1
|
Di Giovannantonio M, Hartley F, Elshenawy B, Barberis A, Hudson D, Shafique HS, Allott VES, Harris DA, Lord SR, Haider S, Harris AL, Buffa FM, Harris BHL. Defining hypoxia in cancer: A landmark evaluation of hypoxia gene expression signatures. CELL GENOMICS 2025; 5:100764. [PMID: 39892389 PMCID: PMC11872601 DOI: 10.1016/j.xgen.2025.100764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/04/2024] [Accepted: 01/07/2025] [Indexed: 02/03/2025]
Abstract
Tumor hypoxia drives metabolic shifts, cancer progression, and therapeutic resistance. Challenges in quantifying hypoxia have hindered the exploitation of this potential "Achilles' heel." While gene expression signatures have shown promise as surrogate measures of hypoxia, signature usage is heterogeneous and debated. Here, we present a systematic pan-cancer evaluation of 70 hypoxia signatures and 14 summary scores in 104 cell lines and 5,407 tumor samples using 472 million length-matched random gene signatures. Signature and score choice strongly influenced the prediction of hypoxia in vitro and in vivo. In cell lines, the Tardon signature was highly accurate in both bulk and single-cell data (94% accuracy, interquartile mean). In tumors, the Buffa and Ragnum signatures demonstrated superior performance, with Buffa/mean and Ragnum/interquartile mean emerging as the most promising for prospective clinical trials. This work delivers recommendations for experimental hypoxia detection and patient stratification for hypoxia-targeting therapies, alongside a generalizable framework for signature evaluation.
Collapse
Affiliation(s)
- Matteo Di Giovannantonio
- Computational Biology and Integrative Genomics Lab, Department of Oncology, University of Oxford, Oxford, UK
| | - Fiona Hartley
- Computational Biology and Integrative Genomics Lab, Department of Oncology, University of Oxford, Oxford, UK
| | - Badran Elshenawy
- Computational Biology and Integrative Genomics Lab, Department of Oncology, University of Oxford, Oxford, UK
| | - Alessandro Barberis
- Computational Biology and Integrative Genomics Lab, Department of Oncology, University of Oxford, Oxford, UK
| | - Dan Hudson
- Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK; The Rosalind Franklin Institute, Didcot, UK
| | | | | | | | - Simon R Lord
- Computational Biology and Integrative Genomics Lab, Department of Oncology, University of Oxford, Oxford, UK
| | - Syed Haider
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, UK
| | - Adrian L Harris
- Computational Biology and Integrative Genomics Lab, Department of Oncology, University of Oxford, Oxford, UK
| | - Francesca M Buffa
- Computational Biology and Integrative Genomics Lab, Department of Oncology, University of Oxford, Oxford, UK; CompBio Lab, Department of Computing Sciences, Bocconi University, Milan, Italy; AI and Systems Biology Lab, IFOM - Istituto Fondazione di Oncologia Molecolare ETS, Milan, Italy.
| | - Benjamin H L Harris
- Computational Biology and Integrative Genomics Lab, Department of Oncology, University of Oxford, Oxford, UK; St. Catherine's College, University of Oxford, Oxford, UK; Cutrale Perioperative and Ageing Group, Imperial College London, London, UK.
| |
Collapse
|
2
|
Shamis SAK, Quinn J, Al‐Badran S, McKenzie M, Hatthakarnkul P, Lynch G, Lian G, Numprasit W, Romics L, Andersen D, Mallon E, McMillan DC, Edwards J. Elucidation of Dysregulated Pathways Associated With Hypoxia in Oestrogen Receptor-Negative Breast Cancer. Cancer Med 2024; 13:e70274. [PMID: 39660488 PMCID: PMC11632397 DOI: 10.1002/cam4.70274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 07/06/2024] [Accepted: 09/20/2024] [Indexed: 12/12/2024] Open
Abstract
PURPOSE Carbonic anhydrase IX (CAIX) is a well-established prognostic marker in breast cancer (BC). Nevertheless, this prognostic value is yet to be confirmed in BC subtypes. This study aims to investigate the prognostic effects of CAIX in oestrogen receptor (ER)-negative (ER-) BCs and to establish pathways related to cytoplasmic CAIX expression in ER- and lymph node-negative BCs. METHODS Immunohistochemistry was performed to identify the prognostic role of CAIX protein expression in ER- tissue microarrays (TMAs) (n = 191). CAIX-positive samples (n = 37) were transcriptionally profiled by TempO-Seq and analysed by STRING. Real-time quantitative PCR (RT-qPCR) analysis was used to validate differentially expressed genes. RESULTS Overexpression of cytoplasmic CAIX was an independent predictor of recurrence free survival, disease-free survival and overall survival in ER- cohort. RNA transcriptomic analysis identified 10 significant genes in ER- cohort and 3 genes in the node-negative group. The STRING database demonstrated a significant interaction between MUCL1 and GALNT6, which were linked with extracellular matrix organisation, degradation of the extracellular matrix and disease of glycosylation pathways. In the node-negative group, SPNS2 is mainly involved in the sphingolipid de novo biosynthesis pathway. A significant correlation between cytoplasmic SphK1 and cytoplasmic hypoxia-inducible factor-1α was observed. Among the 10 genes, 7 genes (SERHL2, GALNT6, MUCL1, MMP7, PITX2, CEACAM6 and SPNS2) were selected, and their expression was quantitatively assessed by RT-qPCR. The PCR data of these genes showed that SERHL2, GALNT6, MUCL1, PITX2, and SPNS2 mRNA levels were expressed in MDA-MB-231 BC cell lines at variable levels of hypoxic exposure. CONCLUSION Cytoplasmic CAIX was independently associated with poor prognosis in ER- BC. Gene expression profiles shed light on the pathways and genes associated with hypoxia in ER- BC. In node-negative patients, SPNS2 was of particular interest.
Collapse
Affiliation(s)
- Suad A. K. Shamis
- Academic Unit of Surgery, School of MedicineUniversity of Glasgow, Royal InfirmaryGlasgowUK
- Wolfson Wohl Cancer Research Centre, School of Cancer SciencesUniversity of GlasgowGlasgowUK
| | - Jean Quinn
- Wolfson Wohl Cancer Research Centre, School of Cancer SciencesUniversity of GlasgowGlasgowUK
| | - Sara Al‐Badran
- Wolfson Wohl Cancer Research Centre, School of Cancer SciencesUniversity of GlasgowGlasgowUK
| | - Molly McKenzie
- Wolfson Wohl Cancer Research Centre, School of Cancer SciencesUniversity of GlasgowGlasgowUK
| | - Phimmada Hatthakarnkul
- Wolfson Wohl Cancer Research Centre, School of Cancer SciencesUniversity of GlasgowGlasgowUK
| | - Gerard Lynch
- Wolfson Wohl Cancer Research Centre, School of Cancer SciencesUniversity of GlasgowGlasgowUK
| | - Guang‐Yu Lian
- Wolfson Wohl Cancer Research Centre, School of Cancer SciencesUniversity of GlasgowGlasgowUK
| | - Warapan Numprasit
- Wolfson Wohl Cancer Research Centre, School of Cancer SciencesUniversity of GlasgowGlasgowUK
| | - Laszlo Romics
- NHS Greater Glasgow and Clyde (New Victoria Hospital)GlasgowUK
| | - Ditte Andersen
- BioClavis Ltd., Teaching & Learning CentreQueen Elizabeth University HospitalGlasgowUK
| | - Elizabeth Mallon
- Department of PathologyQueen Elizabeth University HospitalGlasgowUK
| | - Donald C. McMillan
- Academic Unit of Surgery, School of MedicineUniversity of Glasgow, Royal InfirmaryGlasgowUK
| | - Joanne Edwards
- Wolfson Wohl Cancer Research Centre, School of Cancer SciencesUniversity of GlasgowGlasgowUK
| |
Collapse
|
3
|
Besso MJ, Bitto V, Koi L, Wijaya Hadiwikarta W, Conde-Lopez C, Euler-Lange R, Bonrouhi M, Schneider K, Linge A, Krause M, Baumann M, Kurth I. Transcriptomic and epigenetic landscape of nimorazole-enhanced radiochemotherapy in head and neck cancer. Radiother Oncol 2024; 199:110348. [PMID: 38823583 DOI: 10.1016/j.radonc.2024.110348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 04/27/2024] [Accepted: 05/20/2024] [Indexed: 06/03/2024]
Abstract
BACKGROUND Hypoxia remains a challenge for the therapeutic management of head and neck squamous cell carcinoma (HNSCC). The combination of radiotherapy with nimorazole has shown treatment benefit in HNSCC, but the precise underlying molecular mechanisms remain unclear. PURPOSE To assess and to characterize the transcriptomic/epigenetic landscape of HNSCC tumor models showing differential therapeutic response to fractionated radiochemotherapy (RCTx) combined with nimorazole. MATERIALS/METHODS Bulk RNA-sequencing and DNA methylation experiments were conducted using untreated and treated HNSCC xenografts after 10 fractions of RCTx with and without nimorazole. These tumor models (FaDu, SAS, Cal33, SAT and UT-SCC-45) previously showed a heterogeneous response to RCTx with nimorazole. The prognostic impact of candidate genes was assessed using clinical and gene expression data from HNSCC patients treated with primary RCTx within the DKTK-ROG. RESULTS Nimorazole responder and non-responder tumor models showed no differences in hypoxia gene signatures However, non-responder models showed upregulation of metabolic pathways. From that, a subset of 15 differentially expressed genes stratified HNSCC patients into low and high-risk groups with distinct outcome. CONCLUSION In the present study, we found that nimorazole non-responder models were characterized by upregulation of genes involved in Retinol metabolism and xenobiotic metabolic process pathways, which might contribute to identify mechanisms of resistance to nitroimidazole compounds and potentially expand the repertoire of therapeutic options to treat HNSCC.
Collapse
Affiliation(s)
- María José Besso
- German Cancer Research Center (DKFZ) Heidelberg, Division of Radiooncology Radiobiology, Germany.
| | - Verena Bitto
- German Cancer Research Center (DKFZ) Heidelberg, Division of Radiooncology Radiobiology, Germany; German Cancer Research Center (DKFZ) Heidelberg, Division of Applied Bioinformatics, Germany; Faculty for Mathematics and Computer Science, Heidelberg University, Germany; HIDSS4Health - Helmholtz Information and Data Science School for Health, Karlsruhe/Heidelberg, Germany
| | - Lydia Koi
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
| | - Wahyu Wijaya Hadiwikarta
- German Cancer Research Center (DKFZ) Heidelberg, Division of Radiooncology Radiobiology, Germany; National Center for Radiation Research in Oncology (NCRO), Heidelberg Institute for Radiation Oncology (HIRO), Heidelberg, Germany
| | - Cristina Conde-Lopez
- German Cancer Research Center (DKFZ) Heidelberg, Division of Radiooncology Radiobiology, Germany
| | - Rosemarie Euler-Lange
- German Cancer Research Center (DKFZ) Heidelberg, Division of Radiooncology Radiobiology, Germany
| | - Mahnaz Bonrouhi
- German Cancer Research Center (DKFZ) Heidelberg, Division of Radiooncology Radiobiology, Germany
| | - Karolin Schneider
- German Cancer Research Center (DKFZ) Heidelberg, Division of Radiooncology Radiobiology, Germany
| | - Annett Linge
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; German Cancer Research Center (DKFZ), and German Cancer Consortium (DKTK), Partner Site Dresden, Heidelberg, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, and Helmholtz Association / Helmholtz-Zentrum Dresden Rossendorf (HZDR), Dresden, Germany
| | - Mechthild Krause
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany; German Cancer Research Center (DKFZ), and German Cancer Consortium (DKTK), Partner Site Dresden, Heidelberg, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, and Helmholtz Association / Helmholtz-Zentrum Dresden Rossendorf (HZDR), Dresden, Germany
| | - Michael Baumann
- German Cancer Research Center (DKFZ) Heidelberg, Division of Radiooncology Radiobiology, Germany; OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany; German Cancer Research Center (DKFZ), and German Cancer Consortium (DKTK), Core Center Heidelberg, Heidelberg, Germany; National Center for Radiation Research in Oncology (NCRO), Heidelberg Institute for Radiation Oncology (HIRO), Heidelberg, Germany
| | - Ina Kurth
- German Cancer Research Center (DKFZ) Heidelberg, Division of Radiooncology Radiobiology, Germany; German Cancer Research Center (DKFZ), and German Cancer Consortium (DKTK), Core Center Heidelberg, Heidelberg, Germany; National Center for Radiation Research in Oncology (NCRO), Heidelberg Institute for Radiation Oncology (HIRO), Heidelberg, Germany
| |
Collapse
|
4
|
Lombardi O, Li R, Halim S, Choudhry H, Ratcliffe PJ, Mole DR. Pan-cancer analysis of tissue and single-cell HIF-pathway activation using a conserved gene signature. Cell Rep 2022; 41:111652. [PMID: 36384128 PMCID: PMC9869179 DOI: 10.1016/j.celrep.2022.111652] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 08/05/2022] [Accepted: 10/20/2022] [Indexed: 11/17/2022] Open
Abstract
Activation of cellular hypoxia pathways, orchestrated by HIF (hypoxia-inducible factor) transcription factors, is a common feature of multiple tumor types, resulting from microenvironment factors and oncogenic mutation. Although they help drive many of the "hallmarks" of cancer and are associated with poor outcome and resistance to therapy, the transcriptional targets of HIF vary considerably depending on the cell type. By integrating 72 genome-wide assays of HIF binding and transcriptional regulation from multiple cancer types, we define a consensus set of 48 HIF target genes that is highly conserved across cancer types and cell lineages. These genes provide an effective marker of HIF activation in bulk and single-cell transcriptomic analyses across a wide range of cancer types and in malignant and stromal cell types. This allows the tissue-orchestrated responses to the hypoxic tumor microenvironment and to oncogenic HIF activation to be deconvoluted at the tumor and single-cell level.
Collapse
Affiliation(s)
- Olivia Lombardi
- NDM Research Building, University of Oxford, Old Road Campus, Headington, Oxford OX3 7FZ, UK
| | - Ran Li
- NDM Research Building, University of Oxford, Old Road Campus, Headington, Oxford OX3 7FZ, UK
| | - Silvia Halim
- NDM Research Building, University of Oxford, Old Road Campus, Headington, Oxford OX3 7FZ, UK
| | - Hani Choudhry
- Department of Biochemistry, Faculty of Science, Center of Innovation in Personalized Medicine, King Fahd Center for Medical Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Peter J Ratcliffe
- Ludwig Institute for Cancer Research, University of Oxford, Old Road Campus, Headington, Oxford OX3 7FZ, UK; The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - David R Mole
- NDM Research Building, University of Oxford, Old Road Campus, Headington, Oxford OX3 7FZ, UK.
| |
Collapse
|
5
|
Suthen S, Lim CJ, Nguyen PHD, Dutertre CA, Lai HLH, Wasser M, Chua C, Lim TKH, Leow WQ, Loh TJ, Wan WK, Pang YH, Soon G, Cheow PC, Kam JH, Iyer S, Kow A, Tam WL, Shuen TWH, Toh HC, Dan YY, Bonney GK, Chan CY, Chung A, Goh BKP, Zhai W, Ginhoux F, Chow PKH, Albani S, Chew V. Hypoxia-driven immunosuppression by Treg and type-2 conventional dendritic cells in HCC. Hepatology 2022; 76:1329-1344. [PMID: 35184329 DOI: 10.1002/hep.32419] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/25/2022] [Accepted: 02/07/2022] [Indexed: 01/10/2023]
Abstract
BACKGROUND AND AIMS Hypoxia is one of the central players in shaping the immune context of the tumor microenvironment (TME). However, the complex interplay between immune cell infiltrates within the hypoxic TME of HCC remains to be elucidated. APPROACH AND RESULTS We analyzed the immune landscapes of hypoxia-low and hypoxia-high tumor regions using cytometry by time of light, immunohistochemistry, and transcriptomic analyses. The mechanisms of immunosuppression in immune subsets of interest were further explored using in vitro hypoxia assays. Regulatory T cells (Tregs) and a number of immunosuppressive myeloid subsets, including M2 macrophages and human leukocyte antigen-DR isotype (HLA-DRlo ) type 2 conventional dendritic cell (cDC2), were found to be significantly enriched in hypoxia-high tumor regions. On the other hand, the abundance of active granzyme Bhi PD-1lo CD8+ T cells in hypoxia-low tumor regions implied a relatively active immune landscape compared with hypoxia-high regions. The up-regulation of cancer-associated genes in the tumor tissues and immunosuppressive genes in the tumor-infiltrating leukocytes supported a highly pro-tumorigenic network in hypoxic HCC. Chemokine genes such as CCL20 (C-C motif chemokine ligand 20) and CXCL5 (C-X-C motif chemokine ligand 5) were associated with recruitment of both Tregs and HLA-DRlo cDC2 to hypoxia-high microenvironments. The interaction between Tregs and cDC2 under a hypoxic TME resulted in a loss of antigen-presenting HLA-DR on cDC2. CONCLUSIONS We uncovered the unique immunosuppressive landscapes and identified key immune subsets enriched in hypoxic HCC. In particular, we identified a potential Treg-mediated immunosuppression through interaction with a cDC2 subset in HCC that could be exploited for immunotherapies.
Collapse
Affiliation(s)
- Sheena Suthen
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Center, Singapore
| | - Chun Jye Lim
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Center, Singapore
| | - Phuong H D Nguyen
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Center, Singapore
| | - Charles-Antoine Dutertre
- Gustave Roussy Cancer Campus, Villejuif, France.,Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale Contre le Cancer, Villejuif, France
| | - Hannah L H Lai
- Agency for Science, Technology and Research, Genome Institute of Singapore, Singapore
| | - Martin Wasser
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Center, Singapore
| | - Camillus Chua
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Center, Singapore
| | - Tony K H Lim
- Duke-NUS Medical School, Singapore.,Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Wei Qiang Leow
- Duke-NUS Medical School, Singapore.,Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Tracy Jiezhen Loh
- Duke-NUS Medical School, Singapore.,Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Wei Keat Wan
- Duke-NUS Medical School, Singapore.,Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Yin Huei Pang
- Department of Pathology, National University Hospital, Singapore
| | - Gwyneth Soon
- Department of Pathology, National University Hospital, Singapore
| | - Peng Chung Cheow
- Duke-NUS Medical School, Singapore.,Division of Surgery and Surgical Oncology, Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital and National Cancer Center Singapore, Singapore
| | - Juinn Huar Kam
- Duke-NUS Medical School, Singapore.,Division of Surgery and Surgical Oncology, Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital and National Cancer Center Singapore, Singapore
| | - Shridhar Iyer
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, University Surgical Cluster, National University Health System, Singapore
| | - Alfred Kow
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, University Surgical Cluster, National University Health System, Singapore
| | - Wai Leong Tam
- Agency for Science, Technology and Research, Genome Institute of Singapore, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Timothy W H Shuen
- Division of Medical Oncology, National Cancer Center Singapore, Singapore
| | - Han Chong Toh
- Duke-NUS Medical School, Singapore.,Division of Medical Oncology, National Cancer Center Singapore, Singapore
| | - Yock Young Dan
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Glenn K Bonney
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, University Surgical Cluster, National University Health System, Singapore
| | - Chung Yip Chan
- Duke-NUS Medical School, Singapore.,Division of Surgery and Surgical Oncology, Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital and National Cancer Center Singapore, Singapore
| | - Alexander Chung
- Duke-NUS Medical School, Singapore.,Division of Surgery and Surgical Oncology, Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital and National Cancer Center Singapore, Singapore
| | - Brian K P Goh
- Duke-NUS Medical School, Singapore.,Division of Surgery and Surgical Oncology, Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital and National Cancer Center Singapore, Singapore
| | - Weiwei Zhai
- Gustave Roussy Cancer Campus, Villejuif, France.,Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| | - Florent Ginhoux
- Gustave Roussy Cancer Campus, Villejuif, France.,Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale Contre le Cancer, Villejuif, France
| | - Pierce K H Chow
- Division of Surgery and Surgical Oncology, Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital and National Cancer Center Singapore, Singapore.,Academic Clinical Programme for Surgery, SingHealth Duke-NUS Academic Medical Centre, Singapore
| | - Salvatore Albani
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Center, Singapore
| | - Valerie Chew
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Center, Singapore
| |
Collapse
|
6
|
SOCS5 knockdown suppresses metastasis of hepatocellular carcinoma by ameliorating HIF-1α-dependent mitochondrial damage. Cell Death Dis 2022; 13:918. [PMID: 36319626 PMCID: PMC9626553 DOI: 10.1038/s41419-022-05361-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 10/14/2022] [Accepted: 10/20/2022] [Indexed: 11/17/2022]
Abstract
The Pringle maneuver (PM) is widely used during hepatocellular carcinoma (HCC) resection. However, it inevitably leads to ischemia and hypoxia, which promotes tumor metastasis. In this study, immunohistochemical staining of specimens from 130 HCC patients revealed that long-time PM significantly affected the prognosis of patients with high expression of suppressor of cytokine signaling 5 (SOCS5), but did not affect the prognosis of patients with low expression of SOCS5. The TCGA database showed that patients with high expression of SOCS5 had higher hypoxia scores, and it was proved that SOCS5 could promote the expression of hypoxia-inducible factor 1 subunit alpha (HIF-1α) protein by clinical tissue samples, cell experiments, lung metastases, and subcutaneous tumorigenesis experiments. Then, we used CoCl2 to construct a hypoxia model, and confirmed that SOCS5 knockdown resisted hypoxia-induced mitochondrial damage by inhibiting the expression of HIF-1α, thereby inhibiting the invasion and migration of HCC cells by immunofluorescence, electron microscopy, migration, invasion, and other experiments. We performed rescue experiments using LY294002 and rapamycin and confirmed that the knockdown of SOCS5-inhibited HCC cell invasion and migration by inhibiting the PI3K/Akt/mTOR/HIF-1α signaling axis. More importantly, we obtained consistent conclusions from clinical, cellular, and animal studies that the hypoxia-induced invasion and migration ability of SOCS5-inhibited HCC were weaker than that of normal HCC. In conclusion, we identified a novel role for SOCS5 in regulating HIF-1α-dependent mitochondrial damage and metastasis through the PI3K/Akt/mTOR pathway. The development of a SOCS5-specific inhibitor, an indirect inhibitor of HIF-1α, might be effective at controlling PM-induced tumor micrometastases during HCC resection.
Collapse
|
7
|
Puente-Santamaría L, Sanchez-Gonzalez L, Ramos-Ruiz R, del Peso L. Hypoxia classifier for transcriptome datasets. BMC Bioinformatics 2022; 23:204. [PMID: 35641902 PMCID: PMC9153107 DOI: 10.1186/s12859-022-04741-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 05/17/2022] [Indexed: 12/02/2022] Open
Abstract
Molecular gene signatures are useful tools to characterize the physiological state of cell populations, but most have developed under a narrow range of conditions and cell types and are often restricted to a set of gene identities. Focusing on the transcriptional response to hypoxia, we aimed to generate widely applicable classifiers sourced from the results of a meta-analysis of 69 differential expression datasets which included 425 individual RNA-seq experiments from 33 different human cell types exposed to different degrees of hypoxia (0.1-5%[Formula: see text]) for 2-48 h. The resulting decision trees include both gene identities and quantitative boundaries, allowing for easy classification of individual samples without control or normoxic reference. Each tree is composed of 3-5 genes mostly drawn from a small set of just 8 genes (EGLN1, MIR210HG, NDRG1, ANKRD37, TCAF2, PFKFB3, BHLHE40, and MAFF). In spite of their simplicity, these classifiers achieve over 95% accuracy in cross validation and over 80% accuracy when applied to additional challenging datasets. Our results indicate that the classifiers are able to identify hypoxic tumor samples from bulk RNAseq and hypoxic regions within tumor from spatially resolved transcriptomics datasets. Moreover, application of the classifiers to histological sections from normal tissues suggest the presence of a hypoxic gene expression pattern in the kidney cortex not observed in other normoxic organs. Finally, tree classifiers described herein outperform traditional hypoxic gene signatures when compared against a wide range of datasets. This work describes a set of hypoxic gene signatures, structured as simple decision tress, that identify hypoxic samples and regions with high accuracy and can be applied to a broad variety of gene expression datasets and formats.
Collapse
Affiliation(s)
- Laura Puente-Santamaría
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain
- Instituto de Investigaciones Biomédicas “Alberto Sols” (CSIC-UAM), 28029 Madrid, Spain
- Genomics Unit Cantoblanco, Fundación Parque Científico de Madrid, C/ Faraday 7, 28049 Madrid, Spain
| | | | - Ricardo Ramos-Ruiz
- Genomics Unit Cantoblanco, Fundación Parque Científico de Madrid, C/ Faraday 7, 28049 Madrid, Spain
| | - Luis del Peso
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain
- Instituto de Investigaciones Biomédicas “Alberto Sols” (CSIC-UAM), 28029 Madrid, Spain
- IdiPaz, Instituto de Investigación Sanitaria del Hospital Universitario La Paz, 28029 Madrid, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Unidad Asociada de Biomedicina CSIC-UCLM, 02006 Albacete, Spain
| |
Collapse
|
8
|
Suwa T, Kobayashi M, Shirai Y, Nam JM, Tabuchi Y, Takeda N, Akamatsu S, Ogawa O, Mizowaki T, Hammond EM, Harada H. SPINK1 as a plasma marker for tumor hypoxia and a therapeutic target for radiosensitization. JCI Insight 2021; 6:e148135. [PMID: 34747365 PMCID: PMC8663551 DOI: 10.1172/jci.insight.148135] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 09/24/2021] [Indexed: 11/17/2022] Open
Abstract
Hypoxia is associated with tumor radioresistance; therefore, a predictive marker for tumor hypoxia and a rational target to overcome it have been sought to realize personalized radiotherapy. Here, we show that serine protease inhibitor Kazal type I (SPINK1) meets these 2 criteria. SPINK1 expression was induced upon hypoxia (O2 < 0.1%) at the transcription initiation level in a HIF-dependent manner, causing an increase in secreted SPINK1 levels. SPINK1 proteins were detected both within and around hypoxic regions of xenografted and clinical tumor tissues, and their plasma levels increased in response to decreased oxygen supply to xenografts. Secreted SPINK1 proteins enhanced radioresistance of cancer cells even under normoxic conditions in EGFR-dependent and nuclear factor erythroid 2-related factor 2-dependent (Nrf2-dependent) manners and accelerated tumor growth after radiotherapy. An anti-SPINK1 neutralizing antibody exhibited a radiosensitizing effect. These results suggest that SPINK1 secreted from hypoxic cells protects the surrounding and relatively oxygenated cancer cells from radiation in a paracrine manner, justifying the use of SPINK1 as a target for radiosensitization and a plasma marker for predicting tumor hypoxia.
Collapse
Affiliation(s)
- Tatsuya Suwa
- Laboratory of Cancer Cell Biology and
- Department of Genome Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Department of Radiation Oncology and Image-applied Therapy, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Minoru Kobayashi
- Laboratory of Cancer Cell Biology and
- Department of Genome Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Yukari Shirai
- Laboratory of Cancer Cell Biology and
- Department of Genome Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Jin-Min Nam
- Laboratory of Cancer Cell Biology and
- Department of Genome Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Yoshiaki Tabuchi
- Division of Molecular Genetics Research, Life Science Research Center, University of Toyama, Toyama, Japan
| | - Norihiko Takeda
- Division of Cardiology and Metabolism, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Shusuke Akamatsu
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Osamu Ogawa
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takashi Mizowaki
- Department of Radiation Oncology and Image-applied Therapy, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ester M. Hammond
- MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Hiroshi Harada
- Laboratory of Cancer Cell Biology and
- Department of Genome Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| |
Collapse
|
9
|
Liu J, Cabral H, Song B, Aoki I, Chen Z, Nishiyama N, Huang Y, Kataoka K, Mi P. Nanoprobe-Based Magnetic Resonance Imaging of Hypoxia Predicts Responses to Radiotherapy, Immunotherapy, and Sensitizing Treatments in Pancreatic Tumors. ACS NANO 2021; 15:13526-13538. [PMID: 34355882 DOI: 10.1021/acsnano.1c04263] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Accurate diagnosis of tumors and predicting the therapeutic responses are highly demanded in the clinic to improve the treatment efficacy and survival rates. Since hypoxia develops in the progression of tumors and inversely correlates with prognosis and promotes resistance to radiotherapies and immunotherapies, it is a potential marker for therapeutic prediction. Therefore, effective discrimination of tumor hypoxia for predicting therapeutic outcomes is critical. Here, a magnetic resonance imaging (MRI)-based diagnosis strategy using contrast-amplifying nanoprobes that sense tumor acidosis and real-time observation of hypoxic conditions in tumors has been developed, aiming at accurate detection of pancreatic tumors and prediction of therapeutic effects. Our approach selectively probed xenograft, allograft, and transgenic spontaneous models of intractable pancreatic cancer, which lacks standardized predictive markers to identify patients who benefit most from treatments, and effectively discriminated the intratumoral hypoxia levels. By stratification of pancreatic tumors based on quantitative MR imaging of hypoxia, it enabled prediction of the responses to radiotherapy and immune checkpoint inhibitors. Moreover, the nanoprobe-based MRI could monitor hypoxia reduction by tumor normalization treatments, which permits visualizing pancreatic tumors that will respond to immune checkpoint blockade therapy, enhancing the response rate. The results demonstrate the potential of our strategy for accurate tumor diagnosis, patient stratification, and effective therapy.
Collapse
Affiliation(s)
- Jing Liu
- Department of Radiology, Center for Medical Imaging, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, No. 17, South Renmin Road, Chengdu 610041, China
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Bin Song
- Department of Radiology, Center for Medical Imaging, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, No. 17, South Renmin Road, Chengdu 610041, China
| | - Ichio Aoki
- National Institute of Radiological Sciences, Japan Agency for Quantum and Radiological Science and Technology, Anagawa 4-9-1,
Inage, Chiba 263-8555, Japan
| | - Zhouyun Chen
- Department of Radiology, Center for Medical Imaging, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, No. 17, South Renmin Road, Chengdu 610041, China
| | - Nobuhiro Nishiyama
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, R1-11, 4259 Nagatsuta, Midori-ku, Yokohama 226-8503, Japan
| | - Yuan Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, No. 17, South Renmin Road, Chengdu 610041, China
| | - Kazunori Kataoka
- Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
- Institute for Future Initiatives, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Peng Mi
- Department of Radiology, Center for Medical Imaging, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, No. 17, South Renmin Road, Chengdu 610041, China
| |
Collapse
|
10
|
Rashidieh B, Molakarimi M, Mohseni A, Tria SM, Truong H, Srihari S, Adams RC, Jones M, Duijf PHG, Kalimutho M, Khanna KK. Targeting BRF2 in Cancer Using Repurposed Drugs. Cancers (Basel) 2021; 13:cancers13153778. [PMID: 34359683 PMCID: PMC8345145 DOI: 10.3390/cancers13153778] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/16/2021] [Accepted: 07/21/2021] [Indexed: 11/29/2022] Open
Abstract
Simple Summary BRF2, a subunit of the RNA polymerase III transcription complex, is upregulated in a wide variety of cancers and is a potential therapeutic target; however, no effective drugs are available to target BRF2. The upregulation of BRF2 in cancer cells confers survival via the prevention of oxidative stress-induced apoptosis. In this manuscript, we report the identification of potential BRF2 inhibitors through in silico drug repurposing screening. We further characterized bexarotene as a hit compound for the development of selective BRF2 inhibitors and provide experimental validation to support the repurposing of this FDA-approved drug as an agent to reduce the cellular levels of ROS and consequent BRF2 expression in cancers with elevated levels of oxidative stress. Abstract The overexpression of BRF2, a selective subunit of RNA polymerase III, has been shown to be crucial in the development of several types of cancers, including breast cancer and lung squamous cell carcinoma. Predominantly, BRF2 acts as a central redox-sensing transcription factor (TF) and is involved in rescuing oxidative stress (OS)-induced apoptosis. Here, we showed a novel link between BRF2 and the DNA damage response. Due to the lack of BRF2-specific inhibitors, through virtual screening and molecular dynamics simulation, we identified potential drug candidates that interfere with BRF2-TATA-binding Protein (TBP)-DNA complex interactions based on binding energy, intermolecular, and torsional energy parameters. We experimentally tested bexarotene as a potential BRF2 inhibitor. We found that bexarotene (Bex) treatment resulted in a dramatic decline in oxidative stress and Tert-butylhydroquinone (tBHQ)-induced levels of BRF2 and consequently led to a decrease in the cellular proliferation of cancer cells which may in part be due to the drug pretreatment-induced reduction of ROS generated by the oxidizing agent. Our data thus provide the first experimental evidence that BRF2 is a novel player in the DNA damage response pathway and that bexarotene can be used as a potential inhibitor to treat cancers with the specific elevation of oxidative stress.
Collapse
Affiliation(s)
- Behnam Rashidieh
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (S.M.T.); (H.T.); (S.S.); (R.C.A.); (M.K.)
- Correspondence: (B.R.); (K.K.K.)
| | - Maryam Molakarimi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University (TMU), Nasr Bridge, Tehran 14115-154, Iran; (M.M.); (A.M.)
| | - Ammar Mohseni
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University (TMU), Nasr Bridge, Tehran 14115-154, Iran; (M.M.); (A.M.)
| | - Simon Manuel Tria
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (S.M.T.); (H.T.); (S.S.); (R.C.A.); (M.K.)
- School of Environment and Science, Griffith University, Nathan, QLD 4111, Australia
| | - Hein Truong
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (S.M.T.); (H.T.); (S.S.); (R.C.A.); (M.K.)
| | - Sriganesh Srihari
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (S.M.T.); (H.T.); (S.S.); (R.C.A.); (M.K.)
| | - Rachael C. Adams
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (S.M.T.); (H.T.); (S.S.); (R.C.A.); (M.K.)
| | - Mathew Jones
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4102, Australia;
| | - Pascal H. G. Duijf
- Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia;
- Centre for Data Science, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
| | - Murugan Kalimutho
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (S.M.T.); (H.T.); (S.S.); (R.C.A.); (M.K.)
| | - Kum Kum Khanna
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (S.M.T.); (H.T.); (S.S.); (R.C.A.); (M.K.)
- Correspondence: (B.R.); (K.K.K.)
| |
Collapse
|
11
|
Zhang Y, Zhang H, Wang M, Schmid T, Xin Z, Kozhuharova L, Yu WK, Huang Y, Cai F, Biskup E. Hypoxia in Breast Cancer-Scientific Translation to Therapeutic and Diagnostic Clinical Applications. Front Oncol 2021; 11:652266. [PMID: 33777815 PMCID: PMC7991906 DOI: 10.3389/fonc.2021.652266] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 02/12/2021] [Indexed: 12/13/2022] Open
Abstract
Breast cancer has been the leading cause of female cancer deaths for decades. Intratumoral hypoxia, mainly caused by structural and functional abnormalities in microvasculature, is often associated with a more aggressive phenotype, increased risk of metastasis and resistance to anti-malignancy treatments. The response of cancer cells to hypoxia is ascribed to hypoxia-inducible factors (HIFs) that activate the transcription of a large battery of genes encoding proteins promoting primary tumor vascularization and growth, stromal cell recruitment, extracellular matrix remodeling, cell motility, local tissue invasion, metastasis, and maintenance of the cancer stem cell properties. In this review, we summarized the role of hypoxia specifically in breast cancer, discuss the prognostic and predictive value of hypoxia factors, potential links of hypoxia and endocrine resistance, cancer hypoxia measurements, further involved mechanisms, clinical application of hypoxia-related treatments and open questions.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hongyi Zhang
- Department of Breast Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Minghong Wang
- Department of Health Management, Shanghai Public Health Clinical Center, Shanghai, China
| | - Thomas Schmid
- Department of Medical Oncology, St. Claraspital, Basel, Switzerland
| | - Zhaochen Xin
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | | | - Wai-Kin Yu
- Cellomics International Limited, Hong Kong, China
| | - Yuan Huang
- Cellomics International Limited, Hong Kong, China
| | - Fengfeng Cai
- Department of Breast Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ewelina Biskup
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Division of Internal Medicine, University Hospital of Basel, University of Basel, Basel, Switzerland.,Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
| |
Collapse
|
12
|
Fan T, Feng X, Yokota A, Liu W, Tang Y, Yan X, Xiao H, Wang Y, Deng Z, Zhao P, Wang M, Wang H, Ma R, Hu X, Huang G. Arsenic Dispensing Powder Promotes Erythropoiesis in Myelodysplastic Syndromes via Downregulation of HIF1A and Upregulation of GATA Factors. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2021; 49:461-485. [PMID: 33641653 DOI: 10.1142/s0192415x2150021x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Traditional Chinese Medicine (TCM) is a practical medicine based on thousands of years of medical practice in China. Arsenic dispensing powder (ADP) has been used as a treatment for MDS patients with a superior efficacy on anemia at Xiyuan Hospital of China Academy of Chinese Medical Sciences. In this study, we retrospectively analyzed MDS patients that received ADP treatment in the past 9 years and confirmed that ADP improves patients' anemia and prolongs overall survival in intermediate-risk MDS patients. Then, we used the MDS transgenic mice model and cell line to explore the drug mechanism. In normal and MDS cells, ADP does not show cellular toxicity but promotes differentiation. In mouse MDS models, we observed that ADP showed significant efficacy on promoting erythropoiesis. In the BFU-E and CFU-E assays, ADP could promote erythropoiesis not only in normal clones but also in MDS clones. Mechanistically, we found that ADP could downregulate HIF1A in MDS clones through upregulation of VHL, P53 and MDM2, which is involved in two parallel pathways to downregulate HIF1A. We also confirmed that ADP upregulates GATA factors in normal clones. Thus, our clinical and experimental studies indicate that ADP is a promising drug to promote erythropoiesis in both MDS and normal clones with a superior outcome than current regular therapies. ADP promotes erythropoiesis in myelodysplastic syndromes via downregulation of HIF1A and upregulation of GATA factors.
Collapse
Affiliation(s)
- Teng Fan
- Department of Hematology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, P. R. China.,Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, OH, USA
| | - Xiaomin Feng
- Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, OH, USA
| | - Asumi Yokota
- Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, OH, USA
| | - Weiyi Liu
- Department of Hematology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, P. R. China
| | - Yuting Tang
- Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, OH, USA
| | - Xiaomei Yan
- Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, OH, USA
| | - Haiyan Xiao
- Department of Hematology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, P. R. China
| | - Yue Wang
- Department of Hematology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, P. R. China
| | - Zhongyang Deng
- Department of Hematology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, P. R. China
| | - Pan Zhao
- Department of Hematology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, P. R. China
| | - Mingjing Wang
- Department of Hematology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, P. R. China
| | - Hongzhi Wang
- Department of Hematology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, P. R. China
| | - Rou Ma
- Department of Hematology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, P. R. China
| | - Xiaomei Hu
- Department of Hematology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, P. R. China
| | - Gang Huang
- Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, OH, USA
| |
Collapse
|
13
|
Gray M, Meehan J, Turnbull AK, Martínez-Pérez C, Kay C, Pang LY, Argyle DJ. The Importance of the Tumor Microenvironment and Hypoxia in Delivering a Precision Medicine Approach to Veterinary Oncology. Front Vet Sci 2020; 7:598338. [PMID: 33282935 PMCID: PMC7688625 DOI: 10.3389/fvets.2020.598338] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 10/12/2020] [Indexed: 11/26/2022] Open
Abstract
Treating individual patients on the basis of specific factors, such as biomarkers, molecular signatures, phenotypes, environment, and lifestyle is what differentiates the precision medicine initiative from standard treatment regimens. Although precision medicine can be applied to almost any branch of medicine, it is perhaps most easily applied to the field of oncology. Cancer is a heterogeneous disease, meaning that even though patients may be histologically diagnosed with the same cancer type, their tumors may have different molecular characteristics, genetic mutations or tumor microenvironments that can influence prognosis or treatment response. In this review, we describe what methods are currently available to clinicians that allow them to monitor key tumor microenvironmental parameters in a way that could be used to achieve precision medicine for cancer patients. We further describe exciting novel research involving the use of implantable medical devices for precision medicine, including those developed for mapping tumor microenvironment parameters (e.g., O2, pH, and cancer biomarkers), delivering local drug treatments, assessing treatment responses, and monitoring for recurrence and metastasis. Although these research studies have predominantly focused on and were tailored to humans, the results and concepts are equally applicable to veterinary patients. While veterinary clinical studies that have adopted a precision medicine approach are still in their infancy, there have been some exciting success stories. These have included the development of a receptor tyrosine kinase inhibitor for canine mast cell tumors and the production of a PCR assay to monitor the chemotherapeutic response of canine high-grade B-cell lymphomas. Although precision medicine is an exciting area of research, it currently has failed to gain significant translation into human and veterinary healthcare practices. In order to begin to address this issue, there is increasing awareness that cross-disciplinary approaches involving human and veterinary clinicians, engineers and chemists may be needed to help advance precision medicine toward its full integration into human and veterinary clinical practices.
Collapse
Affiliation(s)
- Mark Gray
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Scotland, United Kingdom
| | - James Meehan
- Translational Oncology Research Group, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Scotland, United Kingdom
| | - Arran K. Turnbull
- Translational Oncology Research Group, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Scotland, United Kingdom
- Breast Cancer Now Edinburgh Research Team, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Scotland, United Kingdom
| | - Carlos Martínez-Pérez
- Translational Oncology Research Group, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Scotland, United Kingdom
- Breast Cancer Now Edinburgh Research Team, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Scotland, United Kingdom
| | - Charlene Kay
- Translational Oncology Research Group, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Scotland, United Kingdom
- Breast Cancer Now Edinburgh Research Team, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Scotland, United Kingdom
| | - Lisa Y. Pang
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Scotland, United Kingdom
| | - David J. Argyle
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Scotland, United Kingdom
| |
Collapse
|
14
|
Sørensen BS, Horsman MR. Tumor Hypoxia: Impact on Radiation Therapy and Molecular Pathways. Front Oncol 2020; 10:562. [PMID: 32373534 PMCID: PMC7186437 DOI: 10.3389/fonc.2020.00562] [Citation(s) in RCA: 149] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/30/2020] [Indexed: 01/25/2023] Open
Abstract
Tumor hypoxia is a common feature of the microenvironment in solid tumors, primarily due to an inadequate, and heterogeneous vascular network. It is associated with resistance to radiotherapy and results in a poorer clinical outcome. The presence of hypoxia in tumors can be identified by various invasive and non-invasive techniques, and there are a number of approaches by which hypoxia can be modified to improve outcome. However, despite these factors and the ongoing extensive pre-clinical studies, the clinical focus on hypoxia is still to a large extent lacking. Hypoxia is a major cellular stress factor and affects a wide range of molecular pathways, and further understanding of the molecular processes involved may lead to greater clinical applicability of hypoxic modifiers. This review is a discussion of the characteristics of tumor hypoxia, hypoxia-related molecular pathways, and the role of hypoxia in treatment resistance. Understanding the molecular aspects of hypoxia will improve our ability to clinically monitor hypoxia and to predict and modify the therapeutic response.
Collapse
Affiliation(s)
- Brita Singers Sørensen
- Experimental Clinical Oncology-Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Michael R Horsman
- Experimental Clinical Oncology-Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
15
|
Soltanalizadeh B, Gonzalez Rodriguez E, Maroufy V, Zheng WJ, Wu H. Modelling of hypoxia gene expression for three different cancer cell lines. ACTA ACUST UNITED AC 2020; 13:124-143. [PMID: 32153660 DOI: 10.1504/ijcbdd.2020.10026794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Gene dynamic analysis is essential in identifying target genes involved pathogenesis of various diseases, including cancer. Cancer prognosis is often influenced by hypoxia. We apply a multi-step pipeline to study dynamic gene expressions in response to hypoxia in three cancer cell lines: prostate (DU145), colon (HT29), and breast (MCF7) cancers. We identified 26 distinct temporal expression patterns for prostate cell line, and 29 patterns for colon and breast cell lines. The module-based dynamic networks have been developed for all three cell lines. Our analyses improve the existing results in multiple ways. It exploits the time-dependence nature of gene expression values in identifying the dynamically significant genes; hence, more key significant genes and transcription factors have been identified. Our gene network returns significant information regarding biologically important modules of genes. Furthermore, the network has potential in learning the regulatory path between transcription factors and the downstream genes. In addition, our findings suggest that changes in genes BMP6 and ARSJ expression might have a key role in the time-dependent response to hypoxia in breast cancer.
Collapse
Affiliation(s)
- Babak Soltanalizadeh
- Department of Biostatistics & Data Science, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Erika Gonzalez Rodriguez
- Center for translational Injury Research, Department of Surgery, McGovern Medical School, UT Houston, Houston, TX, USA
| | - Vahed Maroufy
- Department of Biostatistics & Data Science, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - W Jim Zheng
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Hulin Wu
- Department of Biostatistics & Data Science, University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
16
|
van der Heijden M, Essers PBM, de Jong MC, de Roest RH, Sanduleanu S, Verhagen CVM, Hamming-Vrieze O, Hoebers F, Lambin P, Bartelink H, Leemans CR, Verheij M, Brakenhoff RH, van den Brekel MWM, Vens C. Biological Determinants of Chemo-Radiotherapy Response in HPV-Negative Head and Neck Cancer: A Multicentric External Validation. Front Oncol 2020; 9:1470. [PMID: 31998639 PMCID: PMC6966332 DOI: 10.3389/fonc.2019.01470] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 12/09/2019] [Indexed: 12/20/2022] Open
Abstract
Purpose: Tumor markers that are related to hypoxia, proliferation, DNA damage repair and stem cell-ness, have a prognostic value in advanced stage HNSCC patients when assessed individually. Here we aimed to evaluate and validate this in a multifactorial context and assess interrelation and the combined role of these biological factors in determining chemo-radiotherapy response in HPV-negative advanced HNSCC. Methods: RNA sequencing data of pre-treatment biopsy material from 197 HPV-negative advanced stage HNSCC patients treated with definitive chemoradiotherapy was analyzed. Biological parameter scores were assigned to patient samples using previously generated and described gene expression signatures. Locoregional control rates were used to assess the role of these biological parameters in radiation response and compared to distant metastasis data. Biological factors were ranked according to their clinical impact using bootstrapping methods and multivariate Cox regression analyses that included clinical variables. Multivariate Cox regression analyses comprising all biological variables were used to define their relative role among all factors when combined. Results: Only few biomarker scores correlate with each other, underscoring their independence. The different biological factors do not correlate or cluster, except for the two stem cell markers CD44 and SLC3A2 (r = 0.4, p < 0.001) and acute hypoxia prediction scores which correlated with T-cell infiltration score, CD8+ T cell abundance and proliferation scores (r = 0.52, 0.56, and 0.6, respectively with p < 0.001). Locoregional control association analyses revealed that chronic (Hazard Ratio (HR) = 3.9) and acute hypoxia (HR = 1.9), followed by stem cell-ness (CD44/SLC3A2; HR = 2.2/2.3), were the strongest and most robust determinants of radiation response. Furthermore, multivariable analysis, considering other biological and clinical factors, reveal a significant role for EGFR expression (HR = 2.9, p < 0.05) and T-cell infiltration (CD8+T-cells: HR = 2.2, p < 0.05; CD8+T-cells/Treg: HR = 2.6, p < 0.01) signatures in locoregional control of chemoradiotherapy-treated HNSCC. Conclusion: Tumor acute and chronic hypoxia, stem cell-ness, and CD8+ T-cell parameters are relevant and largely independent biological factors that together contribute to locoregional control. The combined analyses illustrate the additive value of multifactorial analyses and support a role for EGFR expression analysis and immune cell markers in addition to previously validated biomarkers. This external validation underscores the relevance of biological factors in determining chemoradiotherapy outcome in HNSCC.
Collapse
Affiliation(s)
- Martijn van der Heijden
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, Netherlands.,Department of Head and Neck Oncology and Surgery, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Paul B M Essers
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, Netherlands.,Department of Radiation Oncology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Monique C de Jong
- Department of Radiation Oncology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Reinout H de Roest
- Amsterdam UMC, Vrije Universiteit Amsterdam, Otolaryngology/Head and Neck Surgery, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Sebastian Sanduleanu
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Caroline V M Verhagen
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, Netherlands.,Department of Head and Neck Oncology and Surgery, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Olga Hamming-Vrieze
- Department of Head and Neck Oncology and Surgery, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Frank Hoebers
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Philippe Lambin
- The D-Lab and The M-Lab, Department of Precision Medicine, GROW-School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
| | - Harry Bartelink
- Department of Radiation Oncology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - C René Leemans
- Amsterdam UMC, Vrije Universiteit Amsterdam, Otolaryngology/Head and Neck Surgery, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Marcel Verheij
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, Netherlands.,Department of Radiation Oncology, The Netherlands Cancer Institute, Amsterdam, Netherlands.,Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Ruud H Brakenhoff
- Amsterdam UMC, Vrije Universiteit Amsterdam, Otolaryngology/Head and Neck Surgery, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Michiel W M van den Brekel
- Department of Head and Neck Oncology and Surgery, The Netherlands Cancer Institute, Amsterdam, Netherlands.,Department of Oral and Maxillofacial Surgery, Amsterdam UMC, Academic Medical Center, Amsterdam, Netherlands
| | - Conchita Vens
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, Netherlands.,Department of Radiation Oncology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| |
Collapse
|
17
|
Abou Khouzam R, Goutham HV, Zaarour RF, Chamseddine AN, Francis A, Buart S, Terry S, Chouaib S. Integrating tumor hypoxic stress in novel and more adaptable strategies for cancer immunotherapy. Semin Cancer Biol 2020; 65:140-154. [PMID: 31927131 DOI: 10.1016/j.semcancer.2020.01.003] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/10/2019] [Accepted: 01/07/2020] [Indexed: 12/14/2022]
Abstract
Immunotherapy is poised to become an increasingly utilized therapy in the treatment of cancer. However, several abnormalities in the tumor microenvironment (TME) that can thwart the efficacy of immunotherapies have been established. Microenvironmental hypoxia is a determining factor in shaping aggressiveness, metastatic potential and treatment resistance of solid tumors. The characterization of this phenomenon could prove beneficial for determining a patient's treatment path and for the introduction of novel targetable factors that can enhance therapeutic outcome. Indeed, the ablation of hypoxia has the potential to sensitize tumors to immunotherapy by metabolically remodeling their microenvironment. In this review, we discuss the intrinsic contributions of hypoxia to cellular plasticity, heterogeneity, stemness and genetic instability in the context of immune escape. In addition, we will shed light on how managing hypoxia can ameliorate response to immunotherapy and how integrating hypoxia gene signatures could play a role in this pursuit.
Collapse
Affiliation(s)
- Raefa Abou Khouzam
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, 4184, United Arab Emirates.
| | - Hassan Venkatesh Goutham
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, 4184, United Arab Emirates.
| | - Rania Faouzi Zaarour
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, 4184, United Arab Emirates.
| | - Ali N Chamseddine
- Département d'Oncologie Médicale, Gustave Roussy Cancer Campus Grand Paris, Villejuif, France.
| | - Amirtharaj Francis
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, 4184, United Arab Emirates.
| | - Stéphanie Buart
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, EPHE, Faculty. De médecine Univ. Paris-Sud, University Paris-Saclay, Villejuif F-94805, France
| | - Stéphane Terry
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, EPHE, Faculty. De médecine Univ. Paris-Sud, University Paris-Saclay, Villejuif F-94805, France.
| | - Salem Chouaib
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, 4184, United Arab Emirates; INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, EPHE, Faculty. De médecine Univ. Paris-Sud, University Paris-Saclay, Villejuif F-94805, France.
| |
Collapse
|
18
|
Guarischi-Sousa R, Monteiro JS, Alecrim LC, Michaloski JS, Cardeal LB, Ferreira EN, Carraro DM, Nunes DN, Dias-Neto E, Reimand J, Boutros PC, Setubal JC, Giordano RJ. A transcriptome-based signature of pathological angiogenesis predicts breast cancer patient survival. PLoS Genet 2019; 15:e1008482. [PMID: 31846472 PMCID: PMC6917213 DOI: 10.1371/journal.pgen.1008482] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 10/15/2019] [Indexed: 12/30/2022] Open
Abstract
The specific genes and molecules that drive physiological angiogenesis differ from those involved in pathological angiogenesis, suggesting distinct mechanisms for these seemingly related processes. Unveiling genes and pathways preferentially associated with pathologic angiogenesis is key to understanding its mechanisms, thereby facilitating development of novel approaches to managing angiogenesis-dependent diseases. To better understand these different processes, we elucidated the transcriptome of the mouse retina in the well-accepted oxygen-induced retinopathy (OIR) model of pathological angiogenesis. We identified 153 genes changed between normal and OIR retinas, which represent a molecular signature relevant to other angiogenesis-dependent processes such as cancer. These genes robustly predict the survival of breast cancer patients, which was validated in an independent 1,000-patient test cohort (40% difference in 15-year survival; p = 2.56 x 10-21). These results suggest that the OIR model reveals key genes involved in pathological angiogenesis, and these may find important applications in stratifying tumors for treatment intensification or for angiogenesis-targeted therapies.
Collapse
Affiliation(s)
- Rodrigo Guarischi-Sousa
- Biochemistry Department, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Jhonatas S. Monteiro
- Biochemistry Department, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Lilian C. Alecrim
- Biochemistry Department, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Jussara S. Michaloski
- Biochemistry Department, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Laura B. Cardeal
- Biochemistry Department, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Elisa N. Ferreira
- International Research Center (CIPE) A.C. Camargo Cancer Center, São Paulo, SP, Brazil
| | - Dirce M. Carraro
- International Research Center (CIPE) A.C. Camargo Cancer Center, São Paulo, SP, Brazil
| | - Diana N. Nunes
- International Research Center (CIPE) A.C. Camargo Cancer Center, São Paulo, SP, Brazil
| | - Emmanuel Dias-Neto
- International Research Center (CIPE) A.C. Camargo Cancer Center, São Paulo, SP, Brazil
- Laboratory of Neurosciences (LIM27), Institute & Department of Psychiatry, University of São Paulo, São Paulo, Brazil
| | - Jüri Reimand
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Paul C. Boutros
- Department of Human Genetics, University of California Los Angeles (UCLA), Los Angeles, CA, United States of America
- * E-mail: (PCB); (JCS); (RJG)
| | - João C. Setubal
- Biochemistry Department, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
- * E-mail: (PCB); (JCS); (RJG)
| | - Ricardo J. Giordano
- Biochemistry Department, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
- * E-mail: (PCB); (JCS); (RJG)
| |
Collapse
|
19
|
van der Heijden M, de Jong MC, Verhagen CVM, de Roest RH, Sanduleanu S, Hoebers F, Leemans CR, Brakenhoff RH, Vens C, Verheij M, van den Brekel MWM. Acute Hypoxia Profile is a Stronger Prognostic Factor than Chronic Hypoxia in Advanced Stage Head and Neck Cancer Patients. Cancers (Basel) 2019; 11:E583. [PMID: 31027242 PMCID: PMC6520712 DOI: 10.3390/cancers11040583] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 04/11/2019] [Accepted: 04/19/2019] [Indexed: 02/01/2023] Open
Abstract
Hypoxic head and neck tumors respond poorly to radiotherapy and can be identified using gene expression profiles. However, it is unknown whether treatment outcome is driven by acute or chronic hypoxia. Gene expression data of 398 head and neck cancers was collected. Four clinical hypoxia profiles were compared to in vitro acute and chronic hypoxia profiles. Chronic and acute hypoxia profiles were tested for their association to outcome using Cox proportional hazard analyses. In an initial set of 224 patients, scores of the four clinical hypoxia profiles correlated with each other and with chronic hypoxia. However, the acute hypoxia profile showed a stronger association with local recurrence after chemoradiotherapy (p = 0.02; HR = 3.1) than the four clinical (chronic hypoxia) profiles (p = 0.2; HR = 0.9). An independent set of 174 patients confirmed that acute hypoxia is a stronger prognostic factor than chronic hypoxia for overall survival, progression-free survival, local and locoregional control. Multivariable analyses accounting for known prognostic factors substantiate this finding (p = 0.045; p = 0.042; p = 0.018 and p = 0.003, respectively). In conclusion, the four clinical hypoxia profiles are related to chronic hypoxia and not acute hypoxia. The acute hypoxia profile shows a stronger association with patient outcome and should be incorporated into existing prediction models.
Collapse
Affiliation(s)
- Martijn van der Heijden
- Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1006 CX Amsterdam, The Netherlands.
- Department of Head and Neck Oncology and Surgery, The Netherlands Cancer Institute, Plesmanlaan 121, 1006 CX Amsterdam, The Netherlands.
| | - Monique C de Jong
- Department of Radiation Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, 1006 CX Amsterdam, The Netherlands.
| | - Caroline V M Verhagen
- Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1006 CX Amsterdam, The Netherlands.
- Department of Head and Neck Oncology and Surgery, The Netherlands Cancer Institute, Plesmanlaan 121, 1006 CX Amsterdam, The Netherlands.
| | - Reinout H de Roest
- Amsterdam UMC, Vrije Universiteit Amsterdam, Otolaryngology/Head & Neck Surgery, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.
| | - Sebastian Sanduleanu
- Department of Radiation Oncology (MAASTRO), GROW ⁻ School for Oncology and Developmental Biology, Maastricht University Medical Centre, Dr. Tanslaan 12, 6229 ET Maastricht, The Netherlands.
| | - Frank Hoebers
- Department of Radiation Oncology (MAASTRO), GROW ⁻ School for Oncology and Developmental Biology, Maastricht University Medical Centre, Dr. Tanslaan 12, 6229 ET Maastricht, The Netherlands.
| | - C René Leemans
- Amsterdam UMC, Vrije Universiteit Amsterdam, Otolaryngology/Head & Neck Surgery, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.
| | - Ruud H Brakenhoff
- Amsterdam UMC, Vrije Universiteit Amsterdam, Otolaryngology/Head & Neck Surgery, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.
| | - Conchita Vens
- Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1006 CX Amsterdam, The Netherlands.
- Department of Radiation Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, 1006 CX Amsterdam, The Netherlands.
| | - Marcel Verheij
- Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1006 CX Amsterdam, The Netherlands.
- Department of Radiation Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, 1006 CX Amsterdam, The Netherlands.
- Department of Radiation Oncology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands.
| | - Michiel W M van den Brekel
- Department of Head and Neck Oncology and Surgery, The Netherlands Cancer Institute, Plesmanlaan 121, 1006 CX Amsterdam, The Netherlands.
- Department of Oral and Maxillofacial Surgery, Amsterdam UMC, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands.
| |
Collapse
|
20
|
Bhandari V, Hoey C, Liu LY, Lalonde E, Ray J, Livingstone J, Lesurf R, Shiah YJ, Vujcic T, Huang X, Espiritu SMG, Heisler LE, Yousif F, Huang V, Yamaguchi TN, Yao CQ, Sabelnykova VY, Fraser M, Chua MLK, van der Kwast T, Liu SK, Boutros PC, Bristow RG. Molecular landmarks of tumor hypoxia across cancer types. Nat Genet 2019; 51:308-318. [PMID: 30643250 DOI: 10.1038/s41588-018-0318-2] [Citation(s) in RCA: 446] [Impact Index Per Article: 74.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 11/20/2018] [Indexed: 12/27/2022]
Abstract
Many primary-tumor subregions have low levels of molecular oxygen, termed hypoxia. Hypoxic tumors are at elevated risk for local failure and distant metastasis, but the molecular hallmarks of tumor hypoxia remain poorly defined. To fill this gap, we quantified hypoxia in 8,006 tumors across 19 tumor types. In ten tumor types, hypoxia was associated with elevated genomic instability. In all 19 tumor types, hypoxic tumors exhibited characteristic driver-mutation signatures. We observed widespread hypoxia-associated dysregulation of microRNAs (miRNAs) across cancers and functionally validated miR-133a-3p as a hypoxia-modulated miRNA. In localized prostate cancer, hypoxia was associated with elevated rates of chromothripsis, allelic loss of PTEN and shorter telomeres. These associations are particularly enriched in polyclonal tumors, representing a constellation of features resembling tumor nimbosus, an aggressive cellular phenotype. Overall, this work establishes that tumor hypoxia may drive aggressive molecular features across cancers and shape the clinical trajectory of individual tumors.
Collapse
Affiliation(s)
- Vinayak Bhandari
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Christianne Hoey
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Lydia Y Liu
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Emilie Lalonde
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Jessica Ray
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Julie Livingstone
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Robert Lesurf
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Yu-Jia Shiah
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Tina Vujcic
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Xiaoyong Huang
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Shadrielle M G Espiritu
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Lawrence E Heisler
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Fouad Yousif
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Vincent Huang
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Takafumi N Yamaguchi
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Cindy Q Yao
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Veronica Y Sabelnykova
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Michael Fraser
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Melvin L K Chua
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore.,Duke-NUS Graduate Medical School, Singapore, Singapore
| | | | - Stanley K Liu
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Paul C Boutros
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada. .,Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada. .,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada. .,Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA. .,Department of Urology, University of California, Los Angeles, Los Angeles, CA, USA. .,Jonsson Comprehensive Cancer Centre, University of California, Los Angeles, Los Angeles, CA, USA. .,Institute for Precision Health, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Robert G Bristow
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada. .,Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada. .,Princess Margaret Hospital, University Health Network, Toronto, Ontario, Canada. .,Division of Cancer Sciences, Faculty of Biology, Health and Medicine, University of Manchester, Manchester, UK. .,The Christie NHS Foundation Trust, Manchester, UK. .,CRUK Manchester Institute and Manchester Cancer Research Centre, Manchester, UK.
| |
Collapse
|
21
|
Quantification of hypoxia-related gene expression as a potential approach for clinical outcome prediction in breast cancer. PLoS One 2017; 12:e0175960. [PMID: 28430808 PMCID: PMC5400273 DOI: 10.1371/journal.pone.0175960] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 04/03/2017] [Indexed: 01/08/2023] Open
Abstract
Breast cancers are solid tumors frequently characterized by regions with low oxygen concentrations. Cellular adaptations to hypoxia are mainly determined by “hypoxia inducible factors” that mediate transcriptional modifications involved in drug resistance and tumor progression leading to metastasis and relapse occurrence. In this study, we investigated the prognostic value of hypoxia-related gene expression in breast cancer. A systematic review was conducted to select a set of 45 genes involved in hypoxia signaling pathways and breast tumor progression. Gene expression was quantified by RT-qPCR in a retrospective series of 32 patients with invasive ductal carcinoma. Data were analyzed in relation to classical clinicopathological criteria and relapse occurrence. Coordinated overexpression of selected genes was observed in high-grade and HER2+ tumors. Hierarchical cluster analysis of gene expression significantly segregated relapsed patients (p = 0.008, Chi2 test). All genes (except one) were up-regulated and six markers were significantly expressed in tumors from recurrent patients. The expression of this 6-gene set was used to develop a basic algorithm for identifying recurrent patients according to a risk score of relapse. Analysis of Kaplan-Meier relapse-free survival curves allowed the definition of a threshold score of 2 (p = 0.021, Mantel-Haenszel test). The risk of recurrence was increased by 40% in patients with a high score. In addition to classical prognostic factors, we showed that hypoxic markers have potential prognostic value for outcome and late recurrence prediction, leading to improved treatment decision-making for patients with early-stage invasive breast cancer. It will be necessary to validate the clinical relevance of this prognostic approach through independent studies including larger prospective patient cohorts.
Collapse
|
22
|
Lambin P, Zindler J, Vanneste B, van de Voorde L, Jacobs M, Eekers D, Peerlings J, Reymen B, Larue RTHM, Deist TM, de Jong EEC, Even AJG, Berlanga AJ, Roelofs E, Cheng Q, Carvalho S, Leijenaar RTH, Zegers CML, van Limbergen E, Berbee M, van Elmpt W, Oberije C, Houben R, Dekker A, Boersma L, Verhaegen F, Bosmans G, Hoebers F, Smits K, Walsh S. Modern clinical research: How rapid learning health care and cohort multiple randomised clinical trials complement traditional evidence based medicine. Acta Oncol 2015; 54:1289-300. [PMID: 26395528 DOI: 10.3109/0284186x.2015.1062136] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Trials are vital in informing routine clinical care; however, current designs have major deficiencies. An overview of the various challenges that face modern clinical research and the methods that can be exploited to solve these challenges, in the context of personalised cancer treatment in the 21st century is provided. AIM The purpose of this manuscript, without intending to be comprehensive, is to spark thought whilst presenting and discussing two important and complementary alternatives to traditional evidence-based medicine, specifically rapid learning health care and cohort multiple randomised controlled trial design. Rapid learning health care is an approach that proposes to extract and apply knowledge from routine clinical care data rather than exclusively depending on clinical trial evidence, (please watch the animation: http://youtu.be/ZDJFOxpwqEA). The cohort multiple randomised controlled trial design is a pragmatic method which has been proposed to help overcome the weaknesses of conventional randomised trials, taking advantage of the standardised follow-up approaches more and more used in routine patient care. This approach is particularly useful when the new intervention is a priori attractive for the patient (i.e. proton therapy, patient decision aids or expensive medications), when the outcomes are easily collected, and when there is no need of a placebo arm. DISCUSSION Truly personalised cancer treatment is the goal in modern radiotherapy. However, personalised cancer treatment is also an immense challenge. The vast variety of both cancer patients and treatment options makes it extremely difficult to determine which decisions are optimal for the individual patient. Nevertheless, rapid learning health care and cohort multiple randomised controlled trial design are two approaches (among others) that can help meet this challenge.
Collapse
Affiliation(s)
- Philippe Lambin
- a Department of Radiation Oncology (MAASTRO) , GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Jaap Zindler
- a Department of Radiation Oncology (MAASTRO) , GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Ben Vanneste
- a Department of Radiation Oncology (MAASTRO) , GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Lien van de Voorde
- a Department of Radiation Oncology (MAASTRO) , GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Maria Jacobs
- a Department of Radiation Oncology (MAASTRO) , GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Daniëlle Eekers
- a Department of Radiation Oncology (MAASTRO) , GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Jurgen Peerlings
- a Department of Radiation Oncology (MAASTRO) , GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Bart Reymen
- a Department of Radiation Oncology (MAASTRO) , GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Ruben T H M Larue
- a Department of Radiation Oncology (MAASTRO) , GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Timo M Deist
- a Department of Radiation Oncology (MAASTRO) , GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Evelyn E C de Jong
- a Department of Radiation Oncology (MAASTRO) , GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Aniek J G Even
- a Department of Radiation Oncology (MAASTRO) , GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Adriana J Berlanga
- a Department of Radiation Oncology (MAASTRO) , GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Erik Roelofs
- a Department of Radiation Oncology (MAASTRO) , GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Qing Cheng
- a Department of Radiation Oncology (MAASTRO) , GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Sara Carvalho
- a Department of Radiation Oncology (MAASTRO) , GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Ralph T H Leijenaar
- a Department of Radiation Oncology (MAASTRO) , GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Catharina M L Zegers
- a Department of Radiation Oncology (MAASTRO) , GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Evert van Limbergen
- a Department of Radiation Oncology (MAASTRO) , GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Maaike Berbee
- a Department of Radiation Oncology (MAASTRO) , GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Wouter van Elmpt
- a Department of Radiation Oncology (MAASTRO) , GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Cary Oberije
- a Department of Radiation Oncology (MAASTRO) , GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Ruud Houben
- a Department of Radiation Oncology (MAASTRO) , GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Andre Dekker
- a Department of Radiation Oncology (MAASTRO) , GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Liesbeth Boersma
- a Department of Radiation Oncology (MAASTRO) , GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Frank Verhaegen
- a Department of Radiation Oncology (MAASTRO) , GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Geert Bosmans
- a Department of Radiation Oncology (MAASTRO) , GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Frank Hoebers
- a Department of Radiation Oncology (MAASTRO) , GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Kim Smits
- a Department of Radiation Oncology (MAASTRO) , GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Sean Walsh
- a Department of Radiation Oncology (MAASTRO) , GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre , Maastricht , The Netherlands
| |
Collapse
|
23
|
Harris BHL, Barberis A, West CML, Buffa FM. Gene Expression Signatures as Biomarkers of Tumour Hypoxia. Clin Oncol (R Coll Radiol) 2015; 27:547-60. [PMID: 26282471 DOI: 10.1016/j.clon.2015.07.004] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 06/16/2015] [Indexed: 02/08/2023]
Abstract
Hypoxia is a feature of most solid tumours and is associated with a poor prognosis. The hypoxic environment can reduce the efficacy of radiotherapy and some chemotherapeutics, and has been investigated extensively as a therapeutic target. The clinical use of hypoxia-targeting treatment will benefit from the development of a biomarker to assess tumour hypoxia. There are several possible techniques that measure either the level of oxygen or the tumour molecular response to hypoxia. The latter includes gene expression profiling, which measures the transcriptional response of a tumour to its hypoxic microenvironment. A systematic review identified 32 published hypoxia gene expression signatures. The methods used for their derivation varied, but are broadly classified as: (i) identifying genes with significantly higher or lower expression in cancer cells cultured under hypoxic versus normoxic conditions; (ii) using either previously characterised hypoxia-regulated genes/biomarkers to define hypoxic tumours and then identifying other genes that are over- or under-expressed in the hypoxic tumours. Both generated gene signatures useful in furthering our understanding of hypoxia biology. However, signatures derived using the second method seem to be superior in terms of providing prognostic information. Here we summarise all 32 published hypoxia signatures, discuss their commonalities and differences, and highlight their strengths and limitations. This review also highlights the importance of reproducibility and gene annotation, which must be accounted for to transfer signatures robustly for clinical application as biomarkers.
Collapse
Affiliation(s)
- B H L Harris
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - A Barberis
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - C M L West
- Translational Radiobiology Group, Institute of Cancer Sciences, University of Manchester, Christie Hospital, Manchester M13 9PT, UK
| | - F M Buffa
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK.
| |
Collapse
|
24
|
Boidot R, Branders S, Helleputte T, Rubio LI, Dupont P, Feron O. A generic cycling hypoxia-derived prognostic gene signature: application to breast cancer profiling. Oncotarget 2015; 5:6947-63. [PMID: 25216520 PMCID: PMC4196175 DOI: 10.18632/oncotarget.2285] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Temporal and local fluctuations in O2 in tumors require adaptive mechanisms to support cancer cell survival and proliferation. The transcriptome associated with cycling hypoxia (CycHyp) could thus represent a prognostic biomarker of cancer progression. Methods We exposed 20 tumor cell lines to repeated periods of hypoxia/reoxygenation to determine a transcriptomic CycHyp signature and used clinical data sets from 2,150 breast cancer patients to estimate a prognostic Cox proportional hazard model to assess its prognostic performance. Results The CycHyp prognostic potential was validated in patients independently of the receptor status of the tumors. The discriminating capacity of the CycHyp signature was further increased in the ER+ HER2- patient populations including those with a node negative status under treatment (HR=3.16) or not (HR=5.54). The CycHyp prognostic signature outperformed a signature derived from continuous hypoxia and major prognostic metagenes (P<0.001). The CycHyp signature could also identify ER+HER2 node-negative breast cancer patients at high risk based on clinicopathologic criteria but who could have been spared from chemotherapy and inversely those patients classified at low risk based but who presented a negative outcome. Conclusions The CycHyp signature is prognostic of breast cancer and offers a unique decision making tool to complement anatomopathologic evaluation.
Collapse
Affiliation(s)
- Romain Boidot
- Institut de Recherche Expérimentale et Clinique (IREC), Pole of Pharmacology and Therapeutics (FATH), Université catholique de Louvain, Brussels, Belgium. These authors contribued equally to this work
| | - Samuel Branders
- Machine Learning Group, Institute of Information and Communication Technologies, Electronics and Applied Mathematics (ICTEAM), Université catholique de Louvain, Louvain-la-Neuve, Belgium. These authors contribued equally to this work
| | - Thibault Helleputte
- Machine Learning Group, Institute of Information and Communication Technologies, Electronics and Applied Mathematics (ICTEAM), Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Laila Illan Rubio
- Institut de Recherche Expérimentale et Clinique (IREC), Pole of Pharmacology and Therapeutics (FATH), Université catholique de Louvain, Brussels, Belgium
| | - Pierre Dupont
- Machine Learning Group, Institute of Information and Communication Technologies, Electronics and Applied Mathematics (ICTEAM), Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Olivier Feron
- Institut de Recherche Expérimentale et Clinique (IREC), Pole of Pharmacology and Therapeutics (FATH), Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
25
|
Wong PP, Demircioglu F, Ghazaly E, Alrawashdeh W, Stratford MRL, Scudamore CL, Cereser B, Crnogorac-Jurcevic T, McDonald S, Elia G, Hagemann T, Kocher HM, Hodivala-Dilke KM. Dual-action combination therapy enhances angiogenesis while reducing tumor growth and spread. Cancer Cell 2015; 27:123-37. [PMID: 25584895 DOI: 10.1016/j.ccell.2014.10.015] [Citation(s) in RCA: 153] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 08/12/2014] [Accepted: 10/27/2014] [Indexed: 01/04/2023]
Abstract
Increasing chemotherapy delivery to tumors, while enhancing drug uptake and reducing side effects, is a primary goal of cancer research. In mouse and human cancer models in vivo, we show that coadministration of low-dose Cilengitide and Verapamil increases tumor angiogenesis, leakiness, blood flow, and Gemcitabine delivery. This approach reduces tumor growth, metastasis, and minimizes side effects while extending survival. At a molecular level, this strategy alters Gemcitabine transporter and metabolizing enzyme expression levels, enhancing the potency of Gemcitabine within tumor cells in vivo and in vitro. Thus, the dual action of low-dose Cilengitide, in vessels and tumor cells, improves chemotherapy efficacy. Overall, our data demonstrate that vascular promotion therapy is a means to improve cancer treatment.
Collapse
Affiliation(s)
- Ping-Pui Wong
- Centre for Tumor Biology, Barts Cancer Institute-a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Fevzi Demircioglu
- Centre for Tumor Biology, Barts Cancer Institute-a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Essam Ghazaly
- Centre for Haemato-Oncology, Barts Cancer Institute-a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Wasfi Alrawashdeh
- Centre for Molecular Oncology, Barts Cancer Institute-a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Michael R L Stratford
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Cheryl L Scudamore
- Mary Lyon Centre, MRC Harwell, Harwell Science and Innovation Campus, Oxfordshire OX11 0RD, UK
| | - Biancastella Cereser
- Centre for Tumor Biology, Barts Cancer Institute-a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Tatjana Crnogorac-Jurcevic
- Centre for Molecular Oncology, Barts Cancer Institute-a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Stuart McDonald
- Centre for Tumor Biology, Barts Cancer Institute-a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - George Elia
- Centre for Tumor Biology, Barts Cancer Institute-a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Thorsten Hagemann
- Centre for Cancer Inflammation, Barts Cancer Institute-a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK; Barts and the London HPB Centre, The Royal London Hospital, Barts Health NHS Trust, London E1 1BB, UK
| | - Hemant M Kocher
- Centre for Tumor Biology, Barts Cancer Institute-a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK; Barts and the London HPB Centre, The Royal London Hospital, Barts Health NHS Trust, London E1 1BB, UK
| | - Kairbaan M Hodivala-Dilke
- Centre for Tumor Biology, Barts Cancer Institute-a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
26
|
Fox NS, Starmans MHW, Haider S, Lambin P, Boutros PC. Ensemble analyses improve signatures of tumour hypoxia and reveal inter-platform differences. BMC Bioinformatics 2014; 15:170. [PMID: 24902696 PMCID: PMC4061774 DOI: 10.1186/1471-2105-15-170] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2013] [Accepted: 05/27/2014] [Indexed: 12/24/2022] Open
Abstract
Background The reproducibility of transcriptomic biomarkers across datasets remains poor, limiting clinical application. We and others have suggested that this is in-part caused by differential error-structure between datasets, and their incomplete removal by pre-processing algorithms. Methods To test this hypothesis, we systematically assessed the effects of pre-processing on biomarker classification using 24 different pre-processing methods and 15 distinct signatures of tumour hypoxia in 10 datasets (2,143 patients). Results We confirm strong pre-processing effects for all datasets and signatures, and find that these differ between microarray versions. Importantly, exploiting different pre-processing techniques in an ensemble technique improved classification for a majority of signatures. Conclusions Assessing biomarkers using an ensemble of pre-processing techniques shows clear value across multiple diseases, datasets and biomarkers. Importantly, ensemble classification improves biomarkers with initially good results but does not result in spuriously improved performance for poor biomarkers. While further research is required, this approach has the potential to become a standard for transcriptomic biomarkers.
Collapse
Affiliation(s)
| | | | | | | | - Paul C Boutros
- Informatics and Bio-computing Platform, Ontario Institute for Cancer Research, Toronto, Canada.
| |
Collapse
|
27
|
Smits KM, Melotte V, Niessen HE, Dubois L, Oberije C, Troost EG, Starmans MH, Boutros PC, Vooijs M, van Engeland M, Lambin P. Epigenetics in radiotherapy: Where are we heading? Radiother Oncol 2014; 111:168-77. [DOI: 10.1016/j.radonc.2014.05.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 03/17/2014] [Accepted: 05/01/2014] [Indexed: 12/20/2022]
|
28
|
'Rapid Learning health care in oncology' - an approach towards decision support systems enabling customised radiotherapy'. Radiother Oncol 2013; 109:159-64. [PMID: 23993399 DOI: 10.1016/j.radonc.2013.07.007] [Citation(s) in RCA: 142] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 06/30/2013] [Accepted: 07/16/2013] [Indexed: 12/17/2022]
Abstract
PURPOSE An overview of the Rapid Learning methodology, its results, and the potential impact on radiotherapy. MATERIAL AND RESULTS Rapid Learning methodology is divided into four phases. In the data phase, diverse data are collected about past patients, treatments used, and outcomes. Innovative information technologies that support semantic interoperability enable distributed learning and data sharing without additional burden on health care professionals and without the need for data to leave the hospital. In the knowledge phase, prediction models are developed for new data and treatment outcomes by applying machine learning methods to data. In the application phase, this knowledge is applied in clinical practice via novel decision support systems or via extensions of existing models such as Tumour Control Probability models. In the evaluation phase, the predictability of treatment outcomes allows the new knowledge to be evaluated by comparing predicted and actual outcomes. CONCLUSION Personalised or tailored cancer therapy ensures not only that patients receive an optimal treatment, but also that the right resources are being used for the right patients. Rapid Learning approaches combined with evidence based medicine are expected to improve the predictability of outcome and radiotherapy is the ideal field to study the value of Rapid Learning. The next step will be to include patient preferences in the decision making.
Collapse
|
29
|
Kim DK, Choi E, Song BW, Cha MJ, Ham O, Lee SY, Lee CY, Park JH, Song H, Hwang KC. Differentially regulated functional gene clusters identified in early hypoxic cardiomyocytes. Mol Biol Rep 2012; 39:9549-9556. [PMID: 22733491 DOI: 10.1007/s11033-012-1819-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Accepted: 06/10/2012] [Indexed: 12/11/2022]
Abstract
Pathological stress including myocardial infarction and hypertension causes a negative effect on calcium regulation and homeostasis. Nevertheless, few studies reveal that Ca(2+) regulatory genes are related to pathological status in cardiomyocytes under early hypoxia. To determine the alteration of Ca(2+)-related gene in hypoxic myocytes, primary neonatal rat ventricular cardiomyocytes (NRVCMs) was isolated. Survival of hypoxic NRVCMs was significantly decreased in 6 h. We confirmed an increase of reactive oxygen species (ROS) generation and Ca(2+) overload in hypoxic NRVCMs by using 2',7'-dichlorodihydro-fluorescein diacetate (H2DCFDA) and FACS analysis. Furthermore, survival/apoptotic signals were also regulated in same condition. The expression profiles of more than 30,000 genes from NRVCMs that were subjected to early hypoxia revealed 630 genes that were differentially regulated. The intracellular Na(+) overload and Ca(2+) handling genes with at least two-fold changes were confirmed. The levels of Ca(2+)-handling proteins (calsequestrin, calmodulin, and calreticulin), ion channels (NCX, Na(+)-K(+)-ATPase, SERCA2a, and PLB), and stress markers (RyR2, ANP, and BNP) were significantly altered in early hypoxia. These results demonstrate that early hypoxia alters Ca(2+)-related gene expression in NRVCMs, leading to pathological status.
Collapse
Affiliation(s)
- Do Kyun Kim
- Division of Thoracic and Cardiovascular Surgery, National Health Insurance Cooperated Ilsan Hospital, 100 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeongi-do, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Clinically relevant biomarkers in targeted radiotherapy. Clin Exp Metastasis 2012; 29:853-60. [PMID: 22886523 DOI: 10.1007/s10585-012-9523-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 07/24/2012] [Indexed: 01/24/2023]
Abstract
Three classic parameters have been recognized as predictors or biomarkers of radiation response: intrinsic radiosensitivity, degree of hypoxia and repopulation capacity of clonogenic cells during a course of fractionated radiation therapy. Although good functional assays exist to measure these tumor parameters, and their use has led to the understanding of factors affecting outcome after radiotherapy, their application in clinical practice is hampered by technical difficulties, the length of time needed to obtain results and the lack of prospective randomized clinical trials. Recently, with the progress in molecular biology, genome-wide screening methods have been used to look for genetic signatures that can distinguish between good and bad outcome after radiotherapy. One of the most promising candidates is the epidermal growth factor receptor which is overexpressed or mutated in a variety of malignancies, such lung and head and neck cancer. Inhibition of this receptor has led to radio-sensitization with the prolongation of median survival in several cancers. Since there is significant variability in the response of patients with the same disease to radiotherapy, it would be very valuable to be able to predict which patients would benefit from a molecularly targeted therapy administered with concomitant radiation in order to increase the response rate (and cure rate) of those patients with radioresistant tumors. Optimally, this assay should be able to provide results in an efficient and reproducible manner and detect tumor genetic mutations that would provide specificity to the intervention. One approach currently in clinical practice to overcome intrinsic radioresistance and repopulation is stereotactic body radiotherapy coupled with image-guided radiation, a highly precise and powerful form of radiation, allowing radiation oncologist to treat tumors with more aggressive biological doses of radiation without causing serious normal tissues injury.
Collapse
|
31
|
Toustrup K, Sørensen BS, Alsner J, Overgaard J. Hypoxia Gene Expression Signatures as Prognostic and Predictive Markers in Head and Neck Radiotherapy. Semin Radiat Oncol 2012; 22:119-27. [DOI: 10.1016/j.semradonc.2011.12.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
32
|
The prognostic value of temporal in vitro and in vivo derived hypoxia gene-expression signatures in breast cancer. Radiother Oncol 2012; 102:436-43. [DOI: 10.1016/j.radonc.2012.02.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Revised: 01/31/2012] [Accepted: 02/04/2012] [Indexed: 12/21/2022]
|
33
|
Starmans MHW, Fung G, Steck H, Wouters BG, Lambin P. A simple but highly effective approach to evaluate the prognostic performance of gene expression signatures. PLoS One 2011; 6:e28320. [PMID: 22163293 PMCID: PMC3233554 DOI: 10.1371/journal.pone.0028320] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Accepted: 11/06/2011] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Highly parallel analysis of gene expression has recently been used to identify gene sets or 'signatures' to improve patient diagnosis and risk stratification. Once a signature is generated, traditional statistical testing is used to evaluate its prognostic performance. However, due to the dimensionality of microarrays, this can lead to false interpretation of these signatures. PRINCIPAL FINDINGS A method was developed to test batches of a user-specified number of randomly chosen signatures in patient microarray datasets. The percentage of random generated signatures yielding prognostic value was assessed using ROC analysis by calculating the area under the curve (AUC) in six public available cancer patient microarray datasets. We found that a signature consisting of randomly selected genes has an average 10% chance of reaching significance when assessed in a single dataset, but can range from 1% to ∼40% depending on the dataset in question. Increasing the number of validation datasets markedly reduces this number. CONCLUSIONS We have shown that the use of an arbitrary cut-off value for evaluation of signature significance is not suitable for this type of research, but should be defined for each dataset separately. Our method can be used to establish and evaluate signature performance of any derived gene signature in a dataset by comparing its performance to thousands of randomly generated signatures. It will be of most interest for cases where few data are available and testing in multiple datasets is limited.
Collapse
Affiliation(s)
- Maud H W Starmans
- Department of Radiation Oncology (Maastro), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands.
| | | | | | | | | |
Collapse
|
34
|
Abstract
Hypoxia is a feature of most solid tumors and is associated with poor prognosis in several cancer types, including breast cancer. The master regulator of the hypoxic response is the Hypoxia-inducible factor 1α (HIF-1α). It is becoming clear that HIF-1α expression alone is not a reliable marker of tumor response to hypoxia, and recent studies have focused on determining gene and microRNA (miRNA) signatures for this complex process. The results of these studies are likely to pave the way towards the development of a robust hypoxia signature for breast and other cancers that will be useful for diagnosis and therapy. In this review, we outline the existing markers of hypoxia and recently identified gene and miRNA expression signatures, and discuss their potential as prognostic and predictive biomarkers. We also highlight how the hypoxia response is being targeted in the development of cancer therapies.
Collapse
Affiliation(s)
- Elena Favaro
- The Weatherall Institute of Molecular Medicine, Department of Oncology, University of Oxford, Oxford OX3 9DS, UK.
| | | | | | | |
Collapse
|
35
|
Abstract
Radiotherapy is used to treat approximately 50% of all cancer patients, with varying success. The dose of ionizing radiation that can be given to the tumour is determined by the sensitivity of the surrounding normal tissues. Strategies to improve radiotherapy therefore aim to increase the effect on the tumour or to decrease the effects on normal tissues. These aims must be achieved without sensitizing the normal tissues in the first approach and without protecting the tumour in the second approach. Two factors have made such approaches feasible: namely, an improved understanding of the molecular response of cells and tissues to ionizing radiation and a new appreciation of the exploitable genetic alterations in tumours. These have led to the development of treatments combining pharmacological interventions with ionizing radiation that more specifically target either tumour or normal tissue, leading to improvements in efficacy.
Collapse
Affiliation(s)
- Adrian C Begg
- Division of Experimental Therapy, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands.
| | | | | |
Collapse
|
36
|
Jiang W, Hong Y, Peng J, Fu Z, Feng X, Liu J, Shi Y, Lin J. Study on differences in the pathology, T cell subsets and gene expression in susceptible and non-susceptible hosts infected with Schistosoma japonicum. PLoS One 2010; 5:e13494. [PMID: 20976156 PMCID: PMC2956682 DOI: 10.1371/journal.pone.0013494] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2010] [Accepted: 09/24/2010] [Indexed: 11/20/2022] Open
Abstract
More than 40 kinds of mammals in China are known to be naturally infected with Schistosoma japonicum (S. japonicum); Microtus fortis (M. fortis), a species of vole, is the only mammal in which the schistosomes cannot mature or cause significant pathogenic changes. In the current study, we compared the differences in pathology by Hematoxylin-eosin staining and in changes in the T cell subsets with flow cytometry as well as gene expression using genome oligonucleotide microarrays in the lung and liver, before challenge and 10 days post-infection with schistosomes in a S. japonicum-susceptible mouse model of infection, a non-susceptible rat model and the non-permissive host, M. fortis. The results demonstrated that S. japonicum promoted a more intensive immune response and more pathological lesions in M. fortis and rats than in mice. Hematoxylin-eosin staining revealed that the immune effector cells involved were mainly eosinophilic granulocytes supplemented with heterophilic granulocytes and macrophages. The analysis of splenic T cell subsets showed that CD4(+) T cell subsets and the CD4(+)/CD8(+) ratio were increased, while the CD8(+) T cell subsets decreased remarkably in rats; whereas the CD8(+) T cell subsets were increased, but the CD4(+)/CD8(+) ratio was decreased significantly in mice. The analysis of the pattern of gene expression suggested that some immune-associated genes and apoptosis-inducing genes up-regulated, while some development-associated genes were down-regulated in the infected M. fortis compared to the uninfected controls; the three different hosts have different response mechanisms to schistosome infection. The results of this study will be helpful for identifying the key molecules in the immune response to S. japonicum in M. fortis and for understanding more about the underlying mechanism of the response, as well as for elucidating the interaction between S. japonicum and its hosts.
Collapse
Affiliation(s)
- Weibin Jiang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, People's Republic of China
- East China Normal University, School of Life Science, Shanghai, People's Republic of China
| | - Yang Hong
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, People's Republic of China
| | - Jinbiao Peng
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, People's Republic of China
| | - Zhiqiang Fu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, People's Republic of China
| | - Xingang Feng
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, People's Republic of China
| | - Jinming Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, People's Republic of China
| | - Yaojun Shi
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, People's Republic of China
| | - Jiaojiao Lin
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, People's Republic of China
| |
Collapse
|
37
|
Sørensen BS, Toustrup K, Horsman MR, Overgaard J, Alsner J. Identifying pH independent hypoxia induced genes in human squamous cell carcinomas in vitro. Acta Oncol 2010; 49:895-905. [PMID: 20429727 DOI: 10.3109/02841861003614343] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Genes upregulated by low oxygen have been suggested as endogenous markers for tumor hypoxia. Yet, most of the genes investigated have shown inconsistent results, which have led to concerns about their ability to be true hypoxia makers. Previous studies have demonstrated that expression of hypoxia induced genes can be affected by extracellular pH (pH(e)). METHODS Five different human cell lines (SiHa, FaDu(DD), UTSCC5, UTSCC14 and UTSCC15) were exposed to different oxygen concentrations and pH (7.5 or 6.3), and gene expression analyzed with microarray (Affymetrix - Human Genome U133 Plus 2.0 Array). RESULTS An analysis of two of the cell lines using SAM identified 461 probesets that were able to separate the four groups "Normal oxygen, normal pH", "Low oxygen, normal pH", "Normal oxygen, low pH" and "Low oxygen, low pH". From here it was possible to identify a fraction of probesets induced at low oxygen independent of pH in these two cell lines, this fraction included HIG2, NDRG1, PAI1 and RORA. Further verification by qPCR highlighted the necessity of using more cell lines to obtain a robust gene expression profiles. To specifically select pH independent hypoxia regulated genes across more cell lines, data for FaDu(DD), UTSCC5, UTSCC14 and UTSCC15 were analyzed to identify genes that were induced by hypoxia in each cell line, where the induction was not affected by low pH, and where the gene was not significantly induced by low pH alone. Each cell line had 65-122 probesets meeting these criteria. For genes to be considered as target genes (hypoxia inducible pH independent), genes had to be present in three of four cell lines. CONCLUSION The result is a robust hypoxia profile unaffected by pH across cell lines consisting of 27 genes. This study demonstrates a way to identify hypoxia markers by microarray, where other factors in the tumor microenvironment are taken into account.
Collapse
Affiliation(s)
- Brita Singers Sørensen
- Department of Experimental Clinical Oncology, Aarhus University Hospital, DK-8000 Aarhus C, Denmark.
| | | | | | | | | |
Collapse
|
38
|
de Jong MC, Pramana J, van der Wal JE, Lacko M, Peutz-Kootstra CJ, de Jong JM, Takes RP, Kaanders JH, van der Laan BF, Wachters J, Jansen JC, Rasch CR, van Velthuysen MLF, Grénman R, Hoebers FJ, Schuuring E, van den Brekel MW, Begg AC. CD44 Expression Predicts Local Recurrence after Radiotherapy in Larynx Cancer. Clin Cancer Res 2010; 16:5329-38. [DOI: 10.1158/1078-0432.ccr-10-0799] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
39
|
Fardin P, Barla A, Mosci S, Rosasco L, Verri A, Versteeg R, Caron HN, Molenaar JJ, Ora I, Eva A, Puppo M, Varesio L. A biology-driven approach identifies the hypoxia gene signature as a predictor of the outcome of neuroblastoma patients. Mol Cancer 2010; 9:185. [PMID: 20624283 PMCID: PMC2908582 DOI: 10.1186/1476-4598-9-185] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Accepted: 07/12/2010] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Hypoxia is a condition of low oxygen tension occurring in the tumor microenvironment and it is related to poor prognosis in human cancer. To examine the relationship between hypoxia and neuroblastoma, we generated and tested an in vitro derived hypoxia gene signature for its ability to predict patients' outcome. RESULTS We obtained the gene expression profile of 11 hypoxic neuroblastoma cell lines and we derived a robust 62 probesets signature (NB-hypo) taking advantage of the strong discriminating power of the l1-l2 feature selection technique combined with the analysis of differential gene expression. We profiled gene expression of the tumors of 88 neuroblastoma patients and divided them according to the NB-hypo expression values by K-means clustering. The NB-hypo successfully stratifies the neuroblastoma patients into good and poor prognosis groups. Multivariate Cox analysis revealed that the NB-hypo is a significant independent predictor after controlling for commonly used risk factors including the amplification of MYCN oncogene. NB-hypo increases the resolution of the MYCN stratification by dividing patients with MYCN not amplified tumors in good and poor outcome suggesting that hypoxia is associated with the aggressiveness of neuroblastoma tumor independently from MYCN amplification. CONCLUSIONS Our results demonstrate that the NB-hypo is a novel and independent prognostic factor for neuroblastoma and support the view that hypoxia is negatively correlated with tumors' outcome. We show the power of the biology-driven approach in defining hypoxia as a critical molecular program in neuroblastoma and the potential for improvement in the current criteria for risk stratification.
Collapse
Affiliation(s)
- Paolo Fardin
- Laboratory of Molecular Biology, Gaslini Institute, Genoa, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Jubb AM, Buffa FM, Harris AL. Assessment of tumour hypoxia for prediction of response to therapy and cancer prognosis. J Cell Mol Med 2009; 14:18-29. [PMID: 19840191 PMCID: PMC3837600 DOI: 10.1111/j.1582-4934.2009.00944.x] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Tumour cells exploit both genetic and adaptive means to survive and proliferate in hypoxic microenvironments, resulting in the outgrowth of more aggressive tumour cell clones. Direct measurements of tumour oxygenation, and surrogate markers of the hypoxic response in tumours (for instance, hypoxia inducible factor-1α, carbonic anhydrase 9 and glucose transporter-1) are well-established prognostic markers in solid cancers. However, individual markers do not fully capture the complex, dynamic and heterogeneous hypoxic response in cancer. To overcome this, expression profiling has been employed to identify hypoxia signatures in cohorts or models of human cancer. Several of these hypoxia signatures have demonstrated prognostic significance in independent cancer datasets. Nevertheless, individual hypoxia markers have been shown to predict the benefit from hypoxia-modifying or anti-angiogenic therapies. This review aims to discuss the clinical impact of translational work on hypoxia markers and to explore future directions for research in this area.
Collapse
Affiliation(s)
- Adrian M Jubb
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | | | | |
Collapse
|
41
|
Rodemann HP. Molecular radiation biology: Perspectives for radiation oncology. Radiother Oncol 2009; 92:293-8. [PMID: 19726094 DOI: 10.1016/j.radonc.2009.08.023] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2009] [Accepted: 08/15/2009] [Indexed: 12/27/2022]
|
42
|
The use of a comprehensive tumour xenograft dataset to validate gene signatures relevant for radiation response. Radiother Oncol 2009; 92:417-22. [PMID: 19615772 DOI: 10.1016/j.radonc.2009.06.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2009] [Revised: 06/11/2009] [Accepted: 06/24/2009] [Indexed: 11/21/2022]
Abstract
PURPOSE To investigate the use of xenograft models in a novel gene signature validation method using gene expression microarrays. MATERIALS AND METHODS Gene expression profiles of ten human Head and Neck squamous cell carcinomas (HNSCCs) were obtained. Several published prognostic gene expression signatures were evaluated within this set. These consisted of different radiotherapy relevant signatures (i.e. for hypoxia, proliferation and 'stemness'). Signatures were correlated with various endpoints that have been determined in the ten different xenograft models. These include immunohistochemical measures for hypoxia and proliferation, volume doubling time (VDT) and local tumour control after fractionated irradiation or after single dose irradiation under clamp hypoxia. RESULTS We found several significant correlations between the published gene expression signatures and tumour parameters. Several signatures, like the proliferation and wound signature correlated with BrdU labelling index. Further a 'stemness'-related gene signature showed a strong negative correlation with hypoxic fraction. CONCLUSIONS Simultaneous assessment of immunohistochemistry, in vivo tumour properties and gene expression profiling in a comprehensive set of xenograft models can be used to validate and potentially infer biological information about prognostic gene signatures.
Collapse
|
43
|
Ebbesen P, Pettersen EO, Gorr TA, Jobst G, Williams K, Kieninger J, Wenger RH, Pastorekova S, Dubois L, Lambin P, Wouters BG, Van Den Beucken T, Supuran CT, Poellinger L, Ratcliffe P, Kanopka A, Görlach A, Gasmann M, Harris AL, Maxwell P, Scozzafava A. Taking advantage of tumor cell adaptations to hypoxia for developing new tumor markers and treatment strategies. J Enzyme Inhib Med Chem 2009; 24 Suppl 1:1-39. [PMID: 19005871 DOI: 10.1080/14756360902784425] [Citation(s) in RCA: 157] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Cancer cells in hypoxic areas of solid tumors are to a large extent protected against the action of radiation as well as many chemotherapeutic drugs. There are, however, two different aspects of the problem caused by tumor hypoxia when cancer therapy is concerned: One is due to the chemical reactions that molecular oxygen enters into therapeutically targeted cells. This results in a direct chemical protection against therapy by the hypoxic microenvironment, which has little to do with cellular biological regulatory processes. This part of the protective effect of hypoxia has been known for more than half a century and has been studied extensively. However, in recent years there has been more focus on the other aspect of hypoxia, namely the effect of this microenvironmental condition on selecting cells with certain genetic prerequisites that are negative with respect to patient prognosis. There are adaptive mechanisms, where hypoxia induces regulatory cascades in cells resulting in a changed metabolism or changes in extracellular signaling. These processes may lead to changes in cellular intrinsic sensitivity to treatment irrespective of oxygenation and, furthermore, may also have consequences for tissue organization. Thus, the adaptive mechanisms induced by hypoxia itself may have a selective effect on cells, with a fine-tuned protection against damage and stress of many kinds. It therefore could be that the adaptive mechanisms may take advantage of for new tumor labeling/imaging and treatment strategies. One of the Achilles' heels of hypoxia research has always been the exact measurements of tissue oxygenation as well as the control of oxygenation in biological tumor models. Thus, development of technology that can ease this control is vital in order to study mechanisms and perform drug development under relevant conditions. An integrated EU Framework project 2004-2009, termed EUROXY, demonstrates several pathways involved in transcription and translation control of the hypoxic cell phenotype and evidence of cross-talk with responses to pH and redox changes. The carbonic anhydrase isoenzyme CA IX was selected for further studies due to its expression on the surface of many types of hypoxic tumors. The effort has led to marketable culture flasks with sensors and incubation equipment, and the synthesis of new drug candidates against new molecular targets. New labeling/imaging methods for cancer diagnosing and imaging of hypoxic cancer tissue are now being tested in xenograft models and are also in early clinical testing, while new potential anti-cancer drugs are undergoing tests using xenografted tumor cancers. The present article describes the above results in individual consortium partner presentations.
Collapse
Affiliation(s)
- Peter Ebbesen
- Laboratory for Stem Cell Research, Aalborg University, Aarhus, Denmark.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Petit SF, Dekker ALAJ, Seigneuric R, Murrer L, van Riel NAW, Nordsmark M, Overgaard J, Lambin P, Wouters BG. Intra-voxel heterogeneity influences the dose prescription for dose-painting with radiotherapy: a modelling study. Phys Med Biol 2009; 54:2179-96. [PMID: 19293465 DOI: 10.1088/0031-9155/54/7/022] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The purpose of this study was to increase the potential of dose redistribution by incorporating estimates of oxygen heterogeneity within imaging voxels for optimal dose determination. Cellular oxygen tension (pO(2)) distributions were estimated for imaging-size-based voxels by solving oxygen diffusion-consumption equations around capillaries placed at random locations. The linear-quadratic model was used to determine cell survival in the voxels as a function of pO(2) and dose. The dose distribution across the tumour was optimized to yield minimal survival after 30 x 2 Gy fractions by redistributing the dose based on differences in oxygen levels. Eppendorf data of a series of 69 tumours were used as a surrogate of what might be expected from oxygen imaging datasets. Dose optimizations were performed both taking into account cellular heterogeneity in oxygenation within voxels and assuming a homogeneous cellular distribution of oxygen. Our simulations show that dose redistribution based on derived cellular oxygen distributions within voxels result in dose distributions that require less total dose to obtain the same degree of cell kill as dose distributions that were optimized with a model that considered voxels as homogeneous with respect to oxygen. Moderately hypoxic tumours are expected to gain most from dose redistribution. Incorporating cellular-based distributions of radiosensitivity into dose-planning algorithms theoretically improves the potential gains from dose redistribution algorithms.
Collapse
Affiliation(s)
- Steven F Petit
- Department of Radiation Oncology (Maastro), GROW, U.H. Maastricht, Maastricht, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Baumann M, Krause M. Tumor Biology’s Impact on Clinical Cure Rates. THE IMPACT OF TUMOR BIOLOGY ON CANCER TREATMENT AND MULTIDISCIPLINARY STRATEGIES 2009. [DOI: 10.1007/978-3-540-74386-6_19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
46
|
Beyer S, Kristensen MM, Jensen KS, Johansen JV, Staller P. The histone demethylases JMJD1A and JMJD2B are transcriptional targets of hypoxia-inducible factor HIF. J Biol Chem 2008; 283:36542-52. [PMID: 18984585 PMCID: PMC2662309 DOI: 10.1074/jbc.m804578200] [Citation(s) in RCA: 275] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2008] [Revised: 10/29/2008] [Indexed: 12/31/2022] Open
Abstract
Posttranslational histone modifications serve to store epigenetic information and control both nucleosome assembly and recruitment of non-histone proteins. Histone methylation occurs on arginine and lysine residues and is involved in the regulation of gene transcription. A dynamic control of these modifications is exerted by histone methyltransferases and the recently discovered histone demethylases. Here we show that the hypoxia-inducible factor HIF-1alpha binds to specific recognition sites in the genes encoding the jumonji family histone demethylases JMJD1A and JMJD2B and induces their expression. Accordingly, hypoxic cells express elevated levels of JMJD1A and JMJD2B mRNA and protein. Furthermore, we find increased expression of JMJD1A and JMJD2B in renal cancer cells that have lost the von Hippel Lindau tumor suppressor protein VHL and therefore display a deregulated expression of hypoxia-inducible factor. Studies on ectopically expressed JMJD1A and JMJD2B indicate that both proteins retain their histone lysine demethylase activity in hypoxia and thereby might impact the hypoxic gene expression program.
Collapse
Affiliation(s)
- Sophie Beyer
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | |
Collapse
|
47
|
Milani M, Harris AL. Targeting tumour hypoxia in breast cancer. Eur J Cancer 2008; 44:2766-73. [PMID: 18990559 DOI: 10.1016/j.ejca.2008.09.025] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2008] [Accepted: 09/23/2008] [Indexed: 12/28/2022]
|
48
|
Winnard PT, Pathak AP, Dhara S, Cho SY, Raman V, Pomper MG. Molecular imaging of metastatic potential. J Nucl Med 2008; 49 Suppl 2:96S-112S. [PMID: 18523068 DOI: 10.2967/jnumed.107.045948] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
If molecular imaging is to prove clinically useful it will have to surpass current, primarily anatomic techniques in terms of sensitivity and the ability to detect minimal changes in tissue. One of the most important tests for molecular imaging is to determine whether it can image the metastatic potential of tumors. Like all predictive endeavors, the imaging of such "potential" is a daunting task, but one that only molecular imaging--rather than standard, anatomic techniques--is likely to solve. Although difficult, imaging of metastatic potential is also arguably the most important task for molecular imaging of cancer because it is generally the dissemination of malignant tissue, not its prolonged residence in an inopportune site, which kills the patient. Below are examples of uses of molecular imaging of metastases as well as of metastatic potential, the former being a far more developed area of clinical inquiry.
Collapse
Affiliation(s)
- Paul T Winnard
- Russell H Morgan Department of Radiology and Radiological Sciences, Johns Hopkins Medical Institutions, Baltimore 21231, Maryland, USA
| | | | | | | | | | | |
Collapse
|
49
|
Liu SK, Coackley C, Krause M, Jalali F, Chan N, Bristow RG. A novel poly(ADP-ribose) polymerase inhibitor, ABT-888, radiosensitizes malignant human cell lines under hypoxia. Radiother Oncol 2008; 88:258-68. [DOI: 10.1016/j.radonc.2008.04.005] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2008] [Revised: 04/04/2008] [Accepted: 04/08/2008] [Indexed: 12/13/2022]
|
50
|
Begg A, van der Kogel A. Clinical radiobiology in 2008. Radiother Oncol 2008; 86:295-9. [PMID: 18313778 DOI: 10.1016/j.radonc.2008.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2008] [Revised: 02/01/2008] [Accepted: 02/01/2008] [Indexed: 11/18/2022]
|