using automated digital imaging systems and DNA was extracted from sections of formalin-fixed paraffin-embedded (FFPE) tissue for analysis of microsatellite instability. Samples were available from 1092 patients recruited to the QUASAR 2 trial and two large observational series (Gloucester, n = 954; Oslo University Hospital, n = 578). Resultant biomarkers were analysed for prognostic impact using 5-year cancer-specific survival (CSS) as the clinical end point. Results Ploidy and stroma-tumour fraction were significantly prognostic in a multivariate model adjusted for age, adjuvant treatment, and pathological T-stage in stage II patients, and the combination of ploidy and stroma-tumour fraction was found to stratify these patients into three clinically useful groups; 5-year CSS 90% versus 83% versus 73% [hazard ratio (HR) = 1.77 (95% confidence interval (95% CI): 1.13-2.77) and HR = 2.95 (95% CI: 1.73-5.03), P < 0.001]. Conclusion A novel biomarker, combining estimates of ploidy and stroma-tumour fraction, sampled from FFPE tissue, identifies stage II CRC patients with low, intermediate or high risk of CRC disease specific death, and can reliably stratify clinically relevant patient sub-populations with differential risks of tumour recurrence and may support choice of adjuvant therapy for these individuals.
Collapse
Affiliation(s)
- H E Danielsen
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway; Centre for Cancer Biomedicine, University of Oslo, Oslo, Norway; Nuffield Division of Clinical Laboratory Sciences, University of Oxford, Oxford, UK; Department of Informatics, University of Oslo, Oslo, Norway
| | - T S Hveem
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway; Centre for Cancer Biomedicine, University of Oslo, Oslo, Norway; Department of Informatics, University of Oslo, Oslo, Norway
| | - E Domingo
- Department of Oncology, University of Oxford, Oxford, UK; Molecular and Population Genetics Laboratory, University of Oxford, Oxford, UK; Oxford NIHR Comprehensive Biomedical Research Centre, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - M Pradhan
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway; Centre for Cancer Biomedicine, University of Oslo, Oslo, Norway
| | - A Kleppe
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway; Centre for Cancer Biomedicine, University of Oslo, Oslo, Norway; Department of Informatics, University of Oslo, Oslo, Norway
| | - R A Syvertsen
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway; Centre for Cancer Biomedicine, University of Oslo, Oslo, Norway
| | - I Kostolomov
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| | - J A Nesheim
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway; Centre for Cancer Biomedicine, University of Oslo, Oslo, Norway
| | - H A Askautrud
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway; Centre for Cancer Biomedicine, University of Oslo, Oslo, Norway
| | - A Nesbakken
- Centre for Cancer Biomedicine, University of Oslo, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo; Department of Gastrointestinal Surgery, Oslo University Hospital, Oslo; K.G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo
| | - R A Lothe
- Centre for Cancer Biomedicine, University of Oslo, Oslo, Norway; K.G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo; Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital - Norwegian Radium Hospital, Oslo; Department of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo
| | - A Svindland
- Institute of Clinical Medicine, University of Oslo, Oslo; Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - N Shepherd
- Gloucestershire Cellular Pathology Laboratory, Cheltenham General Hospital, Cheltenham
| | - M Novelli
- Research Department of Pathology, University College London Medical School, London, UK
| | - E Johnstone
- Department of Oncology, University of Oxford, Oxford, UK
| | - I Tomlinson
- Molecular and Population Genetics Laboratory, University of Oxford, Oxford, UK
| | - R Kerr
- Department of Oncology, University of Oxford, Oxford, UK
| | - D J Kerr
- Nuffield Division of Clinical Laboratory Sciences, University of Oxford, Oxford, UK.
| |
Collapse
|
8
|
Sekar V, Mehrotra DG, Majumder B. Molecular and Functional Diagnostic Tools in Precision Oncology for Urological Malignancies. Indian J Surg Oncol 2017; 8:24-32. [PMID: 28127179 PMCID: PMC5236026 DOI: 10.1007/s13193-016-0591-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 09/12/2016] [Indexed: 12/29/2022] Open
Abstract
Urological malignancies, represented mainly by prostate, bladder, and renal cancers, are some of the leading causes of cancer-related mortalities worldwide. Despite various efforts over decades to develop early detection tests and effective therapeutic paradigms, the response rate to the existing treatments remains low for both primary and late stage/recurrent phases of these cancers. The evolving landscape of molecular diagnostics, aiming to make the diagnosis and treatment more patient-driven, underpins precision oncology and particularly intends to rationally profile individual tumors and highlight the mechanistic insight and complexity of tumor microenvironment in order to develop biomarkers of toxicity risks and response prediction in a clinically oriented dynamical setting. The present review is an effort to capture some of the recent developments in the area of molecular diagnostics and functional testing platforms and their potential application in clinical decision making in the premises of precision oncology of urological malignancies.
Collapse
Affiliation(s)
- Vasanthakumar Sekar
- Department of Cancer Biology, Mitra Biotech, 202, Narayana Nethralaya, Health City, Hosur Main Road, Bangalore, 560099 India
| | - Debapriya Ghosh Mehrotra
- Department of Molecular Pathology, Mitra Biotech, 202, Narayana Nethralaya, Health City, Hosur Main Road, Bangalore, 560099 India
| | - Biswanath Majumder
- Department of Molecular Pathology, Mitra Biotech, 202, Narayana Nethralaya, Health City, Hosur Main Road, Bangalore, 560099 India
| |
Collapse
|
9
|
Glaire MA, Brown M, Church DN, Tomlinson I. Cancer predisposition syndromes: lessons for truly precision medicine. J Pathol 2017; 241:226-235. [PMID: 27859368 DOI: 10.1002/path.4842] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 11/04/2016] [Accepted: 11/05/2016] [Indexed: 02/11/2024]
Abstract
Cancer predisposition syndromes are typically uncommon, monogenic, high-penetrance disorders. Despite their rarity, they have proven to be highly clinically relevant in directing cancer prevention strategies. As such, they share notable similarities with an expanding class of low-frequency somatic mutations that are associated with a striking prognostic or predictive effect in the tumours in which they occur. In this review, we highlight these commonalities, with particular reference to mutations in the proofreading domain of replicative DNA polymerases. These molecular phenotypes may occur as either germline or somatic events, and in the latter case, have been shown to confer a favourable prognosis and potential increased benefit from immune checkpoint inhibition. We note that incorporation of these variants into clinical management algorithms will help refine patient management, and that this will be further improved by the inclusion of other germline variants, such as those that determine the likelihood of benefit or toxicity from anti-neoplastic therapy. Finally, we propose that such integrated patient and tumour profiling will be essential if we are to deliver truly precision medicine for cancer patients, but in a similar way to rare germline mutations, we must ensure that we identify and utilize rare somatic mutations with strong predictive and prognostic effects. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Mark A Glaire
- Tumour Genomics and Immunology Group, The Oxford Centre for Cancer Gene Research, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - Matthew Brown
- Tumour Genomics and Immunology Group, The Oxford Centre for Cancer Gene Research, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - David N Church
- Tumour Genomics and Immunology Group, The Oxford Centre for Cancer Gene Research, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - Ian Tomlinson
- Molecular and Population Genetics Laboratory, The Oxford Centre for Cancer Gene Research, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK
| |
Collapse
|
10
|
Evensen L, Johansen PL, Koster G, Zhu K, Herfindal L, Speth M, Fenaroli F, Hildahl J, Bagherifam S, Tulotta C, Prasmickaite L, Mælandsmo GM, Snaar-Jagalska E, Griffiths G. Zebrafish as a model system for characterization of nanoparticles against cancer. NANOSCALE 2016; 8:862-77. [PMID: 26648525 DOI: 10.1039/c5nr07289a] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Therapeutic nanoparticles (NPs) have great potential to deliver drugs against human diseases. Encapsulation of drugs in NPs protects them from being metabolized, while they are delivered specifically to a target site, thereby reducing toxicity and other side-effects. However, non-specific tissue accumulation of NPs, for example in macrophages, especially in the spleen and liver is a general problem with many NPs being developed for cancer therapy. To address the problem of non-specific tissue accumulation of NPs we describe the development of the zebrafish embryo as a transparent vertebrate system for characterization of NPs against cancer. We show that injection of human cancer cells results in tumor-like structures, and that subsequently injected fluorescent NPs, either made of polystyrene or liposomes can be imaged in real-time. NP biodistribution and general in vivo properties can be easily monitored in embryos having selective fluorescent labeling of specific tissues. We demonstrate in vitro, by using optical tweezer micromanipulation, microscopy and flow cytometry that polyethylene glycol (PEG) coating of NPs decreases the level of adhesion of NPs to macrophages, and also to cancer cells. In vivo in zebrafish embryos, PEG coating resulted in longer NP circulation times, decreased macrophage uptake, and reduced adhesion to the endothelium. Importantly, liposomes were observed to accumulate passively and selectively in tumor-like structures comprised of human cancer cells. These results show that zebrafish embryo is a powerful system for microscopy-based screening of NPs on the route to preclinical testing.
Collapse
Affiliation(s)
- Lasse Evensen
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway.
| | - Patrick L Johansen
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway.
| | - Gerbrand Koster
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway.
| | - Kaizheng Zhu
- Department of Chemistry, University of Oslo, Sem Sælands vei 26, 0371, Oslo, Norway
| | - Lars Herfindal
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, 5009 Bergen, Norway
| | - Martin Speth
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway.
| | - Federico Fenaroli
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway.
| | - Jon Hildahl
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway.
| | - Shahla Bagherifam
- Department of Chemistry, University of Oslo, Sem Sælands vei 26, 0371, Oslo, Norway
| | - Claudia Tulotta
- Institute of Biology, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Lina Prasmickaite
- Department of Tumour Biology, Oslo University Hospital Radiumhospital, Oslo, Norway
| | - Gunhild M Mælandsmo
- Department of Tumour Biology, Oslo University Hospital Radiumhospital, Oslo, Norway
| | - Ewa Snaar-Jagalska
- Institute of Biology, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Gareth Griffiths
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway.
| |
Collapse
|
11
|
Widder J, van der Schaaf A, Lambin P, Marijnen CAM, Pignol JP, Rasch CR, Slotman BJ, Verheij M, Langendijk JA. The Quest for Evidence for Proton Therapy: Model-Based Approach and Precision Medicine. Int J Radiat Oncol Biol Phys 2015; 95:30-36. [PMID: 26684410 DOI: 10.1016/j.ijrobp.2015.10.004] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 10/01/2015] [Indexed: 02/07/2023]
Abstract
PURPOSE Reducing dose to normal tissues is the advantage of protons versus photons. We aimed to describe a method for translating this reduction into a clinically relevant benefit. METHODS AND MATERIALS Dutch scientific and health care governance bodies have recently issued landmark reports regarding generation of relevant evidence for new technologies in health care including proton therapy. An approach based on normal tissue complication probability (NTCP) models has been adopted to select patients who are most likely to experience fewer (serious) adverse events achievable by state-of-the-art proton treatment. RESULTS By analogy with biologically targeted therapies, the technology needs to be tested in enriched cohorts of patients exhibiting the decisive predictive marker: difference in normal tissue dosimetric signatures between proton and photon treatment plans. Expected clinical benefit is then estimated by virtue of multifactorial NTCP models. In this sense, high-tech radiation therapy falls under precision medicine. As a consequence, randomizing nonenriched populations between photons and protons is predictably inefficient and likely to produce confusing results. CONCLUSIONS Validating NTCP models in appropriately composed cohorts treated with protons should be the primary research agenda leading to urgently needed evidence for proton therapy.
Collapse
Affiliation(s)
- Joachim Widder
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Arjen van der Schaaf
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Philippe Lambin
- Department of Radiation Oncology, School for Oncology and Developmental Biology (GROW), Maastricht University Medical Center, Maastricht, The Netherlands
| | - Corrie A M Marijnen
- Department of Radiation Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jean-Philippe Pignol
- Department of Radiation Oncology, Erasmus Medical Center Cancer Institute, Rotterdam, The Netherlands
| | - Coen R Rasch
- Department of Radiation Oncology, Academic Medical Center, Amsterdam, The Netherlands
| | - Ben J Slotman
- Department of Radiation Oncology, VU Medical Center, Amsterdam, The Netherlands
| | - Marcel Verheij
- Department of Radiation Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Johannes A Langendijk
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
12
|
Callens C, Debled M, Delord M, Turbiez-Stalain I, Veyret C, Bièche I, Brain E. High-throughput pharmacogenetics identifies SLCO1A2 polymorphisms as candidates to elucidate the risk of febrile neutropenia in the breast cancer RAPP-01 trial. Breast Cancer Res Treat 2015; 153:383-9. [DOI: 10.1007/s10549-015-3552-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 08/13/2015] [Indexed: 11/24/2022]
|
13
|
Zheng CL, Ratnakar V, Gil Y, McWeeney SK. Use of semantic workflows to enhance transparency and reproducibility in clinical omics. Genome Med 2015; 7:73. [PMID: 26289940 PMCID: PMC4545705 DOI: 10.1186/s13073-015-0202-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 07/14/2015] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Recent highly publicized cases of premature patient assignment into clinical trials, resulting from non-reproducible omics analyses, have prompted many to call for a more thorough examination of translational omics and highlighted the critical need for transparency and reproducibility to ensure patient safety. The use of workflow platforms such as Galaxy and Taverna have greatly enhanced the use, transparency and reproducibility of omics analysis pipelines in the research domain and would be an invaluable tool in a clinical setting. However, the use of these workflow platforms requires deep domain expertise that, particularly within the multi-disciplinary fields of translational and clinical omics, may not always be present in a clinical setting. This lack of domain expertise may put patient safety at risk and make these workflow platforms difficult to operationalize in a clinical setting. In contrast, semantic workflows are a different class of workflow platform where resultant workflow runs are transparent, reproducible, and semantically validated. Through semantic enforcement of all datasets, analyses and user-defined rules/constraints, users are guided through each workflow run, enhancing analytical validity and patient safety. METHODS To evaluate the effectiveness of semantic workflows within translational and clinical omics, we have implemented a clinical omics pipeline for annotating DNA sequence variants identified through next generation sequencing using the Workflow Instance Generation and Specialization (WINGS) semantic workflow platform. RESULTS We found that the implementation and execution of our clinical omics pipeline in a semantic workflow helped us to meet the requirements for enhanced transparency, reproducibility and analytical validity recommended for clinical omics. We further found that many features of the WINGS platform were particularly primed to help support the critical needs of clinical omics analyses. CONCLUSIONS This is the first implementation and execution of a clinical omics pipeline using semantic workflows. Evaluation of this implementation provides guidance for their use in both translational and clinical settings.
Collapse
Affiliation(s)
- Christina L Zheng
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR, USA.
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.
| | - Varun Ratnakar
- Information Sciences Institute, University of Southern California, Los Angeles, CA, USA.
| | - Yolanda Gil
- Information Sciences Institute, University of Southern California, Los Angeles, CA, USA.
| | - Shannon K McWeeney
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR, USA.
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.
- Division of Biostatistics, Department of Public Health and Preventative Medicine, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
14
|
HUS1 regulates in vivo responses to genotoxic chemotherapies. Oncogene 2015; 35:662-9. [PMID: 25915840 DOI: 10.1038/onc.2015.118] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 03/08/2015] [Accepted: 03/10/2015] [Indexed: 12/14/2022]
Abstract
Cells are under constant attack from genotoxins and rely on a multifaceted DNA damage response (DDR) network to maintain genomic integrity. Central to the DDR are the ATM and ATR kinases, which respond primarily to double-strand DNA breaks (DSBs) and replication stress, respectively. Optimal ATR signaling requires the RAD9A-RAD1-HUS1 (9-1-1) complex, a toroidal clamp that is loaded at damage sites and scaffolds signaling and repair factors. Whereas complete ATR pathway inactivation causes embryonic lethality, partial Hus1 impairment has been accomplished in adult mice using hypomorphic (Hus1(neo)) and null (Hus1(Δ1)) Hus1 alleles, and here we use this system to define the tissue- and cell type-specific actions of the HUS1-mediated DDR in vivo. Hus1(neo/Δ1) mice showed hypersensitivity to agents that cause replication stress, including the crosslinking agent mitomycin C (MMC) and the replication inhibitor hydroxyurea, but not the DSB inducer ionizing radiation. Analysis of tissue morphology, genomic instability, cell proliferation and apoptosis revealed that MMC treatment caused severe damage in highly replicating tissues of mice with partial Hus1 inactivation. The role of the 9-1-1 complex in responding to MMC was partially ATR-independent, as a HUS1 mutant that was proficient for ATR-induced checkpoint kinase 1 phosphorylation nevertheless conferred MMC hypersensitivity. To assess the interplay between the ATM and ATR pathways in responding to replication stress in vivo, we used Hus1/Atm double mutant mice. Whereas Hus1(neo/neo) and Atm(-/-) single mutant mice survived low-dose MMC similar to wild-type controls, Hus1(neo/neo)Atm(-/-) double mutants showed striking MMC hypersensitivity, consistent with a model in which MMC exposure in the context of Hus1 dysfunction results in DSBs to which the ATM pathway normally responds. This improved understanding of the inter-dependency between two major DDR mechanisms during the response to a conventional chemotherapeutic illustrates how inhibition of checkpoint factors such as HUS1 may be effective for the treatment of ATM-deficient and other cancers.
Collapse
|
15
|
Innocenti F. DPYD variants to predict 5-FU toxicity: the ultimate proof. J Natl Cancer Inst 2014; 106:dju351. [PMID: 25381394 DOI: 10.1093/jnci/dju351] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Federico Innocenti
- Eshelman School of Pharmacy, Center for Pharmacogenomics and Individualized Therapy, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC.
| |
Collapse
|
16
|
Abstract
In the past decade, we have witnessed unprecedented changes and some remarkable advances that have enabled true personalized medicine. Nevertheless, many challenges in clinical cancer research remain and need to be overcome if we are to witness similar progress in the next decade. Such hurdles include, but are not limited to, clinical development and testing of multiple agents in combination, design of clinical trials to best accommodate the ever increasing knowledge of heterogeneity of the disease, regulatory challenges relating to drug development and trial design, and funding for basic research. With this in mind, we asked four leading cancer researchers from around the world, and who have been associated with the journal since its launch in November 2004 what, in their opinion, we have learnt over the past 10 years and how we should progress in the next 10 years.
Collapse
|
17
|
Abstract
The cancer community is deeply concerned about the unintended consequences of the current wording of the European Union (EU) draft Regulation on Data Protection, which may challenge the survival of retrospective clinical research, biobanking, and population-based cancer registries in the EU. This directive could negatively affect Europe's competitiveness in cancer research.
Collapse
Affiliation(s)
- David J Kerr
- Department of Medicine, Nuffield Division of Clinical and Laboratory Sciences, Level 4, Academic Block, John Radcliffe Infirmary, Headington, Oxford OX3 9DU, UK
| |
Collapse
|
18
|
Johnson R, Newport R, Kerr R, Kerr D. Toxgnostics: predicting and preventing chemotherapy-induced side effects. Per Med 2014; 11:683-685. [PMID: 29764049 DOI: 10.2217/pme.14.56] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Rebecca Johnson
- Oxford Cancer Centre, Department of Oncology, University of Oxford, Churchill Hospital, Old Road, Headington, Oxford OX3 7LE, UK
| | - Rhiana Newport
- Oxford Cancer Centre, Department of Oncology, University of Oxford, Churchill Hospital, Old Road, Headington, Oxford OX3 7LE, UK
| | - Rachel Kerr
- Oxford Cancer Centre, Department of Oncology, University of Oxford, Churchill Hospital, Old Road, Headington, Oxford OX3 7LE, UK
| | - David Kerr
- Nuffield Department of Clinical Laboratory Sciences, University of Oxford, John Radcliffe Hospital, Headley Way, Oxford OX3 9DU, UK
| |
Collapse
|
19
|
Murray BS, Crot S, Siankevich S, Dyson PJ. Potential of cycloaddition reactions to generate cytotoxic metal drugs in vitro. Inorg Chem 2014; 53:9315-21. [PMID: 25133591 DOI: 10.1021/ic501438k] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Severe general toxicity issues blight many chemotherapeutics utilized in the treatment of cancers, resulting in the need for more selective drugs able to exert their biological activity at only the required location(s). Toward this aim, we report the development of an organometallic ruthenium compound, functionalized through a η(6)-bound arene ligand with a bicyclononyne derivative, able to participate in strain-promoted cycloaddition reactions with tetrazines. We show that combination of the ruthenium compound with a ditetrazine in biological media results in the in situ formation of a dinuclear molecule that is more cytotoxic toward cancer cells than the starting mononuclear ruthenium compound and tetrazine components. Such an approach may be extended to in vivo applications to construct a cytotoxic metallodrug at a tumor site, providing a novel approach toward the turn-on cytotoxicity of metallodrugs in the treatment of cancer.
Collapse
Affiliation(s)
- Benjamin S Murray
- Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL) , CH-1015, Lausanne, Switzerland
| | | | | | | |
Collapse
|