GENERATION AND VALIDATION OF NANOBODIES TARGETING BRAIN TUMORS WITH ABERRANT EXPRESSION OF EPIDERMAL GROWTH FACTOR RECEPTOR AND HUMAN EPIDERMAL GROWTH FACTOR RECEPTOR 2

Heleen Hanssens Janik Puttemans Serge Muyldermans
Persbericht

Hersentumoren genezen met kamelenbloed?

Hersentumoren. We zijn ze liever kwijt dan rijk. Maar toch is het één van de  meest voorkomende kankers bij kinderen jonger dan 15 jaar. Behandeling van dit soort tumoren is moeilijk en meestal is een volledige genezing niet mogelijk. Dat maakt dat de diagnose van een hersentumor nog steeds een quasi doodsvonnis is anno 2018.

Er bestaan twee soorten hersentumoren: primaire hersentumoren, die ontstaan uit gezonde hersencellen, en secundaire hersentumoren, die uitzaaiingen zijn van andere kankersoorten, zoals de belangrijke borstkankers. De behandeling van beide soorten bestaat uit een keuze uit of combinatie van chirurgie, bestraling en chemotherapie waarbij kankerbestrijdende middelen worden toegediend. De laatste decennia wordt er veel onderzoek gedaan naar deze specifieke geneesmiddelen, en ook naar methodes om kankercellen te onderscheiden van gezonde cellen.

Als ons lichaam in contact komt met een indringer, zoals een bacterie of virus, treedt ons natuurlijk afweermechanisme, het immuunsysteem, in werking en worden er antilichamen geproduceerd: moleculen die in staat zijn om de indringers te herkennen en te elimineren. Kankercellen zijn in wezen ook indringers, maar omdat ze zo sterk gelijken op gezonde cellen, worden ze door ons immuunsysteem niet herkend als lichaamsvreemd en dus ook met rust gelaten. In laboratoria heeft men nu technieken ontwikkeld om toch antilichamen aan te maken, die specifiek kankercellen kunnen herkennen en binden, en die via het bloed aan de patiënt kunnen worden toegediend. Dit is het basisprincipe van de immuuntherapie, een techniek die bij verschillende kankers zijn nut en efficiëntie al heeft bewezen. 

Bij hersentumoren ligt dit moeilijker en slaat deze techniek niet aan, en daar is reden voor: de bloedhersenbarrière. Dat is een soort microfilter die onze hersenen beschermt voor mogelijk schadelijke stoffen of indringers. Gewone antilichamen zijn grote moleculen die zeer moeilijk door deze filter geraken. Anderzijds heeft onderzoek uitgewezen dat de filter in geval van een tumor soms niet meer goed werkt en antilichamen de tumor toch zouden kunnen vinden. In dat geval echter, zorgt het gezond hersenweefsel ervoor zorgt dat die antilichamen niet meer goed werken. Pech dus. Of toch niet?

25 jaar geleden werd er aan de Vrije Universiteit Brussel een toevallige en bijzondere ontdekking gedaan in het bloed van kameelachtigen. Deze dieren maken naast de gewone antilichamen ook een speciale soort antilichamen aan: de zware-ketenantilichamen. Deze zijn kleiner dan de gewone antilichamen, die bestaan uit zware én lichte ketens, en hebben bovendien een ander herkenningsgedeelte. Dit deeltje wordt ook nanobody genoemd, en kan gebruikt worden als een aparte molecule om indringers te herkennen en te binden. Een nanobody is ongeveer tien keer kleiner dan een gewoon antilichaam en onderzoek heeft al verschillende voordelen kunnen aantonen ten opzichte van deze laatste. Zo zijn ze sneller in het vinden en bereiken van indringers in het lichaam, en ze worden ook sneller uit het lichaam verwijderd via de urine.

Het doel in deze masterthesis was het onderzoeken naar de capaciteiten van nanobodies om hersentumoren te bereiken, en om na te gaan of ze er meer geschikt voor zouden zijn dan de standaard antilichamen. Mijn onderzoek heeft zich voornamelijk gericht op hersenmetastasen van een veelvoorkomende borstkanker, in een testsetting met muizen. Zowel antilichamen als nanobodies werden gemerkt met een radioactieve stof. De radioactieve straling kunnen we meten wat ons toelaat om te berekenen hoeveel nanobody in de tumor geraakt.

Met dit onderzoek hebben we kunnen aantonen dat zowel antilichamen als nanobodies in staat zijn om de hersentumor te bereiken. Echter wel met wezenlijke verschillen. Antilichamen kunnen veel meer radioactieve straling in de tumor brengen. Maar omwille van hun grootte bewegen ze zich veel trager doorheen het lichaam en bereiken ze de tumor veel later. Nanobodies daarentegen zijn veel vinniger: ze zijn al voldoende meetbaar in de tumor na een uur, terwijl dit voor de antilichamen drie à vier dagen duurt om een maximum dosis te bereiken. Dit opent perspectieven naar de behandeling van deze tumoren. Men kan aan het nanobody een therapeutisch type radioactiviteit koppelen, zodat het de kankercel waaraan het zich hecht kan bestralen en schade toebrengen. Doet met dit met antilichamen, dan zal er ook schade aangericht worden aan gezonde weefsels, waar het antilichaam “treuzelt” omwille van zijn grootte en bijhorende traagheid. Er is echter een maar. De kleine vinnige nanobodies passeren ook vlot de nierfilter en worden dus ook snel via de urine uitgescheiden, hetgeen nierschade zou kunnen veroorzaken bij gebruik van radioactiviteit. Momenteel wordt er dan ook veel onderzoek gedaan naar een manier om de nierschade te verminderen om op die manier een ideaal type molecule te ontwikkelen voor de gerichte radiotherapie van tumoren in de hersenen.

Een andere belangrijke moeilijkheid in de behandeling van hersentumoren is de chirurgische verwijdering. Het is namelijk uitermate moeilijk om gezond hersenweefsel te onderscheiden van kankercellen in het brein. En aangezien hersenen zo delicaat en complex zijn, kunnen dokters niet zomaar meer weefsel wegnemen om zeker te zijn dat de hele tumor is verwijderd. Nanobodies kunnen ook hier een oplossing bieden. Door er een soort fluorescerende stof aan te binden, kan men de slechte cellen laten oplichten, zodat hersentumoroperaties veel preciezer kunnen gebeuren.

Samengevat hebben we in deze masterthesis een aantal veelbelovende mogelijkheden van nanobodies in het domein van de neuro-oncologie aangetoond, zowel op het vlak van meer gerichte radiotherapie, als op het vlak van beeldgeleide operaties. Deze toevallig ontdekte kleine moleculen uit kamelenbloed zouden daarom wel eens een grote stap kunnen betekenen in de behandeling van hersentumoren, een domein waarin de vraag naar meer efficiënte therapieën enorm is.

Bibliografie
  • Abbott, N. J. et al., 2010. Structure and function of the blood–brain barrier. Neurobiology of Disease, Volume 37, pp. 13-25.
  • Agarwala, S. S. & Kirkwood, J. M., 2000. Temozolomide, a novel alkylating agent with activity in the central nervous system, may improve the treatment of advanced metastatic melanoma. The Oncologist, Volume 5, pp. 144-151.
  • Aizer, A. A. & Lee, E. Q., 2018. Brain metastases. Neurologic Clinics, Volume 36, pp. 557- 577. American Cancer Society, 2015. Breast cancer facts & figures 2015-2016.. [Online] Available at: https://www.cancer.org/content/dam/cancer-org/research/cancer-facts-and… breast-cancer-facts-and-figures/breast-cancer-facts-and-figures-2015-2016.pdf [Accessed 19 August 2018].
  • An, Z. et al., 2018. Epidermal growth factor receptor and EGFRvIII in glioblastoma: signaling pathways and targeted therapies. Oncogene, Volume 37, pp. 1561-1575.
  • Assi, H., Candolfi, M., Lowenstein, P. R. & Castro, M. G., 2012. Rodent glioma models: intracranial stereotactic allografts and xenografts. Neuromethods, Volume 77, pp. 229-243.
  • Bachelot, T. et al., 2013. Lapatinib plus capecitabine in patients with previously untreated brain metastases from HER2-positive metastatic breast cancer (LANDSCAPE): a singlegroup phase 2 study. The Lancet Oncology, Volume 14, pp. 64-71.
  • Badr, C. E., 2014. Bioluminescence imaging: basics and practical limitations. Methods in Molecular Biotechnology, Volume 1098, pp. 1-18.
  • Becker, A. et al., 2001. Receptor-targeted optical imaging of tumors with near-infrared fluorescent ligands. Nature Biotechnology, Volume 19, pp. 327-331.
  • Bendell, J. et al., 2003. Central nervous system metastases in women who receive trastuzumab-based therapy for metastatic breast carcinoma. Cancer, Volume 15, pp. 2972- 2977.
  • Berghoff, A. S. & Preusser, M., 2018. New developments in brain metastases. Therapeutic Advances in Neurological Disorders, Volume 11, pp. 1-14.
  • Bruce, J. N., Houseplan, E. M. & Kennedy, B., 2009. Glioblastoma multiforme, New York, New York, USA: Columbia University College of Physicians and Surgeons. REFERENCES 98
  • Bruce, J. N. & Kennedy, B. M., 2017. Glioblastoma multiforme - prognosis, Chicago, Illinois, USA: Medscape.
  • Caljon, G. et al., 2012. Using microdialysis to analyse the passage of monovalent nanobodies through the blood–brain barrier. Brittish Journal of Pharmacology, Volume 165, pp. 2341- 2353.
  • Connell, J. et al., 2013. Selective permeabilization of the blood-brain barrier at sites of metastasis. Journal of the National Cancer Institute, Volume 105, pp. 1634-1643.
  • Conrath, K. et al., 2001. Beta-lactamase inhibitors derived from single-domain antibody fragments elicited in the camelidae. Antimicrobial Agents and Chemotherapy, Volume 45, pp. 2807-2812.
  • Dasgupta, A. et al., 2016. Ultrasound-mediated drug delivery to the brain: principles, progress and prospects. Drug Discovery Today: Technology, Volume 20, pp. 41-48.
  • De Genst, E. et al., 2006. Molecular basis for the preferential cleft recognition by dromedary heavy-chain antibodies. Proceedings of The National Academy of Sciences of the USA, Volume 103, pp. 4586-4591.
  • De Vos, J., 2014. Targeting the plaques with camelid cdAbs binding LOX-1. In: Non-invasive in vivo imaging of atherosclerotic plaques using camelid single-domain antibody fragments as molecular tracers. Brussels, Belgium: Vrije Universiteit Brussel, pp. 101-121.
  • De Vos, J., Devoogdt, N., Lahoutte, T. & Muyldermans, S., 2013. Camelid single-domain antibody-fragment engineering for (pre)clinical in vivo molecular imaging applications: adjusting the bullet to its target. Expert opinion on Biological Therapy, Volume 13, pp. 1149- 1160.
  • Debie, P. et al., 2017. Effect of dye and conjugation chemistry on the biodistribution profile of near-infrared-labeled nanobodies as tracers for image-guided surgery. Molecular Pharmaceutics, Volume 14, pp. 1145-1153.
  • Debie, P. et al., 2018. Improved debulking of peritoneal tumor implants by near-infrared fluorescent nanobody image guidance in an experimental mouse model. Molecular Imaging and Biology, Volume 20, pp. 361-367.
  • Dekempeneer, Y. et al., 2016. Targeted alpha therapy using short-lived alpha-particles and the promise of nanobodies as targeting vehicle. Expert Opinion on Biological Therapy, Volume 16, pp. 1035-1047.
  • D'Huyvetter, M. et al., 2014a. Targeted radionuclide therapy with a 177Lu-labeled anti-HER2 nanobody. Theranostics, Volume 4, pp. 108-720.
  • D'Huyvetter, M. et al., 2014b. Radiolabeled nanobodies as theranostic tools in targeted radionuclide therapy of cancer. Expert Opinion on Drug Delivery, Volume 11, pp. 1939-1954.
  • Dick Jr., L. W., Kim, C., Qiu, D. & Cheng, K.-C., 2007. Determination of the origin of the Nterminal pyro-glutamate variation in monoclonal antibodies using model peptides. Biotechnology and Bioengineering, Volume 97, pp. 544-553.
  • Dijkers, E. C. et al., 2010. Biodistribution of 89Zr-trastuzumab and PET Imaging of HER2- Positive Lesions in Patients With Metastatic Breast Cancer. Clinical Pharmacology and Therapeutics, Volume 87, pp. 586-592.
  • Ding, L. et al., 2015. Small sized EGFR1 and HER2 specific bifunctional antibody for targeted cancer therapy. Theranostics, Volume 5, pp. 378-398.
  • Dumoulin, M. et al., 2002. Single-domain antibody fragments with high conformational stability. Protein Science, Volume 11, p. 500–515.
  • Fan, C.-H., Lin, C.-Y., Liu, H.-L. & Yeh, C.-K., 2017. Ultrasound targeted CNS gene delivery for Parkinson's disease treatment. Journal of Controlled Release, Volume 261, pp. 246-262.
  • Ferlay, J. et al., 2015. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. International journal of Cancer, Volume 136, p. 359–386.
  • Fleetwood, F. et al., 2013. Surface display of a single-domain antibody library on Grampositive bacteria. Cellular and Molecular Life Sciences, Volume 70, pp. 1081-1093. Food and Drug Administration, 2012. accessdata. [Online] Available at: https://www.accessdata.fda.gov/drugsatfda_docs/bla/2012/103792orig1s527… [Accessed 17 8 2018].
  • Furnari, F. B., Cloughesy, T. F., Cavenee, W. K. & Mischel, P. S., 2015. Heterogeneity of epidermal growth factor receptor signalling networks in glioblastoma. Nature Reviews Cancer, Volume 15, pp. 302-310.
  • Gainkam, L. O. T. et al., 2008. Comparison of the biodistribution and tumor targeting of two 99m Tc-labeled anti-EGFR nanobodies in mice, using pinhole SPECT/Micro-CT. Journal of Nuclear Medicine, Volume 49, pp. 788-795.
  • Gancberg, D. et al., 2002. Comparison of HER-2 status between primary breast cancer and corresponding distant metastatic sites. Annals of Oncology, Volume 13, pp. 1036-1043.
  • Gan, H. K., Cvrljevic, A. N. & Johns, T. G., 2013. The epidermal growth factor receptor variant III (EGFRvIII): where wild things are altered. The FEBS Journal, Volume 280, pp. 5350-5370.
  • Garrett, T. P. et al., 2002. Crystal structure of a truncated epidermal growth factor receptor extracellular domain bound to transforming growth factor α. Cell, Volume 110, pp. 763-773.
  • Gedeon, P. C. et al., 2014. An EGFRvIII-targeted bispecific T-cell engager overcomes limitations of the standard of care for glioblastoma. Expert Review of Clinical Pharmacology, Volume 6, pp. 375-386.
  • Gorovits, B. & Krinos-Fiorotti, C., 2013. Proposed mechanism of off-target toxicity for antibody–drug conjugates driven by mannose receptor uptake. Cancer Immunology Immunotherapy, Volume 62, pp. 217-233.
  • Hamers-Casterman, C. et al., 1993. Naturally occurring antibodies devoid of light chains. Nature, Volume 363, p. 446–448.
  • Han, W. et al., 2006. Hypophosphorylation of residue Y1045 leads to defective downregulation of EGFRvIII. Cancer Biology and Therapy, Volume 5, pp. 1361-1368.
  • Hegi, M. et al., 2005. MGMT gene silencing and benefit from temozolomide in glioblastoma. The New England journal of Medicine, Volume 352, pp. 997-1003.
  • Henriksson, R., Asklund, T. & Poulsen, H. S., 2011. Impact of therapy on quality of life, neurocognitive function and their correlates in gliobastoma multiforme: a review. Journal of Neuro Oncology, Volume 104, pp. 639-646.
  • Hicks, D. G. & Kulkarni, S., 2008. HER2+ breast cancer: review of biologic relevance and optimal use of diagnostic tools. American Society for Clinical Pathology, Volume 129, pp. 263-273.
  • Huang, H. et al., 1997. The enhanced tumorigenic activity of a mutant epidermal growth factor receptor common in human cancers is mediated by threshold levels of constitutive tyrosine phosphorylation and unattenuated signaling. The Journal of Biological Chemistry, Volume 272, pp. 2927-2935.
  • Huang, L. et al., 2008. SPECT imaging with 99mTc-labeled EGFR-specific nanobody for in vivo monitoring of EGFR expression. Molecular Imaging and Biology, Volume 10, pp. 167- 175.
  • Huang, P. H., Xu, A. M. & White, F. M., 2009. Oncogenic EGFR signaling networks in glioma. Science Signaling, Volume 2, p. re6. Hussack, G. et al., 2011. Engineered single-domain antibodies with high protease resistance and thermal stability. PLoS ONE, Volume 6, p. e28218.
  • Iqbal, N. & Iqbal, N., 2014. Human epidermal growth factor receptor 2 (HER2) in cancers: overexpression and therapeutic implications. Molecular Biology International, Volume 2014, p. e852748.
  • Iqbal, U. et al., 2010. Molecular imaging of glioblastoma multiforme using anti-insulin-like growth factor-binding protein-7 single-domain antibodies. British Journal of Cancer, Volume 103, pp. 1606-1616.
  • Jain, A. K. et al., 2007. Carbon nanotubes and their toxicity. Nanotoxicology, Volume 1, pp. 167-197. Johnson, D. R. & O'Neill, B. P., 2012. Glioblastoma survival in the United States before and during the temozolomide era. Journal of Neuro Oncology, Volume 107, p. 359–364.
  • Johnson, L. et al., 2015. Rational development and characterization of humanized anti-EGFR variant III chimeric antigen receptor T cells for glioblastoma. Science Translational Medicine, Volume 7, p. 275ra22.
  • Jones, M. L. et al., 2016. Targeting membrane proteins for antibody discovery using phage display. Nature Scientific Reports, Volume 6, p. 26240.
  • Kabraji, S. et al., 2018. Drug resistance in HER2-positive breast cancer brain metastases: blame the barrier or the brain?. Clinical Cancer Research, Volume 24, pp. 1795-1804.
  • Keyaerts, M. et al., 2016. Phase I study of 68Ga-HER2-nanobody for PET/CT assessment of HER2 expression in breast carcinoma. Journal of Nuclear Medicine, Volume 57, pp. 27-33.
  • Kluba, C. A. & Mindt, T. L., 2013. Click-to-chelate: development of technetium and rhenium-tricarbonyl labeled radiopharmaceuticals. Molecules, Volume 18, pp. 3206-3226.
  • Kodack, D. P. et al., 2015. Emerging strategies for treating brain metastases from breast cancer. Cancer Cell, Volume 27, pp. 163-175.
  • Krasniqi, A. et al., 2017. Theranostic radiolabeled anti-CD20 sdAb for targeted radionuclide therapy of non-hodgkin lymphoma. Molecular Cancer Therapeutics, Volume 16, pp. 2828- 2839.
  • Krüwel, T. et al., 2016. In vivo detection of small tumour lesions by multi-pinhole SPECT applying a 99mTc-labelled nanobody targeting the Epidermal Growth Factor Receptor. Scientific Reports, Volume 6, p. e21834 .
  • Lal, A. et al., 2002. Mutant epidermal growth factor receptor up-regulates molecular effectors of tumor invasion. Cancer Research, Volume 62, pp. 3335-3339.
  • Lemaire, M. et al., 2013. Imaging and radioimmunotherapy of multiple myeloma with antiidiotypic Nanobodies. Leukemia, Volume 28, pp. 444-447.
  • Leone, J. P. & Leone, B. A., 2015. Breast cancer brain metastases: the last frontier. Experimental Hematology & Oncology, Volume 4.
  • Li, J. et al., 1997. PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer. Science, Volume 275, pp. 1943-1947.
  • Li, T. et al., 2016. Camelid single-domain antibodies: a versatile tool for in vivo imaging of extracelluar and intracellular brain targets. Journal of Controlled Diseases, Volume 243, pp. 1-10.
  • Liu, S. & Edwards, D. S., 2001. Synthesis and characterization of two 111In-labeled DTPApeptide conjugates. Bioconjugate Chemistry, Volume 12, pp. 630-634.
  • Lockman, P. et al., 2010. Heterogeneous blood-tumor barrier permeability determines drug efficacy in experimental brain metastases of breast cancer. Clinical Cancer Research, Volume 16, pp. 5664-5678.
  • Louis, D. N. et al., 2007. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathologica, Volume 114, pp. 97-109.
  • Luwor, R. et al., 2004. The tumor-specific de2-7 epidermal growth factor receptor (EGFR) promotes cells survival and heterodimerizes with the wild-type EGFR. Oncogene, Volume 23, pp. 6095-6104.
  • Macrocyclics, 2018. Bifinctional Chelators (BFCs). [Online] Available at: https://www.macrocyclics.com/online-catalog/bifunctional-chelators-bfcs…- chx-a-dtpa/
  • Malikki, H., 2016. Trial Watch: glioblastoma vaccine therapy disappointment in phase III trial. Nature Reviews Neurology, Volume 12, p. 190.
  • Massa, S. et al., 2016. Sortase A-mediated site-specific labeling of camelid single-domain antibody-fragments: a versatile strategy for multiple molecular imaging modalities. Contrast Media and Molecular Imaging, Volume 11, pp. 328-339.
  • Massa, S. et al., 2014. Site-specific labeling of cysteine-tagged camelid single-domain antibody-fragments for use in molecular imaging. Bioconjugate Chemistry, Volume 25, pp. 979-988.
  • McMurphy, T. et al., 2014. The anti-tumor activity of a neutralizing nanobody targeting leptin receptor in a mouse model of melanoma. PLoS One, Volume 28, p. e89895.
  • Melchers, L. J. et al., 2014. Head and neck squamous cell carcinomas do not express EGFRvIII. International Journal of Radiation Oncology, Biology, Physics, Volume 90, pp. 454-462.
  • Messaoudi, K., Clavreul, A. & Lagarce, F., 2015. Toward an effective strategy in glioblastoma treatment. Part I: resistance mechanisms and strategies to overcome resistance of glioblastoma to temozolomide. Drug Discovery Today, Volume 20, pp. 899-905.
  • Muyldermans, S., 2001. Single domain camel antibodies: current status. Reviews in Molecular Biotechnology, Volume 74, pp. 277-304.
  • Muyldermans, S., 2013. Nanobodies: natural single-domain antibodies. Annual Review of Biochemistry, Volume 82, pp. 775-797.
  • Nagane, M. et al., 1996. A common mutant epidermal growth factor receptor confers enhanced tumorigenicity on human glioblastoma cells by increasing proliferation and reducing apoptosis. Cancer Research, Volume 56, pp. 5079-5086.
  • National Brain Tumor Society, 2018. Quick Brain Tumor Facts. [Online] Available at: http://braintumor.org/brain-tumor-information/brain-tumor-facts/ [Accessed 14 8 2018]. National Institute of Biomedical Imaging and Bioengineering, 2016.
  • Nuclear Medicine. [Online] Available at: https://www.nibib.nih.gov/science-education/science-topics/nuclear-medi… [Accessed 13 December 2017].
  • National Institute of Biomedical Imaging and Bioengineering, n.d. Computed Tomography (CT). [Online] Available at: https://www.nibib.nih.gov/science-education/science-topics/computedtomo…- ct [Accessed 13 December 2017].
  • Neyns, B. et al., 2009. Stratified phase II trial of cetuximab in patients with recurrent highgrade glioma. Annals of oncology, pp. 1596-1603.
  • Niwinska, A., Murawska, M. & Pogoda, K., 2010. Breast cancer brain metastases: differences in survival depending on biological subtype, RPA RTOG prognostic class and systemic treatment after whole-brain radiotherapy (WBRT). Annals of Oncology, Volume 21, pp. 294- 298.
  • Ogawa, K. et al., 2008. Treatment and prognosis of brain metastases from breast cancer. Journal of Neuro-Oncology, Volume 231-238, p. 86.
  • Omidfar, K. et al., 2009. Expression of EGFRvIII in thyroid carcinoma: immunohistochemical study by camel antibodies. Immunological Investigations, Volume 38, pp. 165-180.
  • Omidfar, K. et al., 2004. Production of a novel camel single-domain antibody specific for the type III mutant EGFR. Tumor Biology, Volume 25, pp. 296-305.
  • Park, C.-K.et al., 2012. The changes in MGMT promoter methylation status in initial and recurrent glioblastomas. Translational Oncology, Volume 5, pp. 393-397.
  • Percy, D. et al., 2011. In vivo characterization of changing blood-tumor barrier permeability in a mouse model of breast cancer metastasis: a complementary magnetic resonance imaging approach. Investigative Radiology, Volume 46, pp. 718-725.
  • Rösch, F., Herzog, H. & Qaim, S. M., 2017. The beginning and development of the theranostic approach in nuclear medicine, as exemplified by the radionuclide pair 86Y and 90Y. Radiopharmaceuticals, Volume 10, p. E56.
  • Saerens, D., Conrath, K., Govaert, J. & Muyldermans, S., 2008. Disulfide bond introduction for general stabilization of immunoglobulin heavy-chain variable domains. Journal of Molecular Biology, Volume 377, pp. 478-488.
  • Sampson, J. et al., 2010. Immunologic escape after prolonged progression-free survival with epidermal growth factor receptor variant III peptide vaccination in patients with newly diagnosed glioblastoma. Journal of Clinical Oncology, Volume 28, pp. 4722-4729.
  • Sanai, N., Alvarez-Buylla, A. & Berger, M. S., 2005. Neural stem cells and the origin of gliomas. The New England Journal of Medicine, Volume 353, pp. 811-822.
  • Sanovich, E. et al., 1995. Pathway across blood-brain barrier opened by the bradykinin agonist, RMP-7. Brain Research, Volume 705, pp. 125-135.
  • Scarpa, L. et al., 2016. The 68Ga/177Lu theragnostic concept in PSMA targeting of castration-resistant prostate cancer: correlation of SUVmax values and absorbed dose estimates. European Journal of Medical Molecular Imaging, Volume 44, pp. 788-800.
  • Schmitz, K. R. et al., 2013. Structural evaluation of EGFR inhibition mechanisms for nanobodies/VHH domains. Structure, Volume 21, pp. 1214-1224.
  • Serlin, Y., Shelef, I., Knyazer, B. & Friedman, A., 2015. Anatomy and physiology of the blood-brain barrier. Seminars in Cell & Developmental Biology, Volume 38, pp. 2-6.
  • Seshacharyulu, P. et al., 2012. Targeting the EGFR signaling pathway in cancer therapy. Expert Opinion on Therapeutic Targets, Volume 16, pp. 15-31.
  • Shaw, R. J., 2006. Glucose metabolism and cancer. Current opinion in Cell Biology, Volume 18, pp. 598-608.
  • Sherriff, J. et al., 2013. Patterns of relapse in glioblastoma multiforme following concomitant chemotherapy with temozolomide. The British journal of radiology, Volume 86, p. 20120414.
  • Shinojima, N. et al., 2003. Prognostic value of epidermal growth factor receptor in patients with glioblastoma multiforme. Cancer Research, Volume 63, pp. 6962-6970.
  • Siegal, T. et al., 2000. In vivo assessment of the window of barrier opening after osmotic blood–brain barrier disruption in humans. Journal of Neurosurgery, Volume 92, pp. 599-605.
  • Silberstein, E. B., 2012. Radioiodine: the classic theranostic agent. Seminars in Nuclear Medicine, Volume 42, pp. 164-170.
  • Slobbe, P. et al., 2015. A comparative PET imaging study with the reversible and irreversible EGFR tyrosine kinase inhibitors [11C]erlotinib and [18F]afatinib in lung cancer-bearing mice. European Journal of Nuclear Medicine and Molecular Imaging, Volume 5, p. 14.
  • Snoeck, V., 2013. Current experience in immunogenicity assessment of next generation biologics-nanobodies. European Immunogenicity Symposium, Ablynx NV.
  • Society of Nuclear Medicine and Molecular Imaging, n.d. Fact sheet: what is nuclear medicine and molecular imaging?. [Online] Available at: http://www.snmmi.org/AboutSNMMI/Content.aspx?ItemNumber=15627 [Accessed 13 December 2017].
  • Steeland, S., Vandenbroucke, R. E. & Libert, C., 2016. Nanobodies as therapeutics: big opportunities for small antibodies. Drug Discovery Today, Volume 21, pp. 1076-1113.
  • Stemmler, H. et al., 2006. Characteristics of patients with brain metastases receiving trastuzumab for HER2 overexpressing metastatic breast cancer. Breast, Volume 15, pp. 219- 225.
  • Structural Biology Brussels, 2008. Nanobody-aided crystallography. [Online] Available at: http://www.structuralbiology.be/chaperones [Accessed 2 December 2017].
  • The American Cancer Society Medical and Editorial Content Team, 2017. Treating breast cancer. [Online] Available at: https://www.cancer.org/cancer/breast-cancer/treatment.html [Accessed 16 Februari 2018].
  • Theile, C. S. et al., 2013. Site-specific N-terminal labeling of proteins using sortase-mediated reactions. Nature Protocols, Volume 8, pp. 1800-1807.
  • U.S. National Library of Medicine, 2018. Clinical trials. [Online] Available at: https://clinicaltrials.gov/ct2/show/NCT02209376?term=NCT02209376&rank=1 [Accessed 18 8 2018].
  • Ullrich, R. T. et al., 2009. Methyl-L-11C-methionine PET as a diagnostic marker for malignant progression in patients with glioma. Journal of Nuclear Medicine, Volume 50, pp. 1962-1968.
  • Vallabhajosula, S., Killeen, R. P. & Osborne, J. R., 2010. Altered biodistribution of radiopharmaceuticals: role of radiochemical/pharmaceutical purity, physiological, and pharmacologic factors. Seminars in Nuclear Medicine, Volume 40, pp. 220-241.
  • van Tellingen, O. et al., 2015. Overcoming the blood-brain tumor barrier for effective glioblastoma treatment. Drug Resistance Updates, Volume 19, pp. 1-12.
  • Vaneycken, I. et al., 2011a. Preclinical screening of anti-HER2 nanobodies for molecular imaging of breast cancer. The FASEB Journal, Volume 25, pp. 2433-2446.
  • Vaneycken, I. et al., 2011b. Immuno-imaging using nanobodies. Current opinion in Biotechnology, Volume 22, pp. 877-881.
  • Vaneycken, I. et al., 2010. In vitro analysis and in vivo tumor targeting of a humanized, grafted nanobody in mice using pinhole SPECT/micro-CT. Journal of Nuclear Medicine, Volume 51, pp. 1099-1106.
  • Vaz-Luis, I. et al., 2012. Impact of hormone receptor status on patterns of recurrence and clinical outcomes among patients with human epidermal growth factor-2-positive breast cancer in the National Comprehensive Cancer Network: a prospective cohort study. Breast Cancer Research, Volume 14, p. R129.
  • Verma, S., 2014. Advances in treating HER2-positive breast cancer: an interview with Sunil Verma. BioMed Central Medicine, Volume 12, p. 129.
  • Verma, V. et al., 2018. Biotin-tagged proteins: reagents for efficient ELISA-based serodiagnosis and phage display-based affinity selection. PLoS One, Volume 13, p. e0191315.
  • Vincke, C. et al., 2012. Generation of single domain antibody fragments derived from camelids and generation of manifold constructs. Methods in Molecular Biology, Volume 907, pp. 145-176.
  • Vincke, C. et al., 2009. General strategy to humanize a camelid single-domain antibody and identification of a universal humanized nanobody scaffold. The Journal of Biological Chemistry, Volume 284, pp. 3273-3284.
  • Vriesendorp, H. M. & Vriesendorp, F. J., 2003. A review of the intravenous administration of radiolabeled immunoglobulin G to cancer patients. High or low protein dose?. Cancer Biotherapy and Radiopharmaceuticals, Volume 18, pp. 35-46.
  • Vrije Universiteit Brussel, 2015. Phase II clinical trial of 68Ga-anti-HER2-Nanobody PET/CT: validation of an innovative imaging technique to optimize patient selection for HER2-targeted therapy in breast carcinoma. [Online] Available at: https://cris.vub.be/en/projects/phase-ii-clinical-trial-of-68gaantiher2…- validation-of-an-innovative-imaging-technique-to-optimize-patient-selection-forher2targeted- therapy-in-breast-carcinoma(d326d395-9659-4135-ad20-a0c44598c784).html [Accessed 17 8 2018].
  • Walid, M. S., 2008. Prognostic factors for long-term survival after glioblastoma. The Permanente Journal, Volume 12, pp. 45-48.
  • Weller, M. et al., 2017. Rindopepimut with temozolomide for patients with newly diagnosed, EGFRvIII-expressing glioblastoma (ACT IV): a randomised, double-blind, international phase 3 trial. The Lancet. Oncology, Volume 18, pp. 1373-1385.
  • Wen, P. Y. & Kesari, S., 2008. Malignant gliomas in adults. The New England Journal of Medicine, Volume 359, pp. 492-507.
  • Werner, R. A. et al., 2015. 68Gallium- and 90Yttrium-/177Lutetium: ‘‘theranostic twins’’ for diagnosis and treatment of NETs. Annals of Nuclear Medicine, Volume 1-7, p. 29.
  • Wikstrand, C. et al., 1998. The class III variant of the epidermal growth factor receptor (EGFRvIII): characterization and utilization as an immunotherapeutic target. Journal of Neurovirology, Volume 4, pp. 148-158.
  • Xavier, C. et al., 2012. Site-specific labeling of His-tagged nanobodies with 99mTc: a practical guide. Methods in Molecular Biology, Volume 911, pp. 485-490.
  • Yarden, Y., 2001. EGFR family and its ligands in human cancer: signalling mechanisms and therapeutic opportunities. European Journal of Cancer, Volume 37, pp. S3-S8.
  • Yip, S. et al., 2009. MSH6 mutations arise in glioblastomas during temozolomide therapy and mediate temozolomide resistance. Clinical Cancer Research, Volume 15, pp. 4622-4629.
  • Zahonero, C. et al., 2015. Preclinical test of dacomitinib, an irreversible EGFR inhibitor, confirms its effectiveness for glioblastoma. Molecular Cancer Therapeutics, Volume 14, pp. 1548-1558.
  • Zhu, M. et al., 2014. Combining magnetic nanoparticle with biotinylated nanobodies for rapid and sensitive detection of influenza H3N2. Nanoscale Research Letters, Volume 9, p. 528.
Universiteit of Hogeschool
Master of Science in Bio Engineering Sciences – Cell and Gene Biotechnology
Publicatiejaar
2018
Promotor(en)
Nick Devoogdt
Kernwoorden
Share this on: