Kiki C. Andree
University of Twente
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Kiki C. Andree.
Molecular Oncology | 2016
Kiki C. Andree; Guus van Dalum; Leon W.M.M. Terstappen
Enumeration and characterization of circulating tumor cells (CTC) hold the promise of a real time liquid biopsy. They are however present in a large background of hematopoietic cells making their isolation technically challenging. In 2004, the CellSearch system was introduced as the first and only FDA cleared method designed for the enumeration of circulating tumor cells in 7.5 mL of blood. Presence of CTC detected by CellSearch is associated with poor prognosis in metastatic carcinomas. CTC remaining in patients after the first cycles of therapy indicates a futile therapy. Here we review challenges faced during the development of the CellSearch system and the difficulties in assigning objects as CTC. The large heterogeneity of CTC and the different approaches introduced in recent years to isolate, enumerate and characterize CTC results in a large variation of the number of CTC reported urging the need for uniform definitions and at least a clear definition of what the criteria are for assigning an object as a CTC.
ACS Applied Materials & Interfaces | 2016
Kiki C. Andree; Ana M.C. Barradas; Ai T. Nguyen; Anouk Mentink; Ivan Stojanovic; Jacob Baggerman; Joost van Dalum; Cees J.M. van Rijn; Leon W.M.M. Terstappen
The presence of tumor cells in blood is predictive of short survival in several cancers and their isolation and characterization can guide toward the use of more effective treatments. These circulating tumor cells (CTC) are, however, extremely rare and require a technology that is sufficiently sensitive and specific to identify CTC against a background of billions of blood cells. Immuno-capture of cells expressing the epithelial cell adhesion molecule (EpCAM) are frequently used to enrich CTC from blood. The choice of bio conjugation strategy and antibody clone is crucial for adequate cell capture but is poorly understood. In this study, we determined the binding affinity constants and epitope binding of the EpCAM antibodies VU1D-9, HO-3, EpAb3-5, and MJ-37 by surface plasmon resonance imaging (SPRi). Glass surfaces were coated using a poly(acrylic acid) based coating and functionalized with anti-EpCAM antibodies. Binding of cells from the breast carcinoma cell line (SKBR-3) to the functionalized surfaces were compared. Although EpAb3-5 displayed the highest binding affinity HO-3 captured the highest amount of cells. Hence we report differences in the performance of the different antibodies and more importantly that the choice of antibody to capture CTC should be based on multiple assays.
The Journal of Nuclear Medicine | 2017
Laurens T. van der Meer; Samantha Y.A. Terry; Dorette van Ingen Schenau; Kiki C. Andree; Gerben M. Franssen; D.M. Roeleveld; Josbert M. Metselaar; Thomas Reinheckel; Peter M. Hoogerbrugge; Otto C. Boerman; Frank N. van Leeuwen
The antileukemic drug asparaginase, a key component in the treatment of acute lymphoblastic leukemia, acts by depleting asparagine from the blood. However, little is known about its pharmacokinetics, and mechanisms of therapy resistance are poorly understood. Here, we explored the in vivo biodistribution of radiolabeled asparaginase, using a combination of imaging and biochemical techniques, and provide evidence for tissue-specific clearance mechanisms, which could reduce the effectiveness of the drug at these specific sites. Methods: In vivo localization of 111In-labeled Escherichia coli asparaginase was performed in C57BL/6 mice by both small-animal SPECT/CT and ex vivo biodistribution studies. Mice were treated with liposomal clodronate to investigate the effect of macrophage depletion on tracer localization and drug clearance in vivo. Moreover, macrophage cell line models RAW264.7 and THP-1, as well as knockout mice, were used to identify the cellular and molecular components controlling asparaginase pharmacokinetics. Results: In vivo imaging and biodistribution studies showed a rapid accumulation of asparaginase in macrophage-rich tissues such as the liver, spleen, and in particular bone marrow. Clodronate-mediated depletion of phagocytic cells markedly prolonged the serum half-life of asparaginase in vivo and decreased drug uptake in these macrophage-rich organs. Immunohistochemistry and in vitro binding assays confirmed the involvement of macrophagelike cells in the uptake of asparaginase. We identified the activity of the lysosomal protease cathepsin B in macrophages as a rate-limiting factor in degrading asparaginase both in vitro and in vivo. Conclusion: We showed that asparaginase is rapidly cleared from the serum by liver-, spleen-, and bone marrow–resident phagocytic cells. As a consequence of this efficient uptake and protease-mediated degradation, particularly bone marrow–resident macrophages may provide a protective niche to leukemic cells.
International Journal of Cancer | 2018
Kiki C. Andree; Anouk Mentink; Leonie L. Zeune; Leon W.M.M. Terstappen; Nikolas H. Stoecklein; Rui Neves; Christiane Driemel; Rita Lampignano; Liwen Yang; Hans Neubauer; Tanja Fehm; Johannes C. Fischer; Elisabetta Rossi; Mariangela Manicone; Umberto Basso; Piero Marson; Rita Zamarchi; Y. Loriot; Valérie Lapierre; Vincent Faugeroux; Marianne Oulhen; Françoise Farace; Gemma Fowler; Mariane Sousa Fontes; Berni Ebbs; Maryou B. Lambros; Mateus Crespo; Penny Flohr; Johann S. de Bono
Frequently, the number of circulating tumor cells (CTC) isolated in 7.5 mL of blood is too small to reliably determine tumor heterogeneity and to be representative as a “liquid biopsy”. In the EU FP7 program CTCTrap, we aimed to validate and optimize the recently introduced Diagnostic LeukApheresis (DLA) to screen liters of blood. Here we present the results obtained from 34 metastatic cancer patients subjected to DLA in the participating institutions. About 7.5 mL blood processed with CellSearch® was used as “gold standard” reference. DLAs were obtained from 22 metastatic prostate and 12 metastatic breast cancer patients at four different institutions without any noticeable side effects. DLA samples were prepared and processed with different analysis techniques. Processing DLA using CellSearch resulted in a 0–32 fold increase in CTC yield compared to processing 7.5 mL blood. Filtration of DLA through 5 μm pores microsieves was accompanied by large CTC losses. Leukocyte depletion of 18 mL followed by CellSearch yielded an increase of the number of CTC but a relative decrease in yield (37%) versus CellSearch DLA. In four out of seven patients with 0 CTC detected in 7.5 mL of blood, CTC were detected in DLA (range 1–4 CTC). The CTC obtained through DLA enables molecular characterization of the tumor. CTC enrichment technologies however still need to be improved to isolate all the CTC present in the DLA.
Clinical Cancer Research | 2018
Maryou B. Lambros; George Seed; Semini Sumanasuriya; Veronica Gil; Mateus Crespo; Mariane Sousa Fontes; Rob Chandler; Niven Mehra; Gemma Fowler; Berni Ebbs; Penelope Flohr; Susana Miranda; Wei Yuan; Alan Mackay; Ana Ferreira; Rita Pereira; Claudia Bertan; Ines Figueiredo; Ruth Riisnaes; Daniel Nava Rodrigues; Adam Sharp; Jane Goodall; Gunther Boysen; Suzanne Carreira; Diletta Bianchini; Pasquale Rescigno; Zafeiris Zafeiriou; Joanne Hunt; Deirdre Moloney; Lucy Hamilton
Purpose: Circulating tumor cells (CTCs) have clinical relevance, but their study has been limited by their low frequency. Experimental Design: We evaluated liquid biopsies by apheresis to increase CTC yield from patients suffering from metastatic prostate cancer, allow precise gene copy-number calls, and study disease heterogeneity. Results: Apheresis was well tolerated and allowed the separation of large numbers of CTCs; the average CTC yield from 7.5 mL of peripheral blood was 167 CTCs, whereas the average CTC yield per apheresis (mean volume: 59.5 mL) was 12,546 CTCs. Purified single CTCs could be isolated from apheresis product by FACS sorting; copy-number aberration (CNA) profiles of 185 single CTCs from 14 patients revealed the genomic landscape of lethal prostate cancer and identified complex intrapatient, intercell, genomic heterogeneity missed on bulk biopsy analyses. Conclusions: Apheresis facilitated the capture of large numbers of CTCs noninvasively with minimal morbidity and allowed the deconvolution of intrapatient heterogeneity and clonal evolution. Clin Cancer Res; 24(22); 5635–44. ©2018 AACR.
Cancer Research | 2016
Thomas Schlange; Nikolas H. Stoecklein; Rui Neves; S. Pleier; Sabrina Bender; Nora Brychta; M.V. Luetke-Eversloh; Kiki C. Andree; Leonardus Wendelinus Mathias Marie Terstappen; Thomas Krahn
Within the European Innovative Medicines Initiative (IMI) consortium CANCER-ID (www.cancer-id.eu), scientists at academic, clinical and industrial sites across Europe and in the US joined forces to evaluate innovative technologies in the field of liquid cancer biopsies. This project aims at implementing standard operating procedures (SOPs) for pre-analytical sample handling, enrichment, isolation and analysis of Circulating Tumor Cells (CTCs), circulating free tumor DNA (ctDNA) and microRNAs (miRNAs) as novel blood-based biomarkers, with a focus on Non-Small Cell Lung Cancer (NSCLC) and HER2-treatment refractory breast cancer. In order to determine sensitivity and specificity of different technologies for CTC isolation and analysis (e.g. detection of mutations, amplifications, protein phosphorylation), complex samples comprising a mixture of NSCLC or breast cancer cell lines spiked in healthy donor blood were distributed to different CANCER-ID partner sites. These cell lines have been selected based on their molecular/genetic properties to reflect clinically relevant subtypes of the disease and have been further characterized in terms of cell-surface marker expression and cell size distribution. The use of complex spiked samples better models the heterogeneity of real-life patient material. Furthermore, healthy donor and patient derived plasma samples are investigated using different technology platforms to validate tumor-specific miRNA or ctDNA profiles that might characterize molecular tumor subtypes. To this end, differences in exosome-derived versus free circulating miRNAs are of special interest. As for CTCs the development of ctDNA and miRNA standards that can be used to compare and validate different technologies are in the focus of this effort. In summary, our results pave the way for the next phase of CANCER-ID, which includes the analysis of cancer patient samples in clinical studies using different technologies and thereby advance the concept of liquid biopsy particularly in indications in which conventional tissue biopsies are difficult to obtain. Citation Format: Thomas Schlange, Nikolas Stoecklein, Rui P. Neves, Sabrina Pleier, Sebastian Bender, Nora Brychta, Merlin V. Luetke-Eversloh, Kiki Andree, Leon Terstappen, Thomas Krahn, Thomas Krahn. Standardization of technologies for CTC, ctDNA and miRNA enrichment, isolation and analysis for liquid biopsies during the first year of IMI9s CANCER-ID. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 513.
Cancer Research | 2017
Maryou B. Lambros; Veronica Gil; Mateus Crespo; Mariane Sousa Fontes; Rui N. Neves; Niven Mahra; Gemma Fowler; Berni Ebbs; Penny Flohr; George Seed; Wei Yuan; Joanne Hunt; Deirdre Moloney; Dionne Ayanda; Joost F. Swennenhuis; Kiki C. Andree; Semini Sumanasuriya; Matthew Clarke; Pasquale Rescigno; Zafeiris Zafeiriou; Joaquin Mateo; Diletta Bianchini; Nikolas H. Stoecklein; Leon W.M.M. Terstappen; Gunther Boysen; Johann S. de Bono
Introduction: CTC count is an independent predictor of overall survival in mCRPC. Isolation of CTC from peripheral blood (PB) for genomic and functional analysis is challenging, especially in patients (pts) with low CTC count. It has been shown that DLA increases CTC yield. However, it has yet to be proven whether CTC isolation from DLA can be used in complementary studies such as molecular characterization and growth of organoid culture for drug sensitivity studies. Here we present preliminary data of an on-going study, which evaluates DLA in mCRPC pts, focusing on safety, CTC enrichment, molecular characterization and feasibility for organoid culture. Methods: mCRPC pts considered for clinical trials were selected according to performance status (ECOG 0-1) and number of CTC found in 7.5ml PB (>20 cells/7.5mL). DLA products (200x106 cells) were processed using the CellSearch CTC kit (Janssen Diagnostics, LLC) according to manufacturer procedures. The contents of CellSearch cartridges were sorted into single cell by fluorescence activated cell sorting (FACS) and subsequently assessed by array comparative genomic hybridization (aCGH) for copy number aberrations (CNA). Enrichment of CTC for organoid culture was performed by density gradient of mononuclear cells followed by positive selection using magnetic beads. Results: Overall 12 mCRPC patients underwent DLA without any complication or toxicity. The mean CTC count was 90 CTC/7.5 ml peripheral blood (median = 31) and ranged from 20 to 324. CellSearch CTC count in the DLA yielded a mean of 466 (median=203) and ranged from 60 to 2496 with an up to 40-fold increase (mean = 13, median = 6) in CTC count separation when comparing 1mL of PB to 1mL of DLA. Molecular analyses of FACS single CTC from the DLA by aCGH showed that these CTC genomic profiles had the typical hallmarks of mCRPC with CNAs including AR and MYC locus (8q) amplification, and PTEN, RB1, TP53, CHD1 loss. Additionally, ex vivo culture of CTC-derived organoids was successfully achieved. aCGH of these organoids matched the genomic profile that of the CTC from the same patient. Conclusion: DLA from mCRPC pts was well tolerated and yields higher CTC capture than PB and may provide an alternative to tissue biopsy and routine blood volumes. Our strategy allowed us to isolate genomic DNA with good quality for molecular characterization and viable CTC for organoid culture and functional studies. Citation Format: Maryou B. Lambros, Veronica S. Gil, Mateus Crespo, Mariane S. Fontes, Rui N. Neves, Niven Mahra, Gemma Fowler, Berni Ebbs, Penny Flohr, George Seed, Wei Yuan, Joanne Hunt, Deirdre Moloney, Dionne Ayanda, Joost F. Swennenhuis, Kiki C. Andree, Semini Sumanasuriya, Matthew Clarke, Pasquale Rescigno, Zafeiris Zafeiriou, Joaquin Mateo, Diletta Bianchini, Nikolas H. Stoecklein, Leon W. Terstappen, Gunther Boysen, Johann S. De Bono. Diagnostic leukapheresis (DLA): Molecular characterisation and organoid culture of circulating tumor cells (CTC) from metastatic castration resistant prostate cancer (mCRPC) [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 993. doi:10.1158/1538-7445.AM2017-993
Cancer Research | 2016
Kiki C. Andree; Anouk Mentink; Martin Scholz; Roland Kirchner; Rui Neves; Christiane Driemel; Rita Lampignano; Hans Neubauer; Dieter Niederacher; Tanja Fehm; Wolfram T. Knoefel; Johannes C. Fischer; Nikolas H. Stoecklein; Leon W.M.M. Terstappen
Introduction At present, the CellSearch system is the only validated method for the detection of circulating tumor cells (CTC) that has been cleared by the U.S. Food and Drug Administration. This system, designed for the enumeration of CTC in 7.5 mL of blood, detects CTC based on their expression of EpCAM and cytokeratins and negativity for CD45. However, the number of CTC that are detected in patients with metastatic carcinomas is in most cases too small to reliably determine tumor heterogeneity and to be representative as a ‘liquid biopsy’. Our aim is to identify and isolate a sufficient number of circulating tumor cells in virtually all metastatic cancer patients to enable their characterization and to represent a real-time liquid biopsy. For this purpose we used Diagnostic LeukApheresis (DLA) to increase the blood volume to be analyzed. We developed several techniques to isolate CTC from DLA to enable a multicenter comparison of CTC detection in DLA products. Methods DLAs were performed for ∼1 hour to obtain 40 mL of product containing ∼4 x10⁁9 mononuclear cells representing ∼1 liter of blood. Using CellSearch a maximum of 2 mL of DLA could be processed for EpCAM+ CTC (Fisher et al. doi: 10.1073/pnas.1313594110) and EpCAM- CTC (de Wit et al doi: 10.1038/srep12270). Using filtration through microsieves with 5 μm pores a maximum of 1.0 mL of DLA could be processed. To process 18 mL DLA product protocols were developed for leukocyte depletion using RosetteSep™ (StemCell Technologies, USA) and for EpCAM selection using an anti-EpCAM coated column (Leukocare AG). All enriched cell fractions were stained using CD45 PerCP, Cytokeratins PE and the nuclear dye DAPI, followed by fluorescence microscopy scanning and analysis. Results Leukocyte depletion using the RosetteSep™ CTC Enrichment cocktail was first optimized using small sample volumes (1 mL) spiked with cells from cancer cell lines. Depletion of leukocytes ranged from 3.1 to 3.9 logs with an average recovery of spiked cancer cells of 50-60%. Isolation of CTC expressing EpCAM was pursued using anti-EpCAM coated columns and optimized for selection and release of EpCAM expressing cells by passage of cells from cancer cell lines through the column resulting in 34-100% recovery. Both procedures were scaled up to enable processing of 18 mL of DLA. Leukocytes were depleted using RosetteSepTM by 3.1 - 3.9 logs whereas with anti-EpCAM columns only 1.7 - 1.8 logs depletion were reached. Using RosetteSepTM 21% and with the anti-EpCAM coated columns 2% of the tumor cells spiked into 18ml DLA were recovered. Conclusion Standard operating procedures were developed to isolate CTC in DLA9s from breast, prostate cancer and lung cancer patients for evaluation and comparison in the EU sponsored consortiums CTCTrap (www.utwente.nl/tnw/ctctrap/) and CANCER-ID (www.CANCER-ID.eu). Isolation of EpCAM expressing CTC using the anti-EpCAM coated columns will need further optimization before it can proceed to multicenter comparison. Citation Format: Kiki C. Andree, Anouk Mentink, Martin Scholz, Roland Kirchner, Rui P. Neves, Christiane Driemel, Rita Lampignano, Hans Neubauer, Dieter Niederacher, Tanja Fehm, Wolfram T. Knoefel, Johannes C. Fischer, Nikolas H. Stoecklein, Leon WMM Terstappen. The isolation of CTC from diagnostic leukapheresis. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 1532.
Analytical Biochemistry | 2016
Richardus B.M. Schasfoort; Kiki C. Andree; N. van der Velde; A. van der Kooi; Ivan Stojanovic; Leonardus Wendelinus Mathias Marie Terstappen
Blood | 2014
Laurens T. van der Meer; Samantha Terry; Dorette S van IngenSchenau; Kiki C. Andree; Peter M. Hoogerbrugge; Otto C. Boerman; Frank N. van Leeuwen