Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Bård Indrevoll is active.

Publication


Featured researches published by Bård Indrevoll.


Nature Medicine | 2015

Detection of colorectal polyps in humans using an intravenously administered fluorescent peptide targeted against c-Met

Jacobus Burggraaf; Ingrid M. C. Kamerling; Paul Gordon; Lenneke Schrier; Marieke L. de Kam; Andrea J Kales; Ragnar Bendiksen; Bård Indrevoll; Roger M. Bjerke; Siver A. Moestue; Siavash Yazdanfar; Alexandra M. J. Langers; Marit Swaerd-Nordmo; Geir Torheim; Madhuri Warren; Hans Morreau; Philip W. Voorneveld; Tessa Buckle; Fijs W. B. van Leeuwen; Liv-Ingrid Ødegårdstuen; Grethe Tang Dalsgaard; Andrew Healey; James C. Hardwick

Colon cancer prevention currently relies on colonoscopy using white light to detect and remove polyps, but small and flat polyps are difficult to detect and frequently missed when using this technique. Fluorescence colonoscopy combined with a fluorescent probe specific for a polyp biomarker may improve polyp detection. Here we describe GE-137, a water-soluble probe consisting of a 26–amino acid cyclic peptide that binds the human tyrosine kinase c-Met conjugated to a fluorescent cyanine dye. Intravenous administration of GE-137 leads to its accumulation specifically in c-Met–expressing tumors in mice, and it is safe and well tolerated in humans. Fluorescence colonoscopy in patients receiving intravenous GE-137 enabled visualization of all neoplastic polyps that were visible with white light (38), as well as an additional nine polyps that were not visible with white light. This first-in-human pilot study shows that molecular imaging using an intravenous fluorescent agent specific for c-Met is feasible and safe, and that it may enable the detection of polyps missed by other techniques.


Jacc-cardiovascular Imaging | 2008

Noninvasive imaging of angiotensin receptors after myocardial infarction.

Johan Verjans; Dagfinn Løvhaug; Navneet Narula; Artiom Petrov; Bård Indrevoll; Emma Bjurgert; Tatiana B. Krasieva; Lizette B. Petersen; Grete Mørk Kindberg; Magne Solbakken; Alan Cuthbertson; Mani A. Vannan; Chris Reutelingsperger; Bruce J. Tromberg; Leonard Hofstra; Jagat Narula

OBJECTIVES The purpose of this study was to evaluate the feasibility of noninvasive imaging of angiotensin II (AT) receptor upregulation in a mouse model of post-myocardial infarction (MI) heart failure (HF). BACKGROUND Circulating AT levels do not reflect the status of upregulation of renin-angiotensin axis in the myocardium, which plays a central role in ventricular remodeling and evolution of HF after MI. Appropriately labeled AT or AT receptor blocking agents should be able to specifically target AT receptors by molecular imaging techniques. METHODS AT receptor imaging was performed in 29 mice at various time points after permanent coronary artery ligation or in controls using a fluoresceinated angiotensin peptide analog (APA) and radiolabeled losartan. The APA was used in 19 animals for intravital fluorescence microscopy on a beating mouse heart. Tc-99m losartan was used for in vivo radionuclide imaging and quantitative assessment of AT receptor expression in 10 mice. After imaging, hearts were harvested for pathological characterization using confocal and 2-photon microscopy. RESULTS No or little APA uptake was observed in control animals or within infarct regions on days 0 and 1. Distinct uptake occurred in the infarct area at 1 to 12 weeks after MI; the uptake was at maximum at 3 weeks and reduced markedly at 12 weeks after MI. Ultrasonographic examination demonstrated left ventricular remodeling, and pathologic characterization revealed localization of the APA tracer with collagen-producing myofibroblasts. Tc-99m losartan uptake in the infarct region (0.524 +/- 0.212% injected dose/g) increased 2.4-fold as compared to uptake in the control animals (0.215 +/- 0.129%; p < 0.05). CONCLUSIONS The present study demonstrates the feasibility of in vivo molecular imaging of AT receptors in the remodeling myocardium. Noninvasive imaging studies aimed at AT receptor expression could play a role in identification of subjects likely to develop heart failure. In addition, such a strategy could allow for optimization of anti-angiotensin therapy in patients after MI.


The Journal of Nuclear Medicine | 2013

Three Methods for 18F Labeling of the HER2-Binding Affibody Molecule ZHER2:2891 Including Preclinical Assessment

Matthias Glaser; Peter Iveson; Susan Hoppmann; Bård Indrevoll; Anthony Wilson; Joseph Arukwe; Antonios Danikas; Rajiv Bhalla; Duncan Hiscock

Human epidermal growth factor receptor (HER2)–targeted Affibody molecules radiolabeled with 18F allow the noninvasive assessment of HER2 status in vivo through PET imaging. Such agents have the potential to improve patient management by selecting individuals for HER2-targeted therapies and allowing therapy monitoring. The aim of this study was to assess different 18F radiolabeling strategies of the HER2-specific Affibody molecule ZHER2:2891, preclinically determine the biologic efficacy of the different radiolabel molecules, and select a preferred radiolabeling strategy to progress for automated manufacture. Methods: Cysteine was added to the C terminus of the Affibody molecule for the coupling of maleimide linkers, and 3 radiolabeling strategies were assessed: silicon-fluoride acceptor approach (18F-SiFA), 18F-AlF-NOTA, and 4-18F-fluorobenzaldehyde (18F-FBA). The biodistributions of the radiolabeled Affibody molecules were then determined in naïve CD-1 nude mice, and tumor targeting was assessed in CD-1 nude mice bearing high-HER2-expressing NCI-N87 tumors and low-HER2-expressing A431 tumors. The 111In-ABY-025 compound, which has demonstrable clinical utility, served as a reference tracer. Results: The non–decay-corrected radiochemical yields based on starting 18F-fluoride using the 18F-FBA, 18F-SiFA, and 18F-AlF-NOTA methods were 13% ± 3% (n = 5), 38% ± 2% (n = 3), and 11% ± 4% (n = 6), respectively. In naïve mice, both the 18F-AlF-NOTA-ZHER2:2891 and the 111In-ABY-025 compounds showed a significant kidney retention (70.3 ± 1.3 and 73.8 ± 3.0 percentage injected dose [%ID], respectively, at 90 min after injection), which was not observed for 18F-FBA-ZHER2:2891 or 18F-SiFA-ZHER2:2891 (4.8 ± 0.6 and 10.1 ± 0.7 %ID, respectively, at 90 min). The 18F-SiFA-ZHER2:2891 conjugate was compromised by increasing bone retention over time (5.3 ± 1.0 %ID/g at 90 min after injection), indicating defluorination. All the radiolabeled Affibody molecules assessed showed significantly higher retention in NCI-N87 tumors than A431 tumors at all time points (P < 0.05), and PET/CT imaging of 18F-FBA-ZHER2:2891 in a dual NCI-N87/A431 xenograft model demonstrated high tumor-to-background contrast for NCI-N87 tumors. Conclusion: The HER2 Affibody molecule ZHER2:2891 has been site-selectively radiolabeled by three 18F conjugation methods. Preliminary biologic data have identified 18F-FBA-ZHER2:2891 (also known as GE226) as a favored candidate for further development and radiochemistry automation.


Bioorganic & Medicinal Chemistry Letters | 2011

Synthesis and in vitro evaluation of [18F]fluoroethyl triazole labelled [Tyr3]octreotate analogues using click chemistry

Lisa Iddon; Julius Leyton; Bård Indrevoll; Matthias Glaser; Edward G. Robins; Andrew J. T. George; Alan Cuthbertson; Sajinder K. Luthra; Eric O. Aboagye

A novel class of alkyne linked [Tyr(3)]octreotate analogues have been labelled by a copper catalysed azide-alkyne cycloaddition reaction (CuAAC) to form a 1,4-substituted triazole using the reagent [(18)F]2-fluoroethyl azide. An unexpected variability in reactivity during the CuAAC reaction was observed for each alkyne analogue which has been investigated. Two lead alkyne linked [Tyr(3)]octreotate analogues, G-TOCA (3a) and βAG-TOCA (5a) have been identified to be highly reactive in the click reaction showing complete conversion to the [(18)F]2-fluoroethyl triazole linked [Tyr(3)]octreotate analogues FET-G-TOCA (3b) and FET-βAG-TOCA (5b) under mild conditions and with short synthesis times (5 min at 20 °C). As well as ease of synthesis, in vitro binding to the pancreatic tumour AR42J cells showed that both FET-G-TOCA and FET-βAG-TOCA have high affinity for the somatostatin receptor with IC(50) of 4.0±1.4, and 1.6±0.2 nM, respectively.


The Journal of Nuclear Medicine | 2011

Targeting Somatostatin Receptors: Preclinical Evaluation of Novel 18F-Fluoroethyltriazole-Tyr3-Octreotate Analogs for PET

Julius Leyton; Lisa Iddon; Meg Perumal; Bård Indrevoll; Matthias Glaser; Edward G. Robins; Andrew J. T. George; Alan Cuthbertson; Sajinder K. Luthra; Eric O. Aboagye

The incidence and prevalence of gastroenteropancreatic neuroendocrine tumors has been increasing over the past 3 decades. Because of high densities of somatostatin receptors (sstr)—mainly sstr-2—on the cell surface of these tumors, 111In-diethylenetriaminepentaacetic acid-octreotide scintigraphy has become an important part of clinical management. 18F-radiolabeled analogs with suitable pharmacokinetics would permit PET with more rapid clinical protocols. Methods: We compared the affinity in vitro and tissue pharmacokinetics by PET of 5 structurally related 19F/18F-fluoroethyltriazole-Tyr3-octreotate (FET-TOCA) analogs: FET-G-polyethylene glycol (PEG)-TOCA, FETE-PEG-TOCA, FET-G-TOCA, FETE-TOCA, and FET-βAG-TOCA to the recently described 18F-aluminum fluoride NOTA-octreotide (18F-AIF-NOTA-OC) and the clinical radiotracer 68Ga-DOTATATE. Results: All 19F-fluoroethyltriazole-Tyr3-octreotate compounds retained high agonist binding affinity to sstr-2 in vitro (half-maximal effective concentration, 4–19 nM vs. somatostatin at 5.6 nM). Dynamic PET showed that incorporation of PEG linkers, exemplified by 18F-FET-G-PEG-TOCA and 18F-FETE-PEG-TOCA, reduced uptake in high sstr-2–expressing AR42J pancreatic cancer xenografts. 18F-FET-βAG-TOCA showed the lowest nonspecific uptake in the liver. Tumor uptake increased in the order 68Ga-DOTATATE < 18F-AIF-NOTA ≤ 18F-FET-βAG-TOCA < 18F-FET-G-TOCA. The uptake of 18F-FET-βAG-TOCA was specific: a radiolabeled scrambled peptide, 18F-FET-βAG-[W-c-(CTFTYC)K], did not show tumor uptake; there was lower uptake of 18F-FET-βAG-TOCA in AR42J xenografts when mice were pretreated with 10 mg of unlabeled octreotide per kilogram; and there was low uptake of 18F-FET-βAG-TOCA in low sstr-2–expressing HCT116 xenografts. Conclusion: We have developed novel fluoroethyltriazole-Tyr3-octreotate radioligands that combine high specific binding with rapid target localization and rapid pharmacokinetics for high-contrast PET. 18F-FET-βAG-TOCA and 18F-FET-G-TOCA are candidates for future clinical evaluation.


Contrast Media & Molecular Imaging | 2009

Whole-body section fluorescence imaging--a novel method for tissue distribution studies of fluorescent substances.

Siver Moestue; Paula Nunez; Andrew Healey; Roger M. Bjerke; Bård Indrevoll; Tore Skotland; Svein Olaf Hustvedt

The present study demonstrates the usefulness of whole body section fluorescence imaging, a novel technique used in optical imaging drug discovery. This method is in principle an analog of whole body autoradiography, except that fluorescence is measured instead of radioactivity. The method was shown to have a linear concentration-response relationship over a 1000-fold concentration range. Densitometric image analysis allowed semiquantitative studies of drug disposition and selective tumor retention of an optical imaging drug candidate.


Proceedings of SPIE | 2008

A targeted molecular probe for colorectal cancer imaging

T. Attramadal; Roger M. Bjerke; Bård Indrevoll; S. Moestue; A. Rogstad; Ragnar Bendiksen; Andrew Healey; Edvin Wilhelm Johannesen

Colorectal cancer is a major cause of cancer death. Morbidity, mortality and healthcare costs can be reduced if the disease can be detected at an early stage. Screening is a viable approach as there is a clear link to risk factors such as age. We have developed a fluorescent contrast agent for use during colonoscopy. The agent is administered intravenously and is targeted to an early stage molecular marker for colorectal cancer. The agent consists of a targeting section comprising a peptide, and a fluorescent reporter molecule. Clinical imaging of the agent is to be performed with a far red fluorescence imaging channel (635 nm excitation/660-700 nm emission) as an adjunct to white light colonoscopy. Preclinical proof of mechanism results are presented. The compound has a Kd of ~3nM. Two human xenograft tumour models were used. Tumour cells were implanted and grown subcutaneously in nude mice. Imaging using a fluorescence reflectance imaging system and quantitative biodistribution studies were performed. Substances tested include the targeted agent, and a scrambled sequence of the peptide (no binding) used as a negative control. Competition studies were also performed by co-administration of 180 times excess unlabelled peptide. Positive imaging contrast was shown in the tumours, with a clear relationship to expression levels (confirmed with quantitative biodistribution data). There was a significant difference between the positive and negative control substances, and a significant reduction in contrast in the competition experiment.


MedChemComm | 2015

Radiosynthesis of high affinity fluorine-18 labeled GnRH peptide analogues: in vitro studies and in vivo assessment of brain uptake in rats

Dag Erlend Olberg; Sven H. Hausner; Nadine Bauer; Jo Klaveness; Bård Indrevoll; Kjetil Wessel Andressen; Marie Dahl; Finn Olav Levy; Julie L. Sutcliffe; Ira Haraldsen

Gonadotropin releasing hormone (GnRH) is recognized as an important neuromodulator affecting behavior and has been associated with the progression of Alzheimers disease. The peptide has been shown to have a bidirectional transport through the blood–brain-barrier (BBB), which may account for the cognitive effects of systemically administered GnRH. In this study, four novel 18F-GnRH peptide analogues were synthesized and their in vitro and in vivo characteristics studied in male rats. GnRH peptides were assembled by solid-phase peptide synthesis, either as the full length D-Lys6-GnRH (pyroGlu1-His2-Trp3-Ser4-Tyr5-D-Lys6-Leu7-Arg8-Pro9-Gly10-NH2) or as D-Lys6-desGly10-GnRH-NHEt. In all, four GnRH peptide analogues were synthesized and reacted with N-succinimidyl-4-fluorobenzoate (SFB) to yield the fluorinated versions. Binding affinities of the analogues were determined in a competitive binding assay for both human and rat GnRH receptors. Ki-values for the GnRH peptides were found to be subnanomolar, with D-Lys6(FBA)-desGly10-GnRH-NHEt (7) being most potent with a Ki-value of around 50 pM for GnRH receptor species. Radiolabeling was performed using N-succinimidyl-4-[18F]fluorobenzoate ([18F]SFB) in 33.3 ± 12.8% isolated decay corrected (d.c.) yield within 1.5–2 h. Rat serum stability over 2 h revealed minor degradation (≤5%). For in vivo studies, 18F-peptides (4–30 MBq) were injected intravenously via the tail vein into rats and brain uptake was evaluated by means of dynamic PET (2 h) followed by biodistribution studies. PET showed limited or no uptake in brain for the 18F-peptides which predominantly cleared rapidly by renal excretion. Specific binding in the pituitary gland was confirmed for the 18F-peptide, 7, by blocking with the GnRH agonist buserelin.


The Journal of Nuclear Medicine | 2014

Reply: Al18F Labeling of Affibody Molecules

Matthias Glaser; Peter Iveson; Susan Hoppmann; Bård Indrevoll; Anthony Wilson; Joseph Arukwe; Antonios Danikas; Rajiv Bhalla; Duncan Hiscock

TO THE EDITOR: Glaser et al. recently described the labeling of F-ZHER2:2891-Cys-NOTA-(COOH)2-AlF (18F-12) (1) and compared it in vivo to the biodistribution of that Affibody (Affibody AB) with 18F attached to carbon and silicon, as well as an 111In-DOTA-Affibody. They reported that the Al18F-labeled Affibody had a similar biodistribution to the 111In-Affibody, as previously noted by Heskamp et al. (2), and also observed that the Al18F-labeled Affibody had high uptake and retention in the kidney (;80 percentage injected dose [%ID], like the 111In-Affibody). This is presumably because the small-sized Affibody is eliminated through the kidneys, where it is rapidly catabolized, with the resulting Al18F complex residualized in the renal tubules in the same manner as the 111In-DOTA complex (3). In contrast, when the carbonand silicon-labeled Affibody molecules are metabolized in the kidney, the 18F-labeled metabolites are eliminated from the kidney cells, greatly reducing renal uptake. Although this clearly serves as an advantage for this product, much like differences between radioiodinated and radiometal-labeled antibody fragments, it is important to emphasize that renal uptake of the Al18F-Affibody product is a property of the Affibody targeting agent and not the Al18F complex. Previous studies with our pretargeting peptide (4) and the Al18F-NOTA-pegylated arginine-glycineaspartic acid dimer (PRGD2) peptide (5) both showed excellent renal clearance in the mouse models, and the Al18F-NOTAPRGD2 peptide also had good renal clearance in humans (6). It should also be noted that the 18F-Affibody labeled through a carbon atom had high hepatobiliary clearance (40–50 %ID in the intestines), whereas the Al18F-labeled Affibody had low uptake in the intestines. The high hepatobiliary accretion might be considered at least as undesirable as the high renal retention, depending on the use of the agent. Glaser et al. also reported a 2-fold lower labeling yield for their Al18F-Affibody than the Al18F-labeling yield of a similar Affibody bearing the same NOTA ligand (11% vs. 21%), and this despite the fact that Heskamp et al. used a lower amount of the Affibody (2). Although we cannot discount the possibility that slight differences in the Affibody structure could have influenced the yields, we strongly suspect the yield differences are attributable to the lack of a co-solvent in the labeling procedure used by Glaser et al. Indeed, we have shown that the use of a co-solvent generally improves yields 2-fold (7). Thus, we believe it is important when comparing labeling technologies to attempt to optimize or normalize each procedure, or if not empirically assessed, to state the conditions that might have affected yields when this information has been published previously. Second, whereas the nonresidualizing 18F-linkage used by Glaser et al. provided lower renal uptake, there likely are other situations, such as in target cells with a more rapid metabolism, in which a residualizing form of 18F afforded by the AlF method would be preferred (8). DISCLOSURE


Cancer Research | 2017

Abstract 5859: HER2-targeted thorium-227 conjugate (HER2-TTC): Efficacy in preclinical models of trastuzumab and T-DM1 resistance

Jenny Karlsson; Urs B. Hagemann; Christoph Schatz; Derek Grant; Alexander Kristian; Christine Ellingsen; Dessislava Mihaylova; Solène Géraudie; Bård Indrevoll; Uta Wirnitzer; Roger M. Bjerke; Olav B. Ryan; Carl Friedrich Nising; Dominik Mumberg; Alan Cuthbertson

The human epidermal growth factor receptor 2 (HER2) is encoded by the proto-oncogene c-erbB-2 and initiates downstream signaling pathways leading to cell proliferation and tumorigenesis. HER2 is overexpressed in several cancer (Ca) types and is one of the most strongly validated targets for the treatment of breast and gastric cancer serving as both a prognostic and predictive biomarker. Several HER2-targeting antibodies as well as antibody-drug conjugates are either approved or are in clinical development. Prolonged treatment with monoclonal antibodies and antibody drug conjugates have resulted in development of resistance and so there is still an unmet medical need for drugs of new mechanism of action targeting this important receptor system. We describe herein the generation of a high energy, alpha-particle emitting HER2 targeted thorium-227 antibody-chelator conjugate. HER2-TTC consists of the humanized HER2 targeting IgG1 antibody (trastuzumab) covalently linked via an amide bond to a 3,2-hydroxypyridino-based chelator moiety, enabling efficient radiolabeling with the alpha particle emitting radionuclide thorium-227 (Th-227). HER2-TTC was prepared at high radiochemical yield and purity. When tested for binding to recombinant HER2, HER2-TTC was shown to retain comparable binding affinity to trastuzumab. In vitro cytotoxicity experiments were performed on 8 cell lines with different HER2 expression levels (from 7 000 - 500 000 mAbs bound/ cell as determined by FACS) of breast, ovarian, gastric and lung cancer origin. HER2-TTC demonstrated target mediated in vitro cytotoxicity in the pM-range. In vivo biodistribution and anti-tumor efficacy of HER2-TTC was evaluated in the dose range 100-500 kBq/kg at a protein dose of 0.14 mg/kg and i.v. injection in the s.c. KPL-4 breast and Calu-3 lung model previously described to be resistant to trastuzumab. The biodistribution study demonstrated specific tumor accumulation of HER2-TTC in both models with a maximum of 77 and 50 %ID/g 227Th at t = 168 h post dose (decay corrected to T0), respectively. Significant antitumor efficacy was shown for HER2-TTC in the JIMT-1 s.c. breast Ca xenograft model resistant to trastuzumab and T-DM1. The promising preclinical anti-tumor activity supports the development of the targeted alpha therapeutic HER2-TTC for the treatment of trastuzumab and T-DM1 resistant patients. Citation Format: Jenny Karlsson, Urs B. Hagemann, Christoph Schatz, Derek Grant, Alexander Kristian, Christine Ellingsen, Dessislava Mihaylova, Solene Geraudie, Bard Indrevoll, Uta Wirnitzer, Roger M. Bjerke, Olav B. Ryan, Carl F. Nising, Dominik Mumberg, Alan Cuthbertson. HER2-targeted thorium-227 conjugate (HER2-TTC): Efficacy in preclinical models of trastuzumab and T-DM1 resistance [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 5859. doi:10.1158/1538-7445.AM2017-5859

Collaboration


Dive into the Bård Indrevoll's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rajiv Bhalla

University of Queensland

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge