Bob Hancock

Contact Information
Centre for Microbial Diseases and Immunity Research
Lower Mall Research Station
#232, 2259 Lower Mall, Vancouver, BC, Canada, V6T 1Z4
Tel: 1-604-822-2682
Fax: 1-604-827-5566
Email: bob@hancocklab.com
Lab Website: http://cmdr.ubc.ca/bobh/

Current Positions

  • Professor, Microbiology and Immunology, Science.
  • Director, Centre for Microbial Diseases and Immunity Research.
  • UBC Killam Professor of Microbiology & Immunology.
  • Canada Research Chair in Health and Genomics.

Research Interests

Infectious diseases influence all areas of human endeavor. They are responsible for a third of all deaths on the planet, are currently the third leading cause of human deaths in North America, and also have a major impact on agriculture and food safety. However current therapeutic approaches based on antibiotics are under severe threat due to antibiotic resistance and the dearth (and ineffectiveness) of antibiotic discovery programs worldwide.

The Hancock laboratory is engaged in three basic types of research to address this growing problem; understanding the mechanism of action of cationic host defence (antimicrobial) peptides and their role as modulators of innate immunity (including basic functional genomic studies to define the innate immunity network in blood cells); the development of novel therapeutics based on the immunomodulatory and antibiotic activities of host defence peptides; and investigating the functional genomics of a prominent nosocomial pathogen, Pseudomonas aeruginosa, with specific reference to antibiotic resistance and the regulation of resistance and virulence. It is situated in approximately 8000 square feet of lab bench space and 2000 square feet of office space at the Lower Mall Research Station on campus and is well equipped, technologically diverse and well funded.

Selected Publications

  1. Wang, D., Y. Shen, R.E.W. Hancock, J. Ma, and M. Haapsalo. Antimicrobial effect of Peptide DJK-5 used alone or mixed with EDTA on Mono- and multispecies biofilms in dentin canals. J. Endodontics, In Press, pii: S0099-2399(18)30514-4. https://doi.org/10.1016/j.joen.2018.07.018.
  2. Forbester, J.L., E.A. Lees, D. Goulding, S. Forrest, A. Yeung, A. Speak, S. Clare, E. Coomber, S. Mukhopadhyay, J. Kraiczy, F. Schreiber, T.D. Lawley, R.E.W. Hancock, H.H. Uhlig, M. Zilbauer, F. Powrie and G. Dougan. 2018. Interleukin-22 promotes phagolysosomal fusion to induce protection against Salmonella enterica Typhimurium in human epithelial cells. Proc. Natl. Acad. Sci. USA, in press https://doi.org/10.1073/pnas.1811866115.
  3. Sun, E., S. Liua, and R.E.W. Hancock. 2018. Surfing motility: a conserved yet diverse adaptation among motile bacteria. J. Bacteriol., 62(9). pii: e00848-18. Covered by Nature Reviews Microbiology.
  4. Hinshaw, S.J., A.H.Y. Lee, E.E. Gill, and R.E.W. Hancock. 2018. MetaBridge: enabling network-based integrative analysis via direct protein interactors of metabolites. Bioinformatics 34:3225-3227.
  5. Dostert, M., C.R. Belanger, and R.E.W. Hancock. 2018. Design and assessment of anti-biofilm peptides: Steps toward clinical application. J. Innate Immun. 22:1-12.
  6. Sun, E., E.E. Gill, R. Falsafi, A. Yeung, S. Liu, and R.E.W. Hancock. 2018. Broad-spectrum adaptive antibiotic resistance associated with Pseudomonas aeruginosamucin-dependent surfing motility. Antimicrob. Agents Chemother. 62:e00848-18.
  7. Wolfmeier, H., S.C. Mansour, L.T. Liu, D. Pletzer, A. Draeger, E.B. Babiychuk and R.E.W. Hancock. 2018. Liposomal therapy attenuates dermonecrosis induced by community-associated methicillin-resistant Staphylococcus aureus by targeting α-type phenol soluble modulins and α-hemolysin. eBiomedicine 33:211-217.
  8. Windham, I.H., S.L. Servetas, J.M. Whitmire, D. Pletzer, R.E.W. Hancock, and D.S. Merrell. 2018. Biofilm formation by Helicobacter pylori is differentially affected by common culture conditions and protein plays a central role in the biofilm matrix. Appl. Environ. Microbiol. 84(14):pii:e00391-18.
  9. Mwanza-Lisulo, M., M.S. Chomba, M. Chama, E.C. Besa, E. Funjika, K. Zyambo, R. Banda, M. Imikendu, S. Sianongo, R.E.W. Hancock, A. Lee, R. Chilengi, A.J. Stagg, B. Namangala, and P.M. Kelly. 2018. Retinoic acid elicits a coordinated expression of gut homing markers on T lymphocytes of Zambian men receiving oral Vivotif, but not Rotarix, Dukoral or OPVERO vaccines. Vaccine 36(28):4134-4141.
  10. Ma, L., S. Feng, C. de la Fuente-Nunez, R.E.W. Hancock, and X. Lu. 2018. Development of molecularly imprinted polymers to block quorum sensing and inhibit bacterial biofilm formation. ACS Applied Materials & Interfaces 10(22):18450-18457.
  11. Haney, E.F., K.C. Wuerth, N. Rahanjam, N.S. Nikouei, A. Ghassemi A. Boey and R.E.W. Hancock. 2018. Identification of an IDR peptide formulation candidate that prevents peptide aggregation and retains immunomodulatory activity. Peptide Science 8(2):pii:E29. https://doi.org/10.1002/pep2.24077.
  12. Pletzer, D., S.C. Mansour, and R.E.W. Hancock. 2018. Synergy between conventional antibiotics and anti-biofilm peptides in a murine, sub-cutaneous abscess model caused by recalcitrant ESKAPE pathogens. PLoS Pathogens 14(6):e1007084. Featured Research Article at PLoS Pathogens.
  13. Haney,E.F., M.J. Trimble, J.T. Cheng, Q. Vallé and R.E.W. Hancock. 2018. Critical assessment of methods to quantify biofilm growth and evaluate antibiofilm activity of host defence peptides. Biomolecules 8:29.
  14. Porto W.F., L. Irazazabala, E.S.F. Alves, S.M. Ribeiro, C.O. Matos, Á.S. Pires, I.C.M. Fensterseifer, V.J. Miranda, E.F. Haney, V. Humblot, M. Torres, R.E.W. Hancock, L.M. Liao, A. Ladram, T. Lu, C. de la Fuente-Núñez, and O.L. Franco. 2018. In silico optimization of a guava antimicrobial peptide enables combinatorial exploration for peptide design. Nature Commun. 9:1490. doi: 10.1038/s41467-018-03746-3. Covered by the international press.
  15. Gill, E.E., L.S. Chan, G.L. Winsor, N. Dobson, R. Lo, S.J. Ho Sui, B.K. Dhillon, P.K. Taylor, R. Shrestha, C. Spencer, R.E.W. Hancock, P.J. Unrau, F.S.L. Brinkman. 2018. High-throughput detection of RNA processing in bacteria. BMC Genomics 19:223.
  16. Marin-Luevanoa, P., V. Trujilloa, A. Rodriguez-Carlosa, I. González-Curielc, J.A. Enciso-Moreno, R.E.W. Hancock, and B. Rivas-Santiago. 2018. Induction by Innate Defence Regulator peptide 1018 of pro-angiogenic molecules and endothelial cell migration in a high glucose environment. Peptides 101:135-144.
  17. Gellatly, S.L., M Bains, E.B.M. Breidenstein, J. Strehmel, F. Reffuveille, P.K. Taylor, A.T.Y. Yeung, J. Overhage, and R.E.W. Hancock. 2018. Novel roles for two-component regulatory systems in cytotoxicity and virulence-related properties in Pseudomonas aeruginosa. AIMS Microbiol. 4:173-191.
  18. Haney, E.F., Y. Brito-Sánchez, M.J. Trimble, S.C. Mansour, A. Cherkasov, and R.E.W. Hancock. 2018. Computer-aided discovery of peptides that specifically attack bacterial biofilms. Sci. Reports 8:1871.
  19. Wolfmeier, H., D. Pletzer, S.C. Mansour and R.E.W. Hancock. 2018. New perspectives in biofilm eradication. ACS Infectious Diseases 4(2):93-106.
  20. Wu, B.C., A.H. Lee, and R.E.W. Hancock. 2017. Mechanisms of the innate defense regulator peptide-1002 anti-inflammatory activity in a sterile inflammation mouse model. J. Immunol. 199:3592-3603.
  21. Qadi, M., S. Izquierdo-Rabassa, M. Mateu Borrás, A. Doménech-Sánchez, C. Juan, J.B. Goldberg, R.E.W. Hancock, and S. Albertí. 2017. Sensing Mg2+contributes to the resistance of Pseudomonas aeruginosato complement-mediated opsonophagocytosis. Environ. Microbiol. 19:4278-4286.
  22. Wuerth, K.C., R. Falsafi, and R.E.W. Hancock. 2017. Synthetic host defense peptide IDR-1002 reduces inflammation in Pseudomonas aeruginosalung infection. PLoS One 12:e0187565.
  23. Wang, D., Y. Shen, J. Ma, R.E.W. Hancock, and M. Haapasalo. 2017. Antibiofilm effect of D-enantiomeric peptide alone and combined with EDTA in vitro. J. Endodontics. 43:1862-1867.
  24. Pletzer, D., H. Wolfmeier, M. Bains and R.E.W. Hancock. 2017. Synthetic peptides to target stringent response-controlled virulence in a Pseudomonas aeruginosamurine cutaneous infection model. Front. Microbiol. 8:1867.
  25. Haney, E.F. B. Wu, K. Lee, A.L. Hilchie and R.E.W. Hancock. 2017. Aggregation and its influence on the immunomodulatory activity of synthetic innate defense regulator peptides. Cell Chem Biol 24:969–980.e4.
  26. Taylor, P.K., A.T.M. Van Kessel, A. Colavita, R.E.W. Hancock, and T.-F. Mah. 2017. A novel small RNA is important for biofilm formation and pathogenicity in Pseudomonas aeruginosa. PLoS One 12: e0182582.
  27. de Freitas,C.G., S.M. de Freitas Lima, M. de Souza Freire, A.P. de Castro Cantuária, N.G. de Oliveira Júnior, T.S. dos Santos, J. dos Santos Folha, T.M.B. Rezende, A.M. Nicolad, Albuquerque, C. de La Fuente-Núñez, S.M. Ribeiro, S.C. Dias, R.E.W. Hancock, O.L. Franco, and M.S.S. Felipe. 2017. An immunomodulatory peptide that confers protection in an experimental candidemia murine model. Antimicrob. Agents Chemother. 61:e02518-16.
  28. Verderosa, A.D., C. de la Fuente-Núñez, S.C. Mansour, J. Cao, T.K. Lu, R.E.W. Hancock, and K.E. Fairfull-Smith. 2017. Ciprofloxacin-nitroxide hybrids with potential for biofilm control. Eur J Med Chem. 138:590-601.
  29. Belanger, C.R.,C. Mansour, Pletzer, D., and R.E.W. Hancock. 2017. Alternative strategies for the study and treatment of clinical bacterial biofilms. Emerg. Topics Life Sci. 1:41-53. doi:10.1042/ETLS20160020.
  30. Yeung, A.T.Y., C. Hale, A.H. Lee, E. E. Gill, W. Bushell, D. Parry-Smith, D. Goulding, D. Pickard, T. Roumeliotis, J. Choudhary, N. Thomson, W.C. Skarnes, G. Dougan and R.E.W. Hancock. 2017. Exploiting induced pluripotent stem cell-derived macrophages to unravel key host factors influencing Chlamydia trachomatis Nature Communications 8:15013. Covered by Nature Microbiology, etc.

Education

  • Postdoctoral, Berkeley (1977-78).
  • Postdoctoral, Tübingen (1975-77).
  • Ph.D., Adelaide, 1974.