1
|
Hurkmans DP, Verdegaal EME, Hogan SA, de Wijn R, Hovestad L, van den Heuvel DMA, Ruijtenbeek R, Welters MJP, van Brakel M, Basak EA, Pinedo HM, Lamers CHJ, van de Werken HJG, Groten JP, Debets R, Levesque MP, Dummer R, Kapiteijn E, Mathijssen RHJ, Aerts JGJV, van der Burg SH. Blood-based kinase activity profiling: a potential predictor of response to immune checkpoint inhibition in metastatic cancer. J Immunother Cancer 2021; 8:jitc-2020-001607. [PMID: 33427690 PMCID: PMC7757459 DOI: 10.1136/jitc-2020-001607] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2020] [Indexed: 12/12/2022] Open
Abstract
Background Many cancer patients do not obtain clinical benefit from immune checkpoint inhibition. Checkpoint blockade targets T cells, suggesting that tyrosine kinase activity profiling of baseline peripheral blood mononuclear cells may predict clinical outcome. Methods Here a total of 160 patients with advanced melanoma or non-small-cell lung cancer (NSCLC), treated with anti-cytotoxic T-lymphocyte-associated protein 4 (anti-CTLA-4) or anti-programmed cell death 1 (anti-PD-1), were divided into five discovery and cross-validation cohorts. The kinase activity profile was generated by analyzing phosphorylation of peripheral blood mononuclear cell lysates in a microarray comprising of 144 peptides derived from sites that are substrates for protein tyrosine kinases. Binary grouping into patients with or without clinical benefit was based on Response Evaluation Criteria in Solid Tumors V.1.1. Predictive models were trained using partial least square discriminant analysis (PLS-DA), performance of the models was evaluated by estimating the correct classification rate (CCR) using cross-validation. Results The kinase phosphorylation signatures segregated responders from non-responders by differences in canonical pathways governing T-cell migration, infiltration and co-stimulation. PLS-DA resulted in a CCR of 100% and 93% in the anti-CTLA-4 and anti-PD1 melanoma discovery cohorts, respectively. Cross-validation cohorts to estimate the accuracy of the predictive models showed CCRs of 83% for anti-CTLA-4 and 78% or 68% for anti-PD-1 in melanoma or NSCLC, respectively. Conclusion Blood-based kinase activity profiling for response prediction to immune checkpoint inhibitors in melanoma and NSCLC revealed increased kinase activity in pathways associated with T-cell function and led to a classification model with a highly accurate classification rate in cross-validation groups. The predictive value of kinase activity profiling is prospectively verified in an ongoing trial.
Collapse
Affiliation(s)
- Daan P Hurkmans
- Department of Pulmonology, Erasmus University Medical Center, Rotterdam, The Netherlands .,Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Els M E Verdegaal
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Sabrina A Hogan
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Rik de Wijn
- PamGene International B.V, HH 's-Hertogenbosch, The Netherlands
| | - Lies Hovestad
- PamGene International B.V, HH 's-Hertogenbosch, The Netherlands
| | | | - Rob Ruijtenbeek
- PamGene International B.V, HH 's-Hertogenbosch, The Netherlands
| | - Marij J P Welters
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Mandy van Brakel
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Edwin A Basak
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - Cor H J Lamers
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Harmen J G van de Werken
- Department of Urology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands.,Cancer Computational Biology Center, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - John P Groten
- PamGene International B.V, HH 's-Hertogenbosch, The Netherlands
| | - Reno Debets
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Ellen Kapiteijn
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Joachim G J V Aerts
- Department of Pulmonology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Sjoerd H van der Burg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
2
|
do Canto LM, Barros-Filho MC, Rainho CA, Marinho D, Kupper BEC, Begnami MDFDS, Scapulatempo-Neto C, Havelund BM, Lindebjerg J, Marchi FA, Baumbach J, Aguiar S, Rogatto SR. Comprehensive Analysis of DNA Methylation and Prediction of Response to NeoadjuvantTherapy in Locally Advanced Rectal Cancer. Cancers (Basel) 2020; 12:cancers12113079. [PMID: 33105711 PMCID: PMC7690383 DOI: 10.3390/cancers12113079] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/09/2020] [Accepted: 10/16/2020] [Indexed: 12/19/2022] Open
Abstract
The treatment for locally advanced rectal carcinomas (LARC) is based on neoadjuvant chemoradiotherapy (nCRT) and surgery, which results in pathological complete response (pCR) in up to 30% of patients. Since epigenetic changes may influence response to therapy, we aimed to identify DNA methylation markers predictive of pCR in LARC patients treated with nCRT. We used high-throughput DNA methylation analysis of 32 treatment-naïve LARC biopsies and five normal rectal tissues to explore the predictive value of differentially methylated (DM) CpGs. External validation was carried out with The Cancer Genome Atlas-Rectal Adenocarcinoma (TCGA-READ 99 cases). A classifier based on three-CpGs DM (linked to OBSL1, GPR1, and INSIG1 genes) was able to discriminate pCR from incomplete responders with high sensitivity and specificity. The methylation levels of the selected CpGs confirmed the predictive value of our classifier in 77 LARCs evaluated by bisulfite pyrosequencing. Evaluation of external datasets (TCGA-READ, GSE81006, GSE75546, and GSE39958) reproduced our results. As the three CpGs were mapped near to regulatory elements, we performed an integrative analysis in regions associated with predicted cis-regulatory elements. A positive and inverse correlation between DNA methylation and gene expression was found in two CpGs. We propose a novel predictive tool based on three CpGs potentially useful for pretreatment screening of LARC patients and guide the selection of treatment modality.
Collapse
Affiliation(s)
- Luisa Matos do Canto
- Department of Clinical Genetics, University Hospital of Southern Denmark, 7100 Vejle, Denmark;
- International Research Center–CIPE, A.C. Camargo Cancer Center, Sao Paulo 04002-010, Brazil; (M.C.B.-F.); (F.A.M.)
| | - Mateus Camargo Barros-Filho
- International Research Center–CIPE, A.C. Camargo Cancer Center, Sao Paulo 04002-010, Brazil; (M.C.B.-F.); (F.A.M.)
- Department of Head and Neck Surgery, Hospital das Clinicas HCFMUSP, Sao Paulo 01246-903, Brazil
| | - Cláudia Aparecida Rainho
- Department of Chemical and Biological Sciences, Institute of Biosciences, Sao Paulo State University (Unesp), Botucatu 18618-689, Brazil;
| | - Diogo Marinho
- Institute of Biological Psychiatry, Psykiatrisk Center Sct. Hans, 4000 Roskilde, Denmark;
| | - Bruna Elisa Catin Kupper
- Colorectal Cancer Service, A.C. Camargo Cancer Center, Sao Paulo 04002-010, Brazil; (B.E.C.K.); (S.A.J.)
| | | | - Cristovam Scapulatempo-Neto
- Molecular Oncology Research Center, Barretos – 14784-400, and Diagnósticos da América (DASA), Barueri 06455010, Brazil;
| | - Birgitte Mayland Havelund
- Department of Oncology, University Hospital of Southern Denmark, 7100 Vejle, Denmark;
- Danish Colorectal Cancer Center South, 7100 Vejle, Denmark;
| | - Jan Lindebjerg
- Danish Colorectal Cancer Center South, 7100 Vejle, Denmark;
- Department of Pathology, University Hospital of Southern Denmark, 7100 Vejle, Denmark
| | - Fabio Albuquerque Marchi
- International Research Center–CIPE, A.C. Camargo Cancer Center, Sao Paulo 04002-010, Brazil; (M.C.B.-F.); (F.A.M.)
| | - Jan Baumbach
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany;
| | - Samuel Aguiar
- Colorectal Cancer Service, A.C. Camargo Cancer Center, Sao Paulo 04002-010, Brazil; (B.E.C.K.); (S.A.J.)
| | - Silvia Regina Rogatto
- Department of Clinical Genetics, University Hospital of Southern Denmark, 7100 Vejle, Denmark;
- Danish Colorectal Cancer Center South, 7100 Vejle, Denmark;
- Institute of Regional Health Research, Faculty of Health Sciences, University of Southern Denmark, 5000 Odense, Denmark
- Correspondence: ; Tel.: +45-7940-6669
| |
Collapse
|
3
|
Moreira J, Noé G, Rangarajan S, Courtin C, Etain B, Geoffroy PA, Laplanche JL, Vidal M, Bellivier F, Marie-Claire C. Lithium effects on serine-threonine kinases activity: High throughput kinomic profiling of lymphoblastoid cell lines from excellent-responders and non-responders bipolar patients. World J Biol Psychiatry 2020; 21:317-324. [PMID: 29893160 DOI: 10.1080/15622975.2018.1487078] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Objectives: Lithium is the leading mood stabiliser for maintenance treatment in bipolar disorder (BD). However, response to lithium is heterogeneous with more than 60% of patients experiencing partial or no response. In vitro and in vivo molecular studies have reported the implication of kinases in the pathophysiology of BD.Methods: Since kinases are putative targets for lithium therapeutic action, we conducted the first pilot study using kinase array technology to evaluate the global serine/threonine kinases (STK) profiles in cell lines from BD I subtype patients classified as lithium excellent-responders (ER) and non-responder (NR) to lithium treatment.Results: We found significant differences in the basal STK profiles between ER and NR to lithium. We also tested lithium influence on the global STK profile and found no significant difference between ER vs NR cell lines.Conclusions: The results obtained in this exploratory study suggest that multiplex kinase activity profiling could provide a complementary approach in the study of biomarkers of therapeutic response in BD.
Collapse
Affiliation(s)
- Jeverson Moreira
- Variabilité de réponse aux psychotropes, INSERM U1144/Faculté de Pharmacie de Paris, Université Paris Descartes, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Gaëlle Noé
- AP-HP, Hôpital Cochin, Biologie du medicament-Toxicologie, Université Paris Descartes, Paris, France.,UMR8638 CNRS, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | | | - Cindie Courtin
- Variabilité de réponse aux psychotropes, INSERM U1144/Faculté de Pharmacie de Paris, Université Paris Descartes, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Bruno Etain
- Variabilité de réponse aux psychotropes, INSERM U1144/Faculté de Pharmacie de Paris, Université Paris Descartes, Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,AP-HP, GH Saint-Louis - Lariboisière - F. Widal, Pôle de Psychiatrie et de Médecine Addictologique, Paris, France.,Fondation FondaMental, Créteil, France
| | - Pierre A Geoffroy
- Variabilité de réponse aux psychotropes, INSERM U1144/Faculté de Pharmacie de Paris, Université Paris Descartes, Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,AP-HP, GH Saint-Louis - Lariboisière - F. Widal, Pôle de Psychiatrie et de Médecine Addictologique, Paris, France.,Fondation FondaMental, Créteil, France
| | - Jean-Louis Laplanche
- Variabilité de réponse aux psychotropes, INSERM U1144/Faculté de Pharmacie de Paris, Université Paris Descartes, Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,Département de Biochimie and Biologie moléculaire, AP-HP, GH Saint-Louis - Lariboisière - F. Widal, Paris, France
| | - Michel Vidal
- AP-HP, Hôpital Cochin, Biologie du medicament-Toxicologie, Université Paris Descartes, Paris, France.,UMR8638 CNRS, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Frank Bellivier
- Variabilité de réponse aux psychotropes, INSERM U1144/Faculté de Pharmacie de Paris, Université Paris Descartes, Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,AP-HP, GH Saint-Louis - Lariboisière - F. Widal, Pôle de Psychiatrie et de Médecine Addictologique, Paris, France.,Fondation FondaMental, Créteil, France
| | - Cynthia Marie-Claire
- Variabilité de réponse aux psychotropes, INSERM U1144/Faculté de Pharmacie de Paris, Université Paris Descartes, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
4
|
Hoorens MWH, Ourailidou ME, Rodat T, van der Wouden PE, Kobauri P, Kriegs M, Peifer C, Feringa BL, Dekker FJ, Szymanski W. Light-controlled inhibition of BRAFV600E kinase. Eur J Med Chem 2019; 179:133-146. [PMID: 31252305 DOI: 10.1016/j.ejmech.2019.06.042] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/22/2019] [Accepted: 06/15/2019] [Indexed: 11/26/2022]
Abstract
Metastatic melanoma is amongst the most difficult types of cancer to treat, with current therapies mainly relying on the inhibition of the BRAFV600E mutant kinase. However, systemic inhibition of BRAF by small molecule drugs in cancer patients results - paradoxically - in increased wild-type BRAF activity in healthy tissue, causing side-effects and even the formation of new tumors. Here we show the development of BRAFV600E kinase inhibitors of which the activity can be switched on and off reversibly with light, offering the possibility to overcome problems of systemic drug activity by selectively activating the drug at the desired site of action. Based on a known inhibitor, eight photoswitchable effectors containing an azobenzene photoswitch were designed, synthesized and evaluated. The most promising inhibitor showed an approximately 10-fold increase in activity upon light-activation. This research offers inspiration for the development of therapies for metastatic melanoma in which tumor tissue is treated with an active BRAFV600E inhibitor with high spatial and temporal resolution, thus limiting the damage to other tissues.
Collapse
Affiliation(s)
- Mark W H Hoorens
- University Medical Center Groningen, Department of Radiology, Medical Imaging Center, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands; Stratingh Institute for Chemistry, Faculty of Science and Engineering, University of Groningen, Nijenborgh 7, 9747 AG Groningen, Netherlands
| | - Maria E Ourailidou
- Department of Chemical and Pharmaceutical Biology, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, Netherlands
| | - Theo Rodat
- Institute of Pharmacy, Christian-Albrechts-University of Kiel, Gutenbergstr. 76, 24118 Kiel, Germany
| | - Petra E van der Wouden
- Department of Chemical and Pharmaceutical Biology, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, Netherlands
| | - Piermichele Kobauri
- Stratingh Institute for Chemistry, Faculty of Science and Engineering, University of Groningen, Nijenborgh 7, 9747 AG Groningen, Netherlands
| | - Malte Kriegs
- Laboratory of Radiobiology & Experimental Radiooncology and UCCH Kinomics Core Facility, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Christian Peifer
- Institute of Pharmacy, Christian-Albrechts-University of Kiel, Gutenbergstr. 76, 24118 Kiel, Germany
| | - Ben L Feringa
- Stratingh Institute for Chemistry, Faculty of Science and Engineering, University of Groningen, Nijenborgh 7, 9747 AG Groningen, Netherlands
| | - Frank J Dekker
- Department of Chemical and Pharmaceutical Biology, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, Netherlands
| | - Wiktor Szymanski
- University Medical Center Groningen, Department of Radiology, Medical Imaging Center, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands; Stratingh Institute for Chemistry, Faculty of Science and Engineering, University of Groningen, Nijenborgh 7, 9747 AG Groningen, Netherlands.
| |
Collapse
|
5
|
Meltzer S, Bjørnetrø T, Lyckander LG, Flatmark K, Dueland S, Samiappan R, Johansen C, Kalanxhi E, Ree AH, Redalen KR. Circulating Exosomal miR-141-3p and miR-375 in Metastatic Progression of Rectal Cancer. Transl Oncol 2019; 12:1038-1044. [PMID: 31146167 PMCID: PMC6542769 DOI: 10.1016/j.tranon.2019.04.014] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 04/17/2019] [Indexed: 12/21/2022] Open
Abstract
As many as 30% to 40% of locally advanced rectal cancer (LARC) patients experience metastatic progression of the disease. Recognizing the potential of the genetic cargo in tumor-derived exosomes, we hypothesized that plasma exosomal microRNA (miRNA) may reflect biological aggressiveness in LARC and provide new markers for rectal cancer aggressiveness and risk stratification. In a prospective LARC cohort (NCT01816607), plasma samples were collected from 29 patients at the time of diagnosis, before neoadjuvant therapy and surgery. Exosomes, precipitated from plasma using a commercial kit, were verified by cryo-electron microscopy, nanoparticle tracking analysis, and western blotting. Expression of exosomal miRNAs was profiled using a miRCURY LNA miRNA microarray and validation of six miRNAs associated with pathological and clinical end-points was undertaken in plasma collected at the time of diagnosis from 64 patients in an independent prospective LARC cohort (NCT00278694). In both cohorts, exosomal miR-141-3p and miR-375 were higher in patients with synchronous liver metastasis than in those without (P = .010 and P = .017 respectively in the investigative cohort, and P < .001 for both in the validation cohort). Further, high exosomal miR-141-3p was associated with post-operative metastatic liver progression in the investigative cohort (P = .034). Because both miRNAs are associated with tumor angiogenesis and immune modulation, we propose that these miRNAs in circulating exosomes may reflect rectal cancer aggressiveness and accordingly be candidate biomarkers for further investigations.
Collapse
Affiliation(s)
- Sebastian Meltzer
- Department of Oncology, Akershus University Hospital, 1478 Lørenskog, Norway.
| | - Tonje Bjørnetrø
- Department of Oncology, Akershus University Hospital, 1478 Lørenskog, Norway; Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway
| | | | - Kjersti Flatmark
- Department of Gastroenterological Surgery, Oslo University Hospital, 0424 Oslo, Norway; Department of Tumor Biology, Oslo University Hospital, 0424 Oslo, Norway; Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway
| | - Svein Dueland
- Department of Oncology, Oslo University Hospital, 0424 Oslo, Norway
| | - Rampradeep Samiappan
- Department of Bioscience and Nutrition, Karolinska Institutet, SE-141 83 Huddinge, Sweden
| | - Christin Johansen
- Department of Oncology, Akershus University Hospital, 1478 Lørenskog, Norway
| | - Erta Kalanxhi
- Department of Oncology, Akershus University Hospital, 1478 Lørenskog, Norway
| | - Anne Hansen Ree
- Department of Oncology, Akershus University Hospital, 1478 Lørenskog, Norway; Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway
| | - Kathrine Røe Redalen
- Department of Oncology, Akershus University Hospital, 1478 Lørenskog, Norway; Department of Physics, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| |
Collapse
|
6
|
Schwill M, Tamaskovic R, Gajadhar AS, Kast F, White FM, Plückthun A. Systemic analysis of tyrosine kinase signaling reveals a common adaptive response program in a HER2-positive breast cancer. Sci Signal 2019; 12:12/565/eaau2875. [PMID: 30670633 DOI: 10.1126/scisignal.aau2875] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Drug-induced compensatory signaling and subsequent rewiring of the signaling pathways that support cell proliferation and survival promote the development of acquired drug resistance in tumors. Here, we sought to analyze the adaptive kinase response in cancer cells after distinct treatment with agents targeting human epidermal growth factor receptor 2 (HER2), specifically those that induce either only temporary cell cycle arrest or, alternatively, apoptosis in HER2-overexpressing cancers. We compared trastuzumab, ARRY380, the combination thereof, and a biparatopic, HER2-targeted designed ankyrin repeat protein (DARPin; specifically, 6L1G) and quantified the phosphoproteome by isobaric tagging using tandem mass tag liquid chromatography/tandem mass spectrometry (TMT LC-MS/MS). We found a specific signature of persistently phosphorylated tyrosine peptides after the nonapoptotic treatments, which we used to distinguish between different treatment-induced cancer cell fates. Next, we analyzed the activation of serine/threonine and tyrosine kinases after treatment using a bait peptide chip array and predicted the corresponding active kinases. Through a combined system-wide analysis, we identified a common adaptive kinase response program that involved the activation of focal adhesion kinase 1 (FAK1), protein kinase C-δ (PRKCD), and Ephrin (EPH) family receptors. These findings reveal potential targets to prevent adaptive resistance to HER2-targeted therapies.
Collapse
Affiliation(s)
- Martin Schwill
- Department of Biochemistry, University of Zurich, Winterthurerstr. 190, 8057 Zurich, Switzerland
| | - Rastislav Tamaskovic
- Department of Biochemistry, University of Zurich, Winterthurerstr. 190, 8057 Zurich, Switzerland
| | - Aaron S Gajadhar
- Department of Biological Engineering, Koch Institute for Integrative Cancer Research, Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Florian Kast
- Department of Biochemistry, University of Zurich, Winterthurerstr. 190, 8057 Zurich, Switzerland
| | - Forest M White
- Department of Biological Engineering, Koch Institute for Integrative Cancer Research, Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Andreas Plückthun
- Department of Biochemistry, University of Zurich, Winterthurerstr. 190, 8057 Zurich, Switzerland.
| |
Collapse
|
7
|
Noé G, Bellesoeur A, Thomas-Schoemann A, Rangarajan S, Naji F, Puszkiel A, Huillard O, Saidu N, Golmard L, Alexandre J, Goldwasser F, Blanchet B, Vidal M. Clinical and kinomic analysis identifies peripheral blood mononuclear cells as a potential pharmacodynamic biomarker in metastatic renal cell carcinoma patients treated with sunitinib. Oncotarget 2018; 7:67507-67520. [PMID: 27589830 PMCID: PMC5341893 DOI: 10.18632/oncotarget.11686] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 07/10/2016] [Indexed: 12/31/2022] Open
Abstract
Background Sunitinib is a protein tyrosine kinase (PTK) inhibitor that has immune-modulating properties. In this context, peripheral blood mononuclear cells (PBMC), mainly constituted by lymphocytes, could be a perfect surrogate tissue for identifying and assaying pharmacodynamic biomarkers of sunitinib. In this study, we investigated the changes in lymphocytes count as pharmacodynamic biomarker in metastatic renal cell carcinoma (mRCC) patients under sunitinib therapy. Thereafter, we studied the ex vivo effect of sunitinib and SU12262 (active metabolite) on PBMC from naïve mRCC patients using a high throughput kinomic profiling method. Methods The prognostic value of total lymphocytes count between Day 0 and Day 21 (expressed as a ratio D21/D0) was retrospectively investigated in 88 mRCC patients under sunitinib therapy. PTK PamChip® microarrays were used to explore prospectively the ex vivo effect of sunitinib and SU12662 on PTK activity in PBMC from 21 naïve mRCC patients. Results In this retrospective study, D21/D0 lymphocytes ratio (Hazard Ratio, 1.83; CI95%, 1.24-2.71; p=0.0023) was independently associated with PFS. Interestingly, kinomic analysis showed that D21/D0 lymphocytes ratio and Heng prognostic model was statistically associated with the ex vivo sunitinib and SU12662 effect in PBMC. Conclusion The present study highlights that D21/D0 total lymphocytes ratio could be a promising pharmacodynamic biomarker in mRCC patients treated with sunitinib. Additionally, it paves the way to investigate the kinomic profile in PBMC as a prognostic factor in a larger cohort of mRCC patients under sunitinib therapy.
Collapse
Affiliation(s)
- Gaёlle Noé
- Assistance Publique Hôpitaux de Paris, Hôpital Cochin, UF Pharmacocinétique et Pharmacochimie, Paris, France.,UMR8638 CNRS, Faculté de Pharmacie, Université Paris Descartes, PRES Sorbonne Paris Cité, Paris, France
| | - Audrey Bellesoeur
- Assistance Publique Hôpitaux de Paris, Hôpital Cochin, UF Pharmacocinétique et Pharmacochimie, Paris, France.,UMR8638 CNRS, Faculté de Pharmacie, Université Paris Descartes, PRES Sorbonne Paris Cité, Paris, France.,Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Service de Cancérologie Médicale, Paris, France
| | - Audrey Thomas-Schoemann
- Assistance Publique Hôpitaux de Paris, Hôpital Cochin, UF Pharmacocinétique et Pharmacochimie, Paris, France.,UMR8638 CNRS, Faculté de Pharmacie, Université Paris Descartes, PRES Sorbonne Paris Cité, Paris, France
| | | | - Faris Naji
- PamGene International BV, 's-Hertogenbosch, The Netherlands
| | - Alicja Puszkiel
- Assistance Publique Hôpitaux de Paris, Hôpital Cochin, UF Pharmacocinétique et Pharmacochimie, Paris, France
| | - Olivier Huillard
- Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Service de Cancérologie Médicale, Paris, France
| | - Nathaniel Saidu
- U1016 INSERM, UMR 8104 CNRS, UMR-S1016, CARPEM, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Lisa Golmard
- Institut Curie, Département de Biopathologie, Paris, France
| | - Jerome Alexandre
- Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Service de Cancérologie Médicale, Paris, France.,U1016 INSERM, UMR 8104 CNRS, UMR-S1016, CARPEM, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Francois Goldwasser
- Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Service de Cancérologie Médicale, Paris, France.,U1016 INSERM, UMR 8104 CNRS, UMR-S1016, CARPEM, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Benoit Blanchet
- Assistance Publique Hôpitaux de Paris, Hôpital Cochin, UF Pharmacocinétique et Pharmacochimie, Paris, France
| | - Michel Vidal
- Assistance Publique Hôpitaux de Paris, Hôpital Cochin, UF Pharmacocinétique et Pharmacochimie, Paris, France.,UMR8638 CNRS, Faculté de Pharmacie, Université Paris Descartes, PRES Sorbonne Paris Cité, Paris, France
| |
Collapse
|
8
|
Large-scale reduction of tyrosine kinase activities in human monocytes stimulated in vitro with N. meningitidis. PLoS One 2018; 13:e0181912. [PMID: 29357362 PMCID: PMC5774972 DOI: 10.1371/journal.pone.0181912] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 07/10/2017] [Indexed: 01/07/2023] Open
Abstract
N. meningitidis induces extensive gene expression changes in human monocytes, suggesting that complex networks of signaling pathways are activated during meningococcal sepsis. These effects are modulated by the anti-inflammatory cytokine interleukin-10 (IL-10). To further study changes in signal transduction suggested by mRNA data, we used kinase substrate arrays to identify composite kinase activities induced by lysates from a primary human monocyte model system. Cell lysates were prepared from monocytes treated with the following experimental conditions: 106 N. meningitidis/mL, 25 ng/mL IL-10, 106 N. meningitidis/mL in combination with 25 ng/mL IL-10, and vehicle. Lysates were subjected to kinase activity profiling with Tyrosine Kinase PamChip® arrays containing 144 kinase peptide substrates. In our experimental model, we were not able to detect a statistically significant large-scale change in ex vivo array peptide phosphorylation by lysates from monocytes treated for 15 minutes. Targets of the IL-10 anti-inflammatory response were not identified. A profound inhibition of array peptide phosphorylation by monocytes treated for 60 minutes was identified, suggesting low activity of a large number of kinases associated with different signaling pathways and immune cell functions, including STAT3 activity, Nf-κB and VEGF signaling, and PTEN signaling activity. The peptide representing ZBTB16, which was reduced in phosphorylation by lysates from all three experimental conditions, was in Ingenuity Pathway Analysis identified to be linked to reduced cytokine release and mRNA levels of tumor necrosis factor (TNF), IL-6, and CXCL10. Further studies should investigate changes in tyrosine kinase-mediated signal transduction in human immune cells, in order to evaluate the potential clinical application of kinome profiling in the study of systemic inflammatory responses to pathogens.
Collapse
|
9
|
PI-273, a Substrate-Competitive, Specific Small-Molecule Inhibitor of PI4KIIα, Inhibits the Growth of Breast Cancer Cells. Cancer Res 2017; 77:6253-6266. [DOI: 10.1158/0008-5472.can-17-0484] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 06/29/2017] [Accepted: 08/14/2017] [Indexed: 11/16/2022]
|
10
|
Technological advances for interrogating the human kinome. Biochem Soc Trans 2017; 45:65-77. [PMID: 28202660 DOI: 10.1042/bst20160163] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 10/20/2016] [Accepted: 10/25/2016] [Indexed: 12/12/2022]
Abstract
There is increasing appreciation among researchers and clinicians of the value of investigating biology and pathobiology at the level of cellular kinase (kinome) activity. Kinome analysis provides valuable opportunity to gain insights into complex biology (including disease pathology), identify biomarkers of critical phenotypes (including disease prognosis and evaluation of therapeutic efficacy), and identify targets for therapeutic intervention through kinase inhibitors. The growing interest in kinome analysis has fueled efforts to develop and optimize technologies that enable characterization of phosphorylation-mediated signaling events in a cost-effective, high-throughput manner. In this review, we highlight recent advances to the central technologies currently available for kinome profiling and offer our perspectives on the key challenges remaining to be addressed.
Collapse
|
11
|
Risberg K, Redalen KR, Sønstevold L, Bjørnetrø T, Sølvernes J, Ree AH. Pro-survival responses to the dual inhibition of anti-apoptotic Bcl-2 family proteins and mTOR-mediated signaling in hypoxic colorectal carcinoma cells. BMC Cancer 2016; 16:531. [PMID: 27461218 PMCID: PMC4962454 DOI: 10.1186/s12885-016-2600-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 07/22/2016] [Indexed: 12/16/2022] Open
Abstract
Background The use of targeted agents to impel dual inhibition of anti-apoptotic mechanisms and mTOR-mediated pro-survival signaling in colorectal carcinoma (CRC) cell lines with KRAS or BRAF mutation has been shown to induce apoptosis, a timely result given CRC entities harboring such mutations are in need of new therapies. Since CRC comprises heterogeneous tumors with predominant hypoxic components, we investigated effects of an inhibitor of anti-apoptotic Bcl-2 family proteins (ABT-737) in combination with an mTOR inhibitor (AZD8055)—collectively referred to as combo-Rx, in hypoxic CRC cell lines. Methods Cell viability measures, expression of proteins implicated in apoptosis and MAPK/PI3K-AKT/mTOR pathway signaling, and profiling of composite kinase activities were undertaken in a panel of 14 cell lines. Results In hypoxic conditions, combo-Rx suppressed viability of 13 of the cell lines, albeit ABT-737 did not significantly potentiate the inhibitory effect of single-agent AZD8055 in six of the models. Hypoxic KRAS/PIK3CA-mutant HCT-116 and HCT-15 cell lines (both with low endogenous expression of the anti-apoptotic Mcl-1 protein and showing augmented inhibition of viability following the addition of ABT-737 to AZD8055) responded to combo-Rx by induction of apoptosis but with the simultaneous strong Mcl-1 up-regulation and activation of MAPK/PI3K-conducted signaling. In contrast, in hypoxic KRAS-mutant LoVo (devoid of PIK3CA mutation), BRAF/PIK3CA-mutant RKO, and wild-type Colo320DM cell lines (all with high endogenous Mcl-1 expression and being resistant to the additional effect of ABT-737 to AZD8055), combo-Rx did not elicit apoptotic or pro-survival responses. Conclusions The concurrent inhibition of anti-apoptotic proteins and mTOR-mediated signaling in hypoxic KRAS/PIK3CA-mutant CRC cell lines resulted in pro-survival responses in parallel with the intended anti-proliferative effects, a finding that should be of note if considering combinatory targeting of multiple pathways in this CRC entity. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2600-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Karianne Risberg
- Department of Oncology, Akershus University Hospital, 1478, Lørenskog, Norway.,Institute of Clinical Molecular Biology, Akershus University Hospital, 1478, Lørenskog, Norway
| | | | - Linda Sønstevold
- Department of Oncology, Akershus University Hospital, 1478, Lørenskog, Norway
| | - Tonje Bjørnetrø
- Department of Oncology, Akershus University Hospital, 1478, Lørenskog, Norway.,Institute of Clinical Medicine, University of Oslo, 0318, Oslo, Norway
| | - Janne Sølvernes
- Department of Oncology, Akershus University Hospital, 1478, Lørenskog, Norway
| | - Anne Hansen Ree
- Department of Oncology, Akershus University Hospital, 1478, Lørenskog, Norway. .,Institute of Clinical Medicine, University of Oslo, 0318, Oslo, Norway.
| |
Collapse
|
12
|
Kalanxhi E, Hektoen HH, Meltzer S, Dueland S, Flatmark K, Ree AH. Circulating proteins in response to combined-modality therapy in rectal cancer identified by antibody array screening. BMC Cancer 2016; 16:536. [PMID: 27461255 PMCID: PMC4962367 DOI: 10.1186/s12885-016-2601-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 07/22/2016] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The increasingly complex programs of contemporary cancer therapy emphasize the need for biological indicators of both therapeutic response and adverse effects. One example is combined-modality treatment aimed at improving long-term outcome in patients with locally advanced rectal cancer, which commonly comes at the price of extended limits of patient tolerance. METHODS In a prospective study with intensified neoadjuvant treatment of rectal cancer patients, using an antibody array, the profiling of approximately 500 proteins was performed in serial serum samples collected at different stages of the treatment course. RESULTS The small number of proteins whose levels significantly changed after induction neoadjuvant chemotherapy (NACT) expanded substantially following the sequential chemoradiotherapy (CRT) and persisted four weeks later at treatment evaluation before pelvic surgery. Serum levels of proteins selected for validation of the experimental design, lipocalin-2 and matrix metalloproteinase-9, declined after NACT and gradually reverted to baseline values during the remaining neoadjuvant course. Of note, the greater the decline in post-NACT and post-CRT matrix metalloproteinase-9 levels, the more favorable progression-free survival. No correlation was found, however, with diarrhea scores, the clinical correlate of adverse therapeutic effects. CONCLUSIONS Even though the findings were indicative of only tumor and not normal tissue effects, multiplex immunoassay analysis of circulating proteins in patients undergoing combined-modality therapy may in principle dissect the contribution of the individual modalities to overall systemic responses in patient outcome and tolerance. TRIAL REGISTRATION ClinicalTrials.gov NCT00278694 ; registration date: January 16, 2006, retrospective to enrollment of the first 10 patients of the current report.
Collapse
Affiliation(s)
- Erta Kalanxhi
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway.,Institute of Clinical Molecular Biology, Akershus University Hospital, P.O. Box 1000, 1478, Lørenskog, Norway
| | - Helga Helseth Hektoen
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway.,Institute of Clinical Medicine, University of Oslo, P.O. Box 1171, Blindern, 0318, Oslo, Norway.,Department of Tumor Biology, Oslo University Hospital - Norwegian Radium Hospital, Oslo, Norway
| | - Sebastian Meltzer
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway.,Institute of Clinical Medicine, University of Oslo, P.O. Box 1171, Blindern, 0318, Oslo, Norway
| | - Svein Dueland
- Department of Oncology, Oslo University Hospital - Norwegian Radium Hospital, Oslo, Norway
| | - Kjersti Flatmark
- Institute of Clinical Medicine, University of Oslo, P.O. Box 1171, Blindern, 0318, Oslo, Norway.,Department of Tumor Biology, Oslo University Hospital - Norwegian Radium Hospital, Oslo, Norway.,Department of Gastroenterological Surgery, Oslo University Hospital - Norwegian Radium Hospital, P.O. Box 4950, Nydalen, 0424, Oslo, Norway
| | - Anne Hansen Ree
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway. .,Institute of Clinical Medicine, University of Oslo, P.O. Box 1171, Blindern, 0318, Oslo, Norway.
| |
Collapse
|
13
|
Abstract
Preoperative radiotherapy has an accepted role in reducing the risk of local recurrence in locally advanced resectable rectal cancer, particularly when the circumferential resection margin is breached or threatened, according to magnetic resonance imaging. Fluoropyrimidine-based chemoradiation can obtain a significant down-sizing response and a curative resection can then be achieved. Approximately, 20% of the patients can also obtain a pathological complete response, which is associated with less local recurrences and increased survival. Patients who achieve a sustained complete clinical response may also avoid radical surgery. In unresectable or borderline resectable tumors, around 20% of the patients still fail to achieve a sufficient down-staging response with the current chemoradiation schedules. Hence, investigators have aspired to increase pathological complete response rates, aiming to improve curative resection rates, enhance survival, and potentially avoid mutilating surgery. However, adding additional cytotoxic or biological agents have not produced dramatic improvements in outcome and often led to excess surgical morbidity and higher levels of acute toxicity, which effects on compliance and in the global efficacy of chemoradiation.
Collapse
Affiliation(s)
- Rob Glynne-Jones
- Mount Vernon Centre for Cancer Treatment, Northwood, Middlesex, UK.
| | | |
Collapse
|
14
|
Hektoen HH, Ree AH, Redalen KR, Flatmark K. Sulfamate inhibitor S4 influences carbonic anhydrase IX ectodomain shedding in colorectal carcinoma cells. J Enzyme Inhib Med Chem 2015; 31:779-86. [PMID: 26244271 DOI: 10.3109/14756366.2015.1069286] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Carbonic anhydrase IX (CAIX) is a pivotal pH regulator under hypoxia, which by its tumor-specific expression represents an attractive target for cancer therapy. Here, we report on effects of the sulfamate CAIX inhibitor S4 (4-(3'-(3″,5″-dimethylphenyl)ureido)phenyl sulfamate) in colorectal carcinoma cell lines. S4 was administered under experimental hypoxia or normoxia to HT29, KM20L2 and HCT116 cells. Effects on survival, proliferation, pH, lactate extrusion and CAIX protein expression were evaluated. S4 treatment resulted in attenuated hypoxia-induced extracellular acidification and reduced clonogenic survival under hypoxia in HT29 cells. The pH effects were present only in a [Formula: see text]-free buffer system and were accompanied by decreased lactate extrusion. The main finding of this work was that S4 treatment caused alterations in CAIX ectodomain shedding. This merits further investigation to understand how sulfamates influence CAIX activity and how such drugs may be of use in cancer treatment.
Collapse
Affiliation(s)
- Helga Helseth Hektoen
- a Institute of Clinical Medicine, University of Oslo , Oslo , Norway .,b Department of Tumor Biology , Oslo University Hospital , Oslo , Norway .,c Department of Oncology , Akershus University Hospital , Lørenskog , Norway , and
| | - Anne Hansen Ree
- a Institute of Clinical Medicine, University of Oslo , Oslo , Norway .,c Department of Oncology , Akershus University Hospital , Lørenskog , Norway , and
| | - Kathrine Røe Redalen
- c Department of Oncology , Akershus University Hospital , Lørenskog , Norway , and
| | - Kjersti Flatmark
- a Institute of Clinical Medicine, University of Oslo , Oslo , Norway .,b Department of Tumor Biology , Oslo University Hospital , Oslo , Norway .,d Department of Gastroenterological Surgery , Oslo University Hospital , Oslo , Norway
| |
Collapse
|
15
|
Ree AH, Redalen KR. Personalized radiotherapy: concepts, biomarkers and trial design. Br J Radiol 2015; 88:20150009. [PMID: 25989697 DOI: 10.1259/bjr.20150009] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In the past decade, and pointing onwards to the immediate future, clinical radiotherapy has undergone considerable developments, essentially including technological advances to sculpt radiation delivery, the demonstration of the benefit of adding concomitant cytotoxic agents to radiotherapy for a range of tumour types and, intriguingly, the increasing integration of targeted therapeutics for biological optimization of radiation effects. Recent molecular and imaging insights into radiobiology will provide a unique opportunity for rational patient treatment, enabling the parallel design of next-generation trials that formally examine the therapeutic outcome of adding targeted drugs to radiation, together with the critically important assessment of radiation volume and dose-limiting treatment toxicities. In considering the use of systemic agents with presumed radiosensitizing activity, this may also include the identification of molecular, metabolic and imaging markers of treatment response and tolerability, and will need particular attention on patient eligibility. In addition to providing an overview of clinical biomarker studies relevant for personalized radiotherapy, this communication will highlight principles in addressing clinical evaluation of combined-modality-targeted therapeutics and radiation. The increasing number of translational studies that bridge large-scale omics sciences with quality-assured phenomics end points-given the imperative development of open-source data repositories to allow investigators the access to the complex data sets-will enable radiation oncology to continue to position itself with the highest level of evidence within existing clinical practice.
Collapse
Affiliation(s)
- A H Ree
- 1 Department of Oncology, Akershus University Hospital, Lørenskog, Norway.,2 Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - K R Redalen
- 1 Department of Oncology, Akershus University Hospital, Lørenskog, Norway
| |
Collapse
|