Squamous skin cancer
Research on squamous skin cancers spans the analysis of fundamental molecular pathways in disease progression through to tumour cell interactions with the immune system and microenvironment.
Staff
Publications
2025
Bone, M., Schreyer, D., Treanor-Taylor, M., Proby, C., Harwood, C., Leigh, I. M., Bailey, P., Inman, G. (2025) The landscape of long non-coding RNA during cSCC progression. British Journal of Dermatology,
Hurley, R., Osbourne, J., Inman, G. J., Conway, D. I., Paterson, C., Douglas, C. (2025) Pre-treatment serum prognostic scores and survival in curatively treated laryngeal cancer. Laryngoscope Investigative Otolaryngology,
White, S. E., Schwartze, T. A., Mukundan, A., Schoenherr, C., Singh, S. P., van Dinther, M., Cunningham, K. T., White, M. P. J., Campion, T., Pritchard, J., Hinck, C. S., ten Dijke, P., Inman, G. J., Maizels, R. M., Hinck, A. P. (2025) TGM6 is a helminth secretory product that mimics TGF-β binding to TGFBR2 to antagonize signaling in fibroblasts. Nature Communications, 16, (doi: 10.1038/s41467-025-56954-z)
Hurley, R., Paterson, C., Conway, D. I., Inman, G. J., Douglas, C. M. (2025) Laryngeal cancer in the West of Scotland 2014–2020: trends and survival in a cohort of 867 patients. Laryngoscope, (doi: 10.1002/lary.31992)
2024
Singh, S. P., Smyth, D. J., Cunningham, K. T., Mukundan, A., Byeon, C.-H., Hinck, C. S., White, M. P.J., Ciancia, C., Wąsowska, N., Sanders, A., Jin, R., White, R. F., Lilla, S., Zanivan, S., Schoenherr, C., Inman, G., van Dinther, M., ten Dijke, P., Hinck, A. P., Maizels, R. M. (2024) The TGF-β mimic TGM4 achieves cell specificity through combinatorial surface co-receptor binding. EMBO Reports, (doi: 10.1038/s44319-024-00323-2)
Kiourtis, C. et al. (2024) Hepatocellular senescence induces multi-organ senescence and dysfunction via TGFβ. Nature Cell Biology, (doi: 10.1038/s41556-024-01543-3)
Laing, A., Elmarghany, A., Alghaith, A. A., Gouma, A., Stevens, T., Winton, A., Cassels, J., Clarke, C. J., Schwab, C., Harrison, C. J., Gibson, B., Keeshan, K. (2024) Paediatric bone marrow mesenchymal stem cells support acute myeloid leukaemia cell survival and enhance chemoresistance via contact-independent mechanism. British Journal of Haematology, (doi: 10.1111/bjh.19884)
Hargrave, K. E., Worrell, J. C., Pirillo, C., Brennan, E., Masdefiol Garriga, A., Gray, J. I., Purnell, T., Roberts, E. W., MacLeod, M. K.L. (2024) Lung influenza virus specific memory CD4 T cell location and optimal cytokine production are dependent on interactions with lung antigen-presenting cells. Mucosal Immunology, 17, pp. 843-857. (doi: 10.1016/j.mucimm.2024.06.001)
Smith, C. D.L., McMahon, A. D., Purkayastha, M., Creaney, G., Clements, K., Inman, G. J., Bhatti, L. A., Douglas, C. M., Paterson, C., Conway, D. I. (2024) Head and neck cancer incidence is rising but the sociodemographic profile is unchanging: a population epidemiological study (2001-2020) BJC Reports, 2, (doi: 10.1038/s44276-024-00089-z)
Strathearn, L. S., Spender, L. C., Schoenherr, C., Mason, S., Edwards, R., Blyth, K., Inman, G. (2024) C1orf106 (INAVA) is a SMAD3-dependent TGF-β target gene that promotes clonogenicity and correlates with poor prognosis in breast cancer. Cells, 13, (doi: 10.3390/cells13181530)
Smith, C. D.L., McMahon, A. D., Lyall, D. M., Goulart, M., Inman, G., Ross, A., Gormley, M., Dudding, T., Macfarlane, G. J., Robinson, M., Richiardi, L., Serraino, D., Polesel, J., Canova, C., Ahrens, W., Healy, C. M., Lagiou, P., Holcatova, I., Alemany, L., Znoar, A., Waterboer, T., Brennan, P., Virani, S., Conway, D. I. (2024) Development and external validation of a head and neck cancer risk prediction model. Head and Neck, 46, pp. 2261-2273. (doi: 10.1002/hed.27834)
Patterson, S. D., Massett, M. E., Huang, X., Jørgensen, H. G., Michie, A. M. (2024) The MYC–NFATC2 axis maintains the cell cycle and mitochondrial function in acute myeloid leukaemia cells. Molecular Oncology, 18, pp. 2234-2254. (doi: 10.1002/1878-0261.13630)
Ramirez-Guzman, L. A., Huang, W., Cole, J. J., Jorgensen, H. G. (2024) GAS2 upregulation is a targetable vulnerability in chronic myeloid leukaemia. International Journal of Translational Medicine, 4, pp. 354-368. (doi: 10.3390/ijtm4020023)
Kumar, S., Basto, A. P., Ribeiro, F., Almeida, S. C.P., Campos, P., Peres, C., Pulvirenti, N., Al-Khalidi, S., Kilbey, A., Tosello, J., Piaggio, E., Russo, M., Gama-Carvalho, M., Coffelt, S. B., Roberts, E. W., Geginat, J., Florindo, H. F., Graca, L. (2024) Specialized Tfh cell subsets driving type-1 and type-21 humoral responses in lymphoid tissue. Cell Discovery, 10, (doi: 10.1038/s41421-024-00681-0)
Mardilovich, K., Naylor, G., Julian, L., Phinichkusolchit, N., Keeshan, K., Blyth, K., Olson, M. F. (2024) Caspase-resistant ROCK1 expression prolongs survival of Eµ-Myc B cell lymphoma mice. Disease Models and Mechanisms, 17, (doi: 10.1242/dmm.050631)
Bentley-Abbot, C., Heslop, R., Pirillo, C., Chandrasegaran, P., McConnell, G., Roberts, E., Hutchinson, E., MacLeod, A. (2024) An easy to use tool for the analysis of subcellular mRNA transcript colocalisation in smFISH data. Scientific Reports, 14, (doi: 10.1038/s41598-024-58641-3)
Mahmood, M., Liu, E. M., Shergold, A. L., Tolla, E., Tait-Mulder, J., Huerta Uribe, A., Shokry, E., Young, A. L., Lilla, S., Kim, M., Park, T., Boscenco, S., Manchon, J. L., Rodríguez-Antona, C., Walters, R. C., Springett, R. J., Blaza, J. N., Mitchell, L., Blyth, K., Zanivan, S., Sumpton, D., Roberts, E. W., Reznik, E., Gammage, P. A. (2024) Mitochondrial DNA mutations drive aerobic glycolysis to enhance checkpoint blockade response in melanoma. Nature Cancer, (doi: 10.1038/s43018-023-00721-w)
Scott, M. T., Liu, W., Mitchell, R., Clarke, C. J., Kinstrie, R., Warren, F., Almasoudi, H., Stevens, T., Dunn, K., Pritchard, J., Drotar, M. E., Michie, A. M., Jorgensen, H. G., Higgins, B., Copland, M., Vetrie, D. (2024) Activating p53 abolishes self-renewal of quiescent leukaemic stem cells in residual CML disease. Nature Communications, 15, (doi: 10.1038/s41467-024-44771-9)
Derby, S. et al. (2024) Inhibition of ATR opposes glioblastoma invasion through disruption of cytoskeletal networks and integrin internalisation via macropinocytosis. Neuro-Oncology, (doi: 10.1093/neuonc/noad210)
2023
Wang, J., Harwood, C. A., Bailey, E., Bewicke-Copley, F., Anene, C. A., Thomson, J., Qamar, M. J., Laban, R., Nourse, C., Schoenherr, C., Treanor-Taylor, M., Healy, E., Lai, C., Craig, P., Moyes, C., Rickaby, W., Martin, J., Proby, C., Inman, G. J., Leigh, I. M. (2023) Transcriptomic analysis of cutaneous squamous cell carcinoma reveals a multigene prognostic signature associated with metastasis. Journal of the American Academy of Dermatology, 89, pp. 1159-1166. (doi: 10.1016/j.jaad.2023.08.012)
Young, A. L., Lorimer, T., Al-Khalidi, S. K., Roberts, E. W. (2023) De novo priming: driver of immunotherapy responses or epiphenomenon? Essays In Biochemistry, 67, pp. 929-939. (doi: 10.1042/EBC20220244)
Bailey, P., Ridgway, R. L., Cammareri, P., Treanor-Taylor, M., Bailey, U.-M., Schoenherr, C., Bone, M., Schreyer, D., Purdie, K., Thomson, J., Rickaby, W., Jackstadt, R., Campbell, A. D., Dimonitsas, E., Stratigos, A. J., Arron, S. T., Wang, J., Blyth, K., Proby, C. M., Harwood, C. A., Sansom, O. J., Leigh, I. M., Inman, G. J. (2023) Driver gene combinations dictate cutaneous squamous cell carcinoma disease continuum progression. Nature Communications, 14, (doi: 10.1038/s41467-023-40822-9)
Darici, S., Jørgensen, H. G., Huang, X., Serafin, V., Antolini, L., Barozzi, P., Luppi, M., Forghieri, F., Marmiroli, S., Zavatti, M. (2023) Improved efficacy of quizartinib in combination therapy with PI3K inhibition in primary FLT3-ITD AML cells. Advances in Biological Regulation, 89, (doi: 10.1016/j.jbior.2023.100974)
Pirillo, C., Al Khalidi, S., Sims, A., Devlin, R., Zhao, H., Pinto, R., Jasim, S., Shearer, P. A., Shergold, A. L., Donnelly, H., Bravo-Blas, A., Loney, C., Perona-Wright, G., Hutchinson, E., Roberts, E. W. (2023) Cotransfer of antigen and contextual information harmonizes peripheral and lymph node conventional dendritic cell activation. Science Immunology, 8, (doi: 10.1126/sciimmunol.adg8249)
Stavrou, V., Fultang, L., Booth, S., De Simone, D., Bartnik, A., Scarpa, U., Gneo, L., Panetti, S., Potluri, S., Almowaled, M., Barlow, J., Jankevics, A., Lloyd, G., Southam, A., Priestman, D. A., Cheng, P., Dunn, W., Platt, F., Endou, H., Craddock, C., Keeshan, K., Mussai, F., De Santo, C. (2023) Invariant NKT cells metabolically adapt to the acute myeloid leukaemia environment. Cancer Immunology, Immunotherapy, 72, pp. 543-560. (doi: 10.1007/s00262-022-03268-4)
2022
Flanagan, D. J. et al. (2022) Epithelial TGFβ engages growth-factor signalling to circumvent apoptosis and drive intestinal tumourigenesis with aggressive features. Nature Communications, 13, (doi: 10.1038/s41467-022-35134-3)
Smith, C. D.L., McMahon, A. D., Ross, A., Inman, G. J., Conway, D. I. (2022) Risk prediction models for head and neck cancer: a rapid review. Laryngoscope Investigative Otolaryngology, 7, pp. 1893-1908. (doi: 10.1002/lio2.982)
Sims, A., Tornaletti, L. B., Jasim, S., Pirillo, C., Devlin, R., Hirst, J. C., Loney, C., Wojtus, J., Sloan, E., Thorley, L., Boutell, C., Roberts, E., Hutchinson, E. (2022) Superinfection exclusion creates spatially distinct influenza virus populations. PLoS Biology, 21, (doi: 10.1371/journal.pbio.3001941)
Jamieson, S. A., Pudjihartono, M., Horne, C. R., Viloria, J. S., Dunlop, J. L., McMillan, H. D., Day, R. C., Keeshan, K., Murphy, J. M., Mace, P. D. (2022) Nanobodies identity an activated state of the TRIB2 pseudokinase. Structure, 30, pp. 1518-1529.e5. (doi: 10.1016/j.str.2022.08.006)
Gómez-Castañeda, E., Hopcroft, L. E.M., Rogers, S., Munje, C., Bittencourt-Silvestre, J., Copland, M., Vetrie, D., Holyoake, T., Jørgensen, H. G. (2022) Tyrosine kinase inhibitor independent gene expression signature in CML offers new targets for LSPC eradication therapy. Cancers, 14, (doi: 10.3390/cancers14215253)
Leslie, J. et al. (2022) CXCR2 inhibition enables NASH-HCC immunotherapy. Gut, 71, pp. 2093-2106. (doi: 10.1136/gutjnl-2021-326259)
Bravo-Blas, A., Pirillo, C., Shergold, A., Andrusaite, A., Roberts, E. W. (2022) Think global but act local: Tuning the dendritic cell response in cancer. International Journal of Biochemistry and Cell Biology, 147, (doi: 10.1016/j.biocel.2022.106227)
Pirillo, C., Birch, F., Tissot, F., Anton, S. G., Haltalli, M., Tini, V., Kong, I., Piot, C., Partridge, B., Pospori, C., Keeshan, K., Santamaria, S., Hawkins, E., Falini, B., Marra, A., Duarte, D., Lee, C. F., Roberts, E., Celso, C. L. (2022) Metalloproteinase inhibition reduces AML growth, prevents stem cell loss and improves chemotherapy effectiveness. Blood Advances, 6, pp. 3126-3141. (doi: 10.1182/bloodadvances.2021004321)
Lal, R., Ritchie, J., Richmond, L., Keeshan, K. (2022) Detecting endogenous TRIB2 protein expression by flow cytometry and western blotting. Academic Press
Devlin, R., Roberts, E. (2022) Building a healthy mouse model ecosystem to interrogate cancer biology. Disease Models and Mechanisms, 15, (doi: 10.1242/dmm.049795)
2021
Darici, S., Zavatti, M., Braglia, L., Accordi, B., Serafin, V., Horne, G. A., Manzoli, L., Palumbo, C., Huang, X., Jørgensen, H. G., Marmiroli, S. (2021) Synergistic cytotoxicity of dual PI3K/mTOR and FLT3 inhibition in FLT3-ITD AML cells. Advances in Biological Regulation, 82, (doi: 10.1016/j.jbior.2021.100830)
Hollis, R. L., Stillie, L. J., Hopkins, S., Bartos, C., Churchman, M., Rye, T., Nussey, F., Fegan, S., Nirsimloo, R., Inman, G. J., Herrington, C. S., Gourley, C. (2021) Clinicopathological determinants of recurrence risk and survival in mucinous ovarian carcinoma. Cancers, 13, (doi: 10.3390/cancers13225839)
Birch, J., Strathdee, K., Stevenson, K., Derby, S., Dutton, L., Clough, E., Koessinger, A., Gilmour, L., McGhee, E., McGarrity-Cottrell, C., Dibekeme, A. a., Collis, S., Rominiyi, O., Lembruber Soares, L., Solecki, G., Winkler, F., Carlin, L., Inman, G., Chalmers, A., Norman, J., Carruthers, R. (2021) Inhibition of ATR prevents macropinocytosis driven retraction of neurites and opposes invasion in GBM. Research Square, (doi: 10.21203/rs.3.rs-967109/v1)
Aung, M. M. K., Mills, M. L., Bittencourt-Silvestre, J., Keeshan, K. (2021) Insights into the molecular profiles of adult and paediatric acute myeloid leukaemia. Molecular Oncology, 15, pp. 2253-2272. (doi: 10.1002/1878-0261.12899)
Patterson, S. D., Huang, X., Jørgensen, H. G., Michie, A. M. (2021) Transcriptional regulation by the NFAT family in acute myeloid leukaemia. Hemato, 2, pp. 556-571. (doi: 10.3390/hemato2030035)
Ntala, C., Salji, M., Salmond, J., Officer, L., Teodosio, A. V., Blomme, A., McGhee, E. J., Powley, I., Ahmad, I., Kruithof-de Julio, M., Thalmann, G., Roberts, E., Goodyear, C. S., Jamaspishvili, T., Berman, D. M., Carlin, L. M., Le Quesne, J., Leung, H. Y. (2021) Analysis of prostate cancer tumor microenvironment identifies reduced stromal CD4 effector T-cell infiltration in tumors with pelvic nodal metastasis. European Urology Open Science, 29, pp. 19-29. (doi: 10.1016/j.euros.2021.05.001)
Leach, J. D.G. et al. (2021) Oncogenic BRAF, unrestrained by TGFβ-receptor signalling, drives right-sided colonic tumorigenesis. Nature Communications, 12, (doi: 10.1038/s41467-021-23717-5)
Massett, M. E., Monaghan, L., Patterson, S., Mannion, N., Bunschoten, R. P., Hoose, A., Marmiroli, S., Liskamp, R. M.J., Jørgensen, H. G., Vetrie, D., Michie, A. M., Huang, X. (2021) A KDM4A-PAF1-mediated epigenomic network is essential for acute myeloid leukemia cell self-renewal and survival. Cell Death and Disease, 12, (doi: 10.1038/s41419-021-03738-0)
Latif, A.-L. et al. (2021) BRD4-mediated repression of p53 is a target for combination therapy in AML. Nature Communications, 12, (doi: 10.1038/s41467-020-20378-8)
Keeshan, K. (2021) Superenhancing AML with Trib1. Blood, 137, pp. 8-9. (doi: 10.1182/blood.2020008315)
Marensi, V., Keeshan, K. R., MacEwan, D. J. (2021) Pharmacological impact of FLT3 mutations on receptor activity and responsiveness to tyrosine kinase inhibitors. Biochemical Pharmacology, 183, (doi: 10.1016/j.bcp.2020.114348)
McMillan, H. D., Keeshan, K., Dunbier, A. K., Mace, P. D. (2021) Structure vs. Function of TRIB1—Myeloid Neoplasms and Beyond. Cancers, 13, (doi: 10.3390/cancers13123060)
2020
Richmond, L., Keeshan, K. (2020) Pseudokinases: a tribble‐edged sword. FEBS Journal, 287, pp. 4170-4182. (doi: 10.1111/febs.15096)
Darici, S., Alkhaldi, H., Horne, G., Jørgensen, H. G., Marmiroli, S., Huang, X. (2020) Targeting PI3K/Akt/mTOR in AML: rationale and clinical evidence. Journal of Clinical Medicine, 9, (doi: 10.3390/jcm9092934)
Mitxitorena, I., Somma, D., Mitchell, J. P., Lepistö, M., Tyrchan, C., Smith, E. L., Kiely, P. A., Walden, H., Keeshan, K., Carmody, R. J. (2020) The deubiquitinase USP7 uses a distinct ubiquitin-like domain to deubiquitinate NF-ĸB subunits. Journal of Biological Chemistry, 295, pp. 11754-11763. (doi: 10.1074/jbc.RA120.014113)
Wei, Q., Young, J., Holle, A., Li, J., Bieback, K., Inman, G., Spatz, J. P., Cavalcanti-Adam, E. A. (2020) Soft hydrogels for balancing cell proliferation and differentiation. ACS Biomaterials Science and Engineering, 6, pp. 4687-4701. (doi: 10.1021/acsbiomaterials.0c00854)
Wei, Q., Holle, A., Liu, J., Posa, F., Biagioni, F., Croci, O., Benk, A. S., Young, J., Noureddine, F., Deng, J., Zhang, M., Inman, G. J., Spatz, J. P., Campaner, S., Cavalcanti‐Adam, E. A. (2020) BMP‐2 signaling and mechanotransduction synergize to drive osteogenic differentiation via YAP/TAZ. Advanced Science, 7, (doi: 10.1002/advs.201902931)
Sarrou, E., Richmond, L., Carmody, R. J., Gibson, B., Keeshan, K. (2020) CRISPR gene editing of murine blood stem and progenitor cells induces MLL-AF9 chromosomal translocation and MLL-AF9 leukaemogenesis. International Journal of Molecular Sciences, 21, (doi: 10.3390/ijms21124266)
2019
Collins, P. E., Somma, D., Kerrigan, D., Herrington, F., Keeshan, K. R., Nibbs, R. J.B., Carmody, R. J. (2019) The IκB-protein BCL-3 controls toll-like receptor-induced MAPK activity by promoting TPL-2 degradation in the nucleus. Proceedings of the National Academy of Sciences of the United States of America, 116, pp. 25828-25838. (doi: 10.1073/pnas.1900408116)
Smith, E. L., Somma, D., Kerrigan, D., McIntyre, Z., Cole, J. J., Liang, K. L., Kiely, P. A., Keeshan, K., Carmody, R. J. (2019) The regulation of sequence specific NF-ĸB DNA binding and transcription by IKKβ Phosphorylation of NF-ĸB p50 at Serine 80. Nucleic Acids Research, 47, pp. 11151-11161. (doi: 10.1093/nar/gkz873)
Mussai, F., Wheat, R., Sarrou, E., Booth, S., Stavrou, V., Fultang, L., Perry, T., Kearns, P., Cheng, P., Keeshan, K., Craddock, C., De Santo, C. (2019) Targeting the arginine metabolic brake enhances immunotherapy for leukaemia. International Journal of Cancer, 145, pp. 2201-2208. (doi: 10.1002/ijc.32028)
Hayatigolkhatmi, K., Padroni, G., Su, W., Fang, L., Gómez-Castañeda, E., Hsieh, Y.C., Jackson, L., Pellicano, F., Burley, G.A., Jørgensen, H.G. (2019) An investigation of targeted inhibition of transcription factor activity with pyrrole imidazole polyamide (PA) in chronic myeloid leukemia (CML) blast crisis cells. Bioorganic and Medicinal Chemistry Letters, 29, pp. 2622-2625. (doi: 10.1016/j.bmcl.2019.07.049)
Monaghan, L., Massett, M. E., Bunschoten, R. P., Hoose, A., Pirvan, P.-A., Liskamp, R. M.J., Jørgensen, H. G., Huang, X. (2019) The emerging role of H3K9me3 as a potential therapeutic target in acute myeloid leukaemia. Frontiers in Oncology, 9, (doi: 10.3389/fonc.2019.00705)
Badshah, I.I., Brown, S., Weibel, L., Rose, A., Way, B., Sebire, N., Inman, G., Harper, J., O'Shaughnessy, R.F.L. (2019) Differential expression of secreted factors SOSTDC1 and ADAMTS8 cause profibrotic changes in linear morphoea fibroblasts. British Journal of Dermatology, 180, pp. 1135-1149. (doi: 10.1111/bjd.17352)
Binnewies, M., Mujal, A. M., Pollack, J. L., Combes, A. J., Hardison, E. A., Barry, K. C., Tsui, J., Ruhland, M. K., Kersten, K., Abushawish, M. A., Spasic, M., Giurintano, J. P., Chan, V., Daud, A. I., Ha, P., Ye, C. J., Roberts, E. W., Krummel, M. F. (2019) Unleashing type-2 dendritic cells to drive protective antitumor CD4+ T cell immunity. Cell, 177, pp. 556-571.e16. (doi: 10.1016/j.cell.2019.02.005)
Spender, L. C., Ferguson, G. J., Hughes, G. D., Davies, B. R., Goldberg, F. W., Herrera, B., Taylor, R. G., Strathearn, L. S., Sansom, O. J., Barry, S. T., Inman, G. I. (2019) Preclinical evaluation of AZ12601011 and AZ12799734, inhibitors of transforming growth factor β superfamily type 1 receptors. Molecular Pharmacology, 95, pp. 222-234. (doi: 10.1124/mol.118.112946)
Hassan, S., Purdie, K. J., Wang, J., Harwood, C. A., Proby, C. M., Pourreyron, C., Mladkova, N., Nagano, A., Dhayade, S., Athineos, D., Caley, M., Mannella, V., Blyth, K., Inman, G. J., Leigh, I. M. (2019) A unique panel of patient-derived cutaneous squamous cell carcinoma cell lines provides a preclinical pathway for therapeutic testing. International Journal of Molecular Sciences, 20, (doi: 10.3390/ijms20143428)
2018
Chaudhury, S., O'Connor, C., Cañete, A., Bittencourt-Silvestre, J., Sarrou, E., Prendergast, Á., Choi, J., Johnston, P., Wells, C. A., Gibson, B., Keeshan, K. (2018) Age-specific biological and molecular profiling distinguishes paediatric from adult acute myeloid leukaemias. Nature Communications, 9, (doi: 10.1038/s41467-018-07584-1)
Salomé, M., Hopcroft, L., Keeshan, K. (2018) Inverse and correlative relationships between TRIBBLES genes indicate non-redundant functions during normal and malignant hemopoiesis. Experimental Hematology, 66, pp. 63-78.e13. (doi: 10.1016/j.exphem.2018.07.005)
Foulkes, D. M., Byrne, D. P., Yeung, W., Shrestha, S., Bailey, F. P., Ferries, S., Eyers, C. E., Keeshan, K., Wells, C., Drewry, D. H., Zuercher, W. J., Kannan, N., Eyers, P. A. (2018) Covalent inhibitors of EGFR family protein kinases induce degradation of human Tribbles 2 (TRIB2) pseudokinase in cancer cells. Science Signaling, 11, (doi: 10.1126/scisignal.aat7951)
Toofan, P., Busch, C., Morrison, H., O'Brien, S., Jørgensen, H., Copland, M., Wheadon, H. (2018) Chronic myeloid leukemia cells require the bone morphogenic protein pathway for cell cycle progression and self-renewal. Cell Death and Disease, 9, (doi: 10.1038/s41419-018-0905-2)
Inman, G. J., Wang, J., Nagano, A., Alexandrov, L. B., Purdie, K. J., Taylor, R. G., Sherwood, V., Thomson, J., Hogan, S., Spender, L. C., South, A. P., Stratton, M., Chelala, C., Harwood, C. A., Proby, C. M., Leigh, I. M. (2018) The genomic landscape of cutaneous SCC reveals drivers and a novel azathioprine associated mutational signature. Nature Communications, 9, (doi: 10.1038/s41467-018-06027-1)
Purdie, K. J., Proby, C. M., Rizvi, H., Griffin, H., Doorbar, J., Sommerlad, M., Feltkamp, M. C., Meijden, E. V. d., Inman, G. J., South, A. P., Leigh, I. M., Harwood, C. A. (2018) The role of human papillomaviruses and polyomaviruses in BRAF-inhibitor induced cutaneous squamous cell carcinoma and benign squamoproliferative lesions. Frontiers in Microbiology, 9, (doi: 10.3389/fmicb.2018.01806)
Newcombe, A. A., Gibson, B. E.S., Keeshan, K. (2018) Harnessing the potential of epigenetic therapies for childhood acute myeloid leukemia. Experimental Hematology, 63, pp. 1-11. (doi: 10.1016/j.exphem.2018.03.008)
Morrissey, M. A., Williamson, A. P., Steinbach, A. M., Roberts, E. W., Kern, N., Headley, M. B., Vale, R. D. (2018) Chimeric antigen receptors that trigger phagocytosis. eLife, 7, (doi: 10.7554/eLife.36688.001)
Salomé, M., Magee, A., Yalla, K., Chaudhury, S., Sarrou, E., Carmody, R. J., Keeshan, K. (2018) A Trib2-p38 axis controls myeloid leukaemia cell cycle and stress response signalling. Cell Death and Disease, 9, (doi: 10.1038/s41419-018-0467-3)
Binnewies, M., Roberts, E. W., Kersten, K., Chan, V., Fearon, D. F., Merad, M., Coussens, L. M., Gabrilovich, D. I., Ostrand-Rosenberg, S., Hedrick, C. C., Vonderheide, R. H., Pittet, M. J., Jain, R. K., Zou, W., Howcroft, T. K., Woodhouse, E. C., Weinberg, R. A., Krummel, M. F. (2018) Understanding the tumor immune microenvironment (TIME) for effective therapy. Nature Medicine, 24, pp. 541-550. (doi: 10.1038/s41591-018-0014-x)
O'Connor, C., Yalla, K., Salome, M., Moka, H. A., Gómez Castañeda, E., Eyers, P. A., Keeshan, K. (2018) Trib2 expression in granulocyte-monocyte progenitors drives a highly drug resistant acute myeloid leukaemia linked to elevated Bcl2. Oncotarget, 9, pp. 14977-14992. (doi: 10.18632/oncotarget.24525)
Rose, A. M., Spender, L. C., Stephen, C., Mitchell, A., Rickaby, W., Bray, S., Evans, A. T., Dayal, J., Purdie, K. J., Harwood, C. A., Proby, C. M., Leigh, I. M., Coates, P. J., Inman, G. j. (2018) Reduced SMAD2/3 activation independently predicts increased depth of human cutaneous squamous cell carcinoma. Oncotarget, 9, pp. 14552-14566. (doi: 10.18632/oncotarget.24545)
Hayatigolkhatmi, K., Padroni, G., Su, W., Fang, L., Gómez-Castañeda, E., Hsieh, Y.-C., Jackson, L., Holyoake, T. L., Pellicano, F., Burley, G. A., Jørgensen, H. (2018) Investigation of a minor groove-binding polyamide targeted to E2F1 transcription factor in chronic myeloid leukaemia (CML) cells. Blood Cells, Molecules, and Diseases, 69, pp. 119-122. (doi: 10.1016/j.bcmd.2017.11.002)
Hurst, L. A., Dunmore, B. J., Long, L., Crosby, A., Al-Lamki, R., Deighton, J., Southwood, M., Yang, X., Nikolic, M. Z., Herrera, B., Inman, G. J., Bradley, J. R., Rana, A. A., Upton, P. D., Morrell, N. W. (2018) TNFα drives pulmonary arterial hypertension by suppressing the BMP type-II receptor and altering NOTCH signalling. Nature Communications, 8, (doi: 10.1038/ncomms14079)
Denton, A. E., Roberts, E. W., Fearon, D. T. (2018) Stromal cells in the tumor microenvironment. Springer
2017
Cammareri, P., Vincent, D. F., Hodder, M. C., Ridgway, R. A., Murgia, C., Nobis, M., Campbell, A. D., Varga, J., Huels, D. J., Subramani, C., Prescott, K. L.H., Nixon, C., Hedley, A., Barry, S. T., Greten, F. R., Inman, G. J., Sansom, O. J. (2017) TGFβ pathway limits dedifferentiation following WNT and MAPK pathway activation to suppress intestinal tumourigenesis. Cell Death and Differentiation, 24, pp. 1681-1693. (doi: 10.1038/cdd.2017.92)
Laing, A. A., Harrison, C. J., Gibson, B. E.S., Keeshan, K. (2017) Unlocking the potential of anti-CD33 therapy in adult and childhood acute myeloid leukaemia. Experimental Hematology, 54, pp. 40-50. (doi: 10.1016/j.exphem.2017.06.007)
Sarrou, E., Gibson, B., Keeshan, K. (2017) Absence of Mcm6 mimics haemopoietic ageing and has implications for leukaemic transformation. (doi: 10.1016/j.exphem.2017.06.134)
Phillips, T. J. et al. (2017) Treating the placenta to prevent adverse effects of gestational hypoxia on fetal brain development. Scientific Reports, 7, (doi: 10.1038/s41598-017-06300-1)
Ganugula, R., Arora, M., Jaisamut, P., Wiwattanapatapee, R., Jørgensen, H. G., Venkatpurwar, V. P., Zhou, B., Rodrigues Hoffmann, A., Basu, R., Guo, S., Ravi Kumar, M. N. V. (2017) Nano-curcumin safely prevents streptozotocin-induced inflammation and apoptosis in pancreatic ß-cells for effective management of type 1 diabetes mellitus. British Journal of Pharmacology, 174, pp. 2074-2084. (doi: 10.1111/bph.13816)
Carmody, R., Keeshan, K. (2017) The Tribble with APL: a new road to therapy. Cancer Cell, 31, pp. 612-613. (doi: 10.1016/j.ccell.2017.04.011)
Ben Batalla, I., Erdmann, R., Jorgensen, H., Mitchell, R., Ernst, T., von Amsberg, G., Schafhausen, P., Velthaus, J. L., Rankin, S., Clark, R. E., Koschmieder, S., Schultze, A., Mitra, S., Vandenberghe, P., Bru mmendorf, T. H., Carmeliet, P., Hochhaus, A., Pantel, K., Bokemeyer, C., Helgason, G. V., Holyoake, T. L., Loges, S. (2017) Axl blockade by BGB324 inhibits BCR-ABL tyrosine kinase inhibitor-sensitive and -resistant chronic myeloid leukemia. Clinical Cancer Research, 23, pp. 2289-2300. (doi: 10.1158/1078-0432.CCR-16-1930)
Eyers, P. A., Keeshan, K., Kannan, N. (2017) Tribbles in the 21st Century: the evolving roles of Tribbles pseudokinases in biology and disease. Trends in Cell Biology, 27, pp. 284-298. (doi: 10.1016/j.tcb.2016.11.002)
Ahmed, A., Wan, X., Mitxitorena, I., Lindsay, A. J., Pandolfi, P. P., McCaffrey, M. W., Keeshan, K., Chen, Y. H., Carmody, R. J. (2017) Regulation of NF-κB by PML and PML-RARα. Scientific Reports, 7, (doi: 10.1038/srep44539)
Wong, P. T., Roberts, E. W., Tang, S., Mukherjee, J., Cannon, J., Nip, A. J., Corbin, K., Krummel, M. F., Choi, S. K. (2017) Control of an unusual photo-claisen rearrangement in coumarin caged tamoxifen through an extended spacer. ACS Chemical Biology, 12, pp. 1001-1010. (doi: 10.1021/acschembio.6b00999)
Spender, L. C., Inman, G. J. (2017) Targeting BRAF-mutant tumours with TGFBR1 inhibitors. Aging, 9, pp. 5-6. (doi: 10.18632/aging.101169)
Tarafdar, A., Hopcroft, L. E.M., Gallipolli, P., Pellicano, F., Cassels, J., Hair, A., Korfi, K., Jørgensen, H. G., Vetrie, D., Holyoake, T. L., Michie, A. M. (2017) CML cells actively evade host immune surveillance through cytokine-mediated downregulation of MHC-II expression. Blood, 129, pp. 199-208. (doi: 10.1182/blood-2016-09-742049)
Rose, A. M., Sansom, O. J., Inman, G. J. (2017) Loss of TGF-β signaling drives cSCC from skin stem cells – More evidence. Cell Cycle, 16, pp. 386-387. (doi: 10.1080/15384101.2016.1259892)
Jackson, L., Gómez-Castañeda, E., Jørgensen, H. G., Hopcroft, L. E.M., Rogers, S., Holyoake, T. L., Huang, X. (2017) Oncogene addiction in chronic myeloid leukaemia. John Wiley & Sons, Ltd
2016
Spender, L. C., Ferguson, G. J., Liu, S., Cui, C., Girotti, M. R., Sibbet, G., Higgs, E. B., Shuttleworth, M. K., Hamilton, T., Lorigan, P., Weller, M., Vincent, D. F., Sansom, O. J., Frame, M., Dijke, P. t., Marais, R., Inman, G. J. (2016) Mutational activation of BRAF confers sensitivity to transforming growth factor beta inhibitors in human cancer cells. Oncotarget, 7, pp. 81995-82012. (doi: 10.18632/oncotarget.13226)
Chaudhury, S. S., Wheadon, H., Gibson, B., Keeshan, K. (2016) The Bone Marrow Niche Distinguishes Young and Old Leukemia.
Flint, T. R., Janowitz, T., Connell, C. M., Roberts, E. W., Denton, A. E., Coll, A. P., Jodrell, D. I., Fearon, D. T. (2016) Tumor-induced IL-6 reprograms host metabolism to suppress anti-tumor immunity. Cell Metabolism, 24, pp. 672-684. (doi: 10.1016/j.cmet.2016.10.010)
Scott, M. T., Korfi, K., Saffrey, P., Hopcroft, L. E.M., Kinstrie, R., Pellicano, F., Guenther, C., Gallipoli, P., Cruz, M., Dunn, K., Jorgensen, H. G., Cassels, J. E., Hamilon, A., Crossan, A., Sinclair, A., Holyoake, T. L., Vetrie, D. (2016) Epigenetic reprogramming sensitizes CML stem cells to combined EZH2 and tyrosine kinase inhibition. Cancer Discovery, 6, pp. 1248-1257. (doi: 10.1158/2159-8290.CD-16-0263)
O'Connor, C., Lohan, F., Campos, J., Ohlsson, E., Salome, M., Forde, C., Artschwager, R., Liskamp, R. M., Cahill, M. A., Kiely, P. A., Porse, B., Keeshan, K. (2016) The presence of C/EBPα and its degradation are both required for TRIB2 mediated leukaemia. Oncogene, 35, pp. 5272-5281. (doi: 10.1038/onc.2016.66)
Keeshan, K., Vieugué, P., Chaudhury, S., Rishi, L., Gaillard, C., LIang, L., Garcia, E., Nakumura, T., Omidvar, N., Kogan, S. C. (2016) Co-operative leukaemogenesis in acute myeloid leukaemia and acute promyelocytic leukaemia reveal C/EBPα as a common target of TRIB1 and PML/RARA. Haematologica, 101, pp. 1228-1236. (doi: 10.3324/haematol.2015.138503)
Cammareri, P. et al. (2016) Inactivation of TGFβ receptors in stem cells drives cutaneous squamous cell carcinoma. Nature Communications, 7, (doi: 10.1038/ncomms12493)
Liang, K. L., Paredes, R., Carmody, R., Eyers, P. A., Meyer, S., McCarthy, T. V., Keeshan, K. (2016) Human TRIB2 oscillates during the cell cycle and promotes ubiquitination and degradation of CDC25C. International Journal of Molecular Sciences, 17, (doi: 10.3390/ijms17091378)
Roberts, E. W., Broz, M. L., Binnewies, M., Headley, M. B., Nelson, A. E., Wolf, D. M., Kaisho, T., Bogunovic, D., Bhardwaj, N., Krummel, M. F. (2016) Critical role for CD103 + /CD141 + dendritic cells bearing CCR7 for tumor antigen trafficking and priming of T cell immunity in melanoma. Cancer Cell, 30, pp. 324-336. (doi: 10.1016/j.ccell.2016.06.003)
Laidlaw, K. M.E., Berhan, S., Liu, S., Silvestri, G., Holyoake, T. L., Frank, D. A., Aggarwal, B., Bonner, M. Y., Perrotti, D., Jorgensen, H. G., Arbiser, J. L. (2016) Cooperation of imipramine blue and tyrosine kinase blockade demonstrates activity against chronic myeloid leukemia. Oncotarget, 7, pp. 51651-51664. (doi: 10.18632/oncotarget.10541)
Chaudhury, S., Gibson, B., Keeshan, K. (2016) The Impact of Cellular Age on Leukaemic Transformation.
Sarrou, S., Gibson, B., Keeshan, K. (2016) Efficient Genome Editing of Mouse Hematopoietic Cells Using Crispr/Cas9 Technology.
Salomè, M., Keeshan, K. (2016) TRIB2 Regulates the Cell Cycle Under Stress Conditions in a Murine Cell Model of Leukaemia.
Yalla, K., O'Connor, C., Campos, J., Keeshan, K. (2016) TRIB2 Transformed GMP is the Myeloid Leukaemia Initiating Cell.
Languino, L. R., Singh, A., Prisco, M., Inman, G. J., Luginbuhl, A., Curry, J. M., South, A. P. (2016) Exosome-mediated transfer from the tumor microenvironment increases TGFβ signaling in squamous cell carcinoma. American Journal of Translational Research, 8, pp. 2432-2437.
Michie, A.M., Tarafdar, A., Gallipoli, P., Pellicano, F., Hopcroft, L., Korfi, K., Cassels, J., Jorgensen, H.J., Vetrie, D., Holyoake, T.L. (2016) CML Cells Actively Evade Host Immune Surveillance Through Cytokine-Mediated Downregulation of MHC-I I Expression. (doi: 10.1111/bjh.14019)
Jackson, L., Hopcroft, L.E.M., Rogers, S., Jorgensen, H., Pellicano, F., Wells, C., Mosbergen, R., Chen, T., Vetrie, D., Holyoake, T.L. (2016) Identifying Genes and Pathways Deregulated in Chronic Myeloid Leukaemia Stem Cells Through Meta-Analysis of Transcriptomic Data. (doi: 10.1111/bjh.14019)
Gómez-Castañeda, E., Hopcroft, L.E.M., Rogers, S., Jorgensen, H.G., Pellicano, F., Vetrie, D., Copland, M., Grimmond, S., Holyoake, T.L. (2016) Uncovering the BCR-ABL1 Tyrosine Kinase Independent Signature in Chronic Myeloid Leukaemia Stem Cells. (doi: 10.1111/bjh.14019)
Headley, M. B., Bins, A., Nip, A., Roberts, E. W., Looney, M. R., Gerard, A., Krummel, M. F. (2016) Visualization of immediate immune responses to pioneer metastatic cells in the lung. Nature, 531, pp. 513-517. (doi: 10.1038/nature16985)
Liang, K. L., O'Connor, C., Veiga, J. P., McCarthy, T. V., Keeshan, K. (2016) TRIB2 regulates normal and stress-induced thymocyte proliferation. Cell Discovery, 2, (doi: 10.1038/celldisc.2015.50)
Spender, L.C., Inman, G.J. (2016) Fatal attractions? Correlations of CXCL12-CXCR4-CXCR7 expression with disease progression in melanoma and Kaposi sarcoma. British Journal of Dermatology, 175, pp. 1140-1141. (doi: 10.1111/bjd.15136)
Zhang, B., Chu, S., Agarwal, P., Campbell, V. L., Hopcroft, L., Jørgensen, H. G., Lin, A., Gaal, K., Holyoake, T. L., Bhatia, R. (2016) Inhibition of interleukin-1 signaling enhances elimination of tyrosine kinase inhibitor treated CML stem cells. Blood, 128, pp. 2671-2682. (doi: 10.1182/blood-2015-11-679928)
Roberts, E.W., Denton, A.E., Fearon, D.T. (2016) Roles of stromal cells in the immune system. Elsevier
Harwood, C. A., Proby, C. M., Inman, G. J., Leigh, I. M. (2016) The promise of genomics and the development of targeted therapies for cutaneous squamous cell carcinoma. Acta Dermato Venereologica, 96, pp. 3-16. (doi: 10.2340/00015555-2181)
We have three fundamental questions that we are trying to answer in the laboratory using both in vitro cell biological and in vivo techniques coupled with analysis of primary patient tumour material:
1) How do TGFβ/BMP act as tumour suppressors and how do tumour cells avoid this?
2) How do TGFβ/BMP act on tumour cells to promote cancer progression?
3) When and where do these events occur? Our ultimate goals are to develop therapeutics that selectively target the pro-oncogenic actions of these cytokines and to identify patient selection criteria for their deployment.
It has previously been shown that both TGFBR1 and TGFBR2 are frequently mutationally inactivated in human cutaneous squamous cell carcinoma (cSCC) and that combined deletion of TGFBR1 coupled with activation of the MAPK pathway is sufficient to drive rapid invasive cSCC formation from the Lgr5+ve hair follicle bulge stem cells in the mouse.
This has led us into studies profiling the molecular landscape of human cSCC. Our recent whole exome sequencing studies have identified driver genes and pathways that are implicated in cSCC progression and we are currently building upon these observations using whole genome sequencing and RNAseq analysis.
The mammalian skin is an excellent model system to functionally interrogate fundamental cell biological processes required for epithelial homeostasis. The intricate and dynamic relationship between cell adhesion, migration, and basement membrane organisation, in the context of the local immune microenvironment, is critical to normal skin development and healthy tissue function. Gaining insight into the complex interplay between these processes allows us to understand how they go awry in pathological conditions such as inflammatory skin disorders and cancer. There are research programs into two different aspects of skin and involvement with tumour involvement: - Epithelial-immune metabolic crosstalk and inflammatory skin diseases:
This research focuses on understanding the crosstalk between epithelial cells, immune cells and the extracellular matrix (ECM) in maintaining homeostasis and exploring the metabolic drivers of inflammatory skin diseases and cancer - Stem cell homoeostasis and nuclear mechanosensing: This research focuses on focuses on understanding the mechanical underpinning of the crosstalk between the ECM and cell junctions with the cytoskeleton and nucleus in maintaining stem cell quiescence and the role altered nuclear mechanotransduction in driving diseases such as metastatic cancers.
Tumour immunotherapy, most notably checkpoint blockade therapy, has produced remarkable benefits for patients with cancers that previously had poor outcomes.
Blockade of the T cell inhibitory checkpoint molecules CTLA-4 and PD-1 has led to dramatic remissions that have lasted more than a decade in many patients. However, these responses are unfortunately restricted to only a subset of patients.
Our work broadly aims to understand how the immune response to cancer is generated so as to understand what may limit the quality or quantity of that response. In this way, we hope to find new means of augmenting the response to immunotherapy in a broader subset of patients.
T cell responses begin in the lymph node, where they are directed by signals received from the peripheral tissue, where the challenge occurs. During responses to viruses, numerous signals drive re-organisation of the lymph node and choreograph a highly regulated process by which appropriate T cell activation can occur. It has been observed that the tumour-draining lymph node is improperly activated, suggesting priming may be less efficient against the tumour. Indeed vaccination, which improves priming, can improve immunotherapeutic approaches in mice, suggesting this as a viable approach. The aim is to determine how the periphery communicates with and educates the lymph node during an efficient immune response and how this is subverted in the tumour setting. Of particular interest, is the role of different dendritic cell subsets in organising and directing efficient immune responses in the lymph node, and how these are manipulated by the tumour microenvironment. In this way, we aim to discover signals by which we can convert the lymph node microenvironment into a more effective site of immune priming to augment existing immunotherapeutic responses.