209 results on '"Sabine Tejpar"'
Search Results
2. Targeting the bicarbonate transporter SLC4A4 overcomes immunosuppression and immunotherapy resistance in pancreatic cancer
- Author
-
Federica Cappellesso, Marie-Pauline Orban, Niranjan Shirgaonkar, Emanuele Berardi, Jens Serneels, Marie-Aline Neveu, Daria Di Molfetta, Francesca Piccapane, Rosa Caroppo, Lucantonio Debellis, Tessa Ostyn, Nicolas Joudiou, Lionel Mignion, Elena Richiardone, Bénédicte F. Jordan, Bernard Gallez, Cyril Corbet, Tania Roskams, Ramanuj DasGupta, Sabine Tejpar, Mario Di Matteo, Daniela Taverna, Stephan J. Reshkin, Baki Topal, Federico Virga, Massimiliano Mazzone, UCL - SSS/IREC/FATH - Pôle de Pharmacologie et thérapeutique, and UCL - SSS/LDRI - Louvain Drug Research Institute
- Subjects
LACTIC-ACID ,Cancer Research ,Science & Technology ,MIGRATION ,PH ,GLYCOLYSIS ,ANGIOGENESIS ,FAMILY ,Oncology ,CELLS ,SECRETION ,Life Sciences & Biomedicine ,SPECIFICITY - Abstract
Solid tumors are generally characterized by an acidic tumor microenvironment (TME) that favors cancer progression, therapy resistance and immune evasion. By single-cell RNA-sequencing analysis in individuals with pancreatic ductal adenocarcinoma (PDAC), we reveal solute carrier family 4 member 4 (SLC4A4) as the most abundant bicarbonate transporter, predominantly expressed by epithelial ductal cells. Functionally, SLC4A4 inhibition in PDAC cancer cells mitigates the acidosis of the TME due to bicarbonate accumulation in the extracellular space and a decrease in lactate production by cancer cells as the result of reduced glycolysis. In PDAC-bearing mice, genetic or pharmacological SLC4A4 targeting improves T cell-mediated immune response and breaches macrophage-mediated immunosuppression, thus inhibiting tumor growth and metastases. In addition, Slc4a4 targeting in combination with immune checkpoint blockade is able to overcome immunotherapy resistance and prolong survival. Overall, our data propose SLC4A4 as a therapeutic target to unleash an antitumor immune response in PDAC. ispartof: NATURE CANCER vol:3 issue:12 ispartof: location:England status: Published online
- Published
- 2022
3. Mex3a marks drug-tolerant persister colorectal cancer cells that mediate relapse after chemotherapy
- Author
-
Adrián Álvarez-Varela, Laura Novellasdemunt, Francisco M. Barriga, Xavier Hernando-Momblona, Adrià Cañellas-Socias, Sara Cano-Crespo, Marta Sevillano, Carme Cortina, Diana Stork, Clara Morral, Gemma Turon, Felipe Slebe, Laura Jiménez-Gracia, Ginevra Caratù, Peter Jung, Giorgio Stassi, Holger Heyn, Daniele V. F. Tauriello, Lidia Mateo, Sabine Tejpar, Elena Sancho, Camille Stephan-Otto Attolini, and Eduard Batlle
- Subjects
Cancer Research ,Cell Differentiation ,Colorectal cancer ,Organoids ,Mice ,Quimioteràpia del càncer ,Oncology ,Recurrence ,Càncer colorectal ,Neoplastic Stem Cells ,Animals ,Cancer chemotherapy ,Colorectal Neoplasms ,Nanomedicine Radboud Institute for Molecular Life Sciences [Radboudumc 19] - Abstract
Colorectal cancer (CRC) patient-derived organoids predict responses to chemotherapy. Here we used them to investigate relapse after treatment. Patient-derived organoids expand from highly proliferative LGR5(+) tumor cells; however, we discovered that lack of optimal growth conditions specifies a latent LGR5(+) cell state. This cell population expressed the gene MEX3A, is chemoresistant and regenerated the organoid culture after treatment. In CRC mouse models, Mex3a(+) cells contributed marginally to metastatic outgrowth; however, after chemotherapy, Mex3a(+) cells produced large cell clones that regenerated the disease. Lineage-tracing analysis showed that persister Mex3a(+) cells downregulate the WNT/stem cell gene program immediately after chemotherapy and adopt a transient state reminiscent to that of YAP(+) fetal intestinal progenitors. In contrast, Mex3a-deficient cells differentiated toward a goblet cell-like phenotype and were unable to resist chemotherapy. Our findings reveal that adaptation of cancer stem cells to suboptimal niche environments protects them from chemotherapy and identify a candidate cell of origin of relapse after treatment in CRC. Batlle and colleagues report that after chemotherapy, Mex3a(+) colorectal cancer cells represent drug-tolerant persister cells that lead to recurrence by downregulating the WNT-Lgr5(+) stem cell program and adopting a transient regenerative state.
- Published
- 2022
4. Abstract 6770: Spatial characterization of immune microenvironment from early onset metastatic colorectal cancer (EOmCRC) showed a dual prognostic role for IDO1 expression
- Author
-
Raghavendar Thyagaraja Nagaraju, Jakub Franczak, Markus Möhler, Elisa Fontana, Anne Giraut, Lieve Dirix, Jose Casas-Martin, Beatrice Borelli, Florian Lordick, Elizabeth Smith, Sabine Tejpar, Gunnar Folprecht, and Jorge Barriuso
- Subjects
Cancer Research ,Oncology - Abstract
Background: Epidemiological studies demonstrate that the incidence of CRC in young adults is continually rising in both Europe and the USA. The differences between the spatial distribution of immune related markers (IRM) and their prognostic role in EOmCRC compared to late onset (LOmCRC) remains to be elucidated. Methods: Forty paraffin embedded formalin fixed unstained sections from patients (pts) diagnosed with either EOmCRC (≤45) or (≥50) LOmCRC, matched for chemotherapy (chx) and molecular features, all with microsatellite stable disease were included. All tissue samples were obtained from pts enrolled in SPECTAcolor (EORTC-40CRC-GITCG) platform trial. The expression of IRM was studied using the GeoMX Digital Spatial Profiler (DSP). Fifty-seven IRMs were used and three morphology markers (MMs) anti-CD45, pan-cytokeratin and SYTO 13. Three 300 μm regions of interest (ROIs) were selected within the core of the tumour and further 3 ROIs within the active margin of the tumour. Each ROI was segmented based on MMs to obtain a tumour microenvironment (TME) and a tumour (T) mask. The analysis used the GeoMX DSP Advanced Analysis Suite software, R and IBM SPSS. Control for multiple comparisons use a false discovery rate (FDR) threshold of 0.05. Results: Samples from 40 pts were collected; 19 (47.5%) were EOmCRCs; 16 (40%) were female; 23 (57.5%) with pathological stage IV. The majority being, 23 (57.5%) grade ≥ 2, left sided 26 (65%), oxaliplatin based first line chx 23 (57.5%). Ten (25%) were KRAS mutant (mt), 1 (2.5%) NRAS mt and 1 (2.5%) BRAF mt. The median overall (OS) and progression free survival (PFS) for the entire cohort was 36.6 months (m) 95%CI (33.5 - 39.7) and 11.7 m (8.1 - 15.3), respectively.The Conclusion: Differential expression analysis of IRMs found IDO1 as differentially expressed in both segments (T and TME). Furthermore, high IDO1 expression seemed to have a dual prognostic role depending on the segment analysed, showing worse OS when found in the tumour but better OS when found in tumour microenvironment for early onset mCRC. Citation Format: Raghavendar Thyagaraja Nagaraju, Jakub Franczak, Markus Möhler, Elisa Fontana, Anne Giraut, Lieve Dirix, Jose Casas-Martin, Beatrice Borelli, Florian Lordick, Elizabeth Smith, Sabine Tejpar, Gunnar Folprecht, Jorge Barriuso. Spatial characterization of immune microenvironment from early onset metastatic colorectal cancer (EOmCRC) showed a dual prognostic role for IDO1 expression. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 6770.
- Published
- 2023
5. Clinical and molecular characteristics and treatment outcomes of advanced right-colon, left-colon and rectal cancers: data from 1180 patients in a phase III trial of panitumumab with an extended biomarker panel
- Author
-
Jenny F. Seligmann, Bart Jacobs, Gemma Hemmings, Philip Quirke, Susan D. Richman, Christopher Williams, Sarah Brown, Faye Elliott, Matthew T. Seymour, Sabine Tejpar, and Jennifer H. Barrett
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Colorectal cancer ,tumour location ,Epiregulin ,Proto-Oncogene Proteins p21(ras) ,03 medical and health sciences ,0302 clinical medicine ,Amphiregulin ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Panitumumab ,colorectal ,Predictive marker ,Rectal Neoplasms ,business.industry ,Hazard ratio ,Hematology ,medicine.disease ,Irinotecan ,Treatment Outcome ,030104 developmental biology ,030220 oncology & carcinogenesis ,Mutation ,embryonic structures ,epiregulin ,Biomarker (medicine) ,precision ,amphiregulin ,panitumumab ,Colorectal Neoplasms ,business ,Biomarkers ,medicine.drug - Abstract
BACKGROUND: Primary tumour location (PTL) is being adopted by clinicians to guide treatment decisions in metastatic colorectal cancer (mCRC). Here we test PTL as a predictive marker for panitumumab efficacy, and examine its relationship with an extended biomarker profile. We also examine rectal tumours as a separate location. PATIENTS AND METHODS: mCRC patients from the second-line PICCOLO trial of irinotecan versus irinotecan/panitumumab (IrPan). PTL was classified as right-PTL, left-PTL or rectal-PTL. PTL was assessed as a predictive biomarker for IrPan effect in RAS-wild-type (RAS-wt) patients (compared with irinotecan alone), then tested for independence alongside an extended biomarker profile (BRAF, epiregulin/amphiregulin (EREG/AREG) and HER3 mRNA expression). RESULTS: PTL data were available for 1180 patients (98.5%), of whom 558 were RAS-wt. High HER3 expression was independently predictive of panitumumab overall survival improvement, but PTL and EREG/AREG were not. IrPan progression-free survival (PFS) improvement compared with irinotecan was seen in left-PTL [hazard ratio (HR) = 0.61, P = 0.002) but not right-PTL (HR = 0.98, P = 0.90) (interaction P = 0.05; RAS/BRAF-wt interaction P = 0.10), or in rectal-PTL (HR = 0.82, P = 0.20) (interaction P = 0.14 compared with left-PTL; RAS/BRAF-wt interaction P = 0.04). Patients with right-PTL and high EREG/AREG or HER3 expression, had IrPan PFS improvement (high EREG/AREG HR = 0.20, P = 0.04; high HER3 HR = 0.33, P = 0.10) compared with irinotecan. Similar effect was seen for rectal-PTL patients (high EREG/AREG HR = 0.44, P = 0.03; high HER3 HR = 0.34, P = 0.05). CONCLUSIONS: RAS-wt patients with left-PTL are more likely to have panitumumab PFS advantage than those with right-PTL or rectal-PTL. However, an extended biomarker panel demonstrated significant heterogeneity in panitumumab PFS effect within a tumour location. AREG/EREG and HER3 mRNA expression identifies patients with right-PTL or rectal-PTL who achieve similar PFS effect with panitumumab as left-colon patients. Testing could provide a more reliable basis for clinical decision making. Further validation and development of these biomarkers is required to optimise routine patient care. CLINICAL TRIAL REGISTRATION: ISRCTN identifier: ISRCTN93248876. ispartof: ANNALS OF ONCOLOGY vol:31 issue:8 pages:1021-1029 ispartof: location:England status: published
- Published
- 2020
6. Zeb2 drives invasive and microbiota-dependent colon carcinoma
- Author
-
Emre Etlioglu, Joachim Taminau, Bart N. Lambrecht, Pieter Van Vlierberghe, Geert van Loo, Niels Vandamme, Enrico Radaelli, Steven Goossens, Gillian Blancke, Jody J. Haigh, Louis Boon, Pratyaksha Wirapati, Pamela Baldin, Mozes Sze, Hanna-Kaisa Vikkula, Chris Callewaert, Gert De Hertogh, Emilie Dumas, Eugene Tulchinsky, Ioanna Petta, Andy Wullaert, Geert Berx, Sabine Tejpar, Sven Jonckheere, David Nittner, Karolina Slowicka, Esther Hoste, and Lars Vereecke
- Subjects
Cancer Research ,Intestinal permeability ,business.industry ,Colorectal cancer ,Transgene ,Cell ,Inflammation ,medicine.disease ,Intestinal epithelium ,Epithelium ,Metastasis ,medicine.anatomical_structure ,Oncology ,medicine ,Cancer research ,medicine.symptom ,business - Abstract
Colorectal cancer (CRC) is highly prevalent in Western society, and increasing evidence indicates strong contributions of environmental factors and the intestinal microbiota to CRC initiation, progression and even metastasis. We have identified a synergistic inflammatory tumor-promoting mechanism through which the resident intestinal microbiota boosts invasive CRC development in an epithelial-to-mesenchymal transition-prone tissue environment. Intestinal epithelial cell (IEC)-specific transgenic expression of the epithelial-to-mesenchymal transition regulator Zeb2 in mice (Zeb2IEC-Tg/+) leads to increased intestinal permeability, myeloid cell-driven inflammation and spontaneous invasive CRC development. Zeb2IEC-Tg/+ mice develop a dysplastic colonic epithelium, which progresses to severely inflamed neoplastic lesions while the small intestinal epithelium remains normal. Zeb2IEC-Tg/+ mice are characterized by intestinal dysbiosis, and microbiota depletion with broad-spectrum antibiotics or germ-free rederivation completely prevents cancer development. Zeb2IEC-Tg/+ mice represent the first mouse model of spontaneous microbiota-dependent invasive CRC and will help us to better understand host–microbiome interactions driving CRC development in humans. Van Loo and colleagues report that loss of the Zeb2 regulator of epithelial-to-mesenchymal transition from the intestinal epithelium leads to inflammation, increased intestinal permeability and colorectal cancer development, which is enhanced by the resident intestinal microbiome.
- Published
- 2020
7. Next-generation whole blood transcriptome-based assay for advanced adenoma detection
- Author
-
Sahar Hosseinian Ehrensberger, Victoria Wosika, Noushin Hadadi, Sara Fonseca Costa, Eric Durandau, Sylvain Monnier-Benoit, Laura Ciarloni, Stefan J. Scherer, and Sabine Tejpar
- Subjects
Cancer Research ,Oncology - Abstract
77 Background: Colorectal Cancer (CRC) is the second leading cause of cancer mortality worldwide although highly curable if detected early. An accurate liquid biopsy test for early cancer and precancerous lesion, detection is key to promote prevention and reduce mortality. We previously developed a liquid biopsy test based on immune cells response to the tumor, commercialised under the name of Colox, with best-in-class clinical performances for AA detection (52% sensitivity and 92% specificity) (Ciarloni L et al, Clin Cancer Res, 2016). An improved second generation of the test, leveraging transcriptome profiling and advanced machine learning tools, is under development and first results are presented. Methods: Prospective peripheral whole-blood (PAXgene) samples from subjects diagnosed with CRC, advanced adenoma (AA), non-advanced adenoma, other types of cancer as well as controls without colorectal neoplasia (CON) were divided into discovery and validation sets. Transcriptome profiles were generated by RNA-seq and gene expression signatures identified leveraging Novigenix’s proprietary LITOseek platform, which integrates several advanced Machine Learning (ML) methods into a ranking systems. Biological functional analysis was performed by over-representation (ORA), gene set enrichment (GSEA) and gene network analyses (STRINGdb). Results: Significant changes in the whole blood transcriptome profile of AA compared to CON were identified. Functional analysis highlighted upregulation of fatty acid derivative biosynthesis, interferon signaling, tryptophan catabolism to kynurenine, and downregulation of DNA repair in AA blood samples compared to CON. A novel gene classifier was generated, showing for AA detection 71% sensitivity, 94% specificity and an AUC of 74% by cross-validation on the discovery set. Validation of the gene classifier in an independent set is currently ongoing and will be presented at the congress. Conclusions: Capturing immune-related information using whole blood trascriptome profiling and applying cutting-edge machine learning technologies demonstrated to be valuable for identifying gene signatures for advanced adenoma detection. This differentiated solution has demonstrated best-in-class potential to significantly improve patient outcomes by detecting precancerous lesions with a simple non-invasive blood test.
- Published
- 2023
8. Prevalence of Germline Pathogenic Variants in Cancer Predisposing Genes in Czech and Belgian Pancreatic Cancer Patients
- Author
-
Jenneke van den Ende, Encarna Gomez Garcia, Marianna Borecka, Marinus J. Blok, Marketa Safarikova, Robin de Putter, Mattias Van Heetvelde, Sabine Tejpar, Bruce Poppe, Marta Kalousová, Bettina Blaumeiser, Bram Parton, Jan Kral, Greet Wieme, Kathleen Claes, Michal Vocka, Zdenek Kleibl, Jana Soukupova, Petr Nehasil, Marketa Janatova, Petra Zemankova, Kim De Leeneer, Petra Kleiblova, Karen Geboes, Toon Rosseel, RS: GROW - R4 - Reproductive and Perinatal Medicine, MUMC+: DA KG Polikliniek (9), Klinische Genetica, and MUMC+: DA KG Lab Centraal Lab (9)
- Subjects
Oncology ,medicine.medical_specialty ,Cancer Research ,Family Cancer History ,Cancer-Predisposing Gene ,endocrine system diseases ,Colorectal cancer ,overall survival ,pancreatic ductal adenocarcinoma ,MUTATION PREVALENCE ,germline ,Germline ,Article ,multigene panel testing ,Pancreatic cancer ,Internal medicine ,medicine ,Medicine and Health Sciences ,Family history ,CHEK2 ,RC254-282 ,RISK ,family history ,Science & Technology ,business.industry ,Melanoma ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,ASSOCIATION ,medicine.disease ,BRCA1 ,digestive system diseases ,SURVIVAL ,Human medicine ,business ,Life Sciences & Biomedicine - Abstract
(1) Background: The proportion and spectrum of germline pathogenic variants (PV) associated with an increased risk for pancreatic ductal adenocarcinoma (PDAC) varies among populations. (2) Methods: We analyzed 72 Belgian and 226 Czech PDAC patients by multigene panel testing. The prevalence of pathogenic variants (PV) in relation to personal/family cancer history were evaluated. PDAC risks were calculated using both gnomAD-NFE and population-matched controls. (3) Results: In 35/298 (11.7%) patients a PV in an established PDAC-predisposition gene was found. BRCA1/2 PV conferred a high risk in both populations, ATM and Lynch genes only in the Belgian subgroup. PV in other known PDAC-predisposition genes were rarer. Interestingly, a high frequency of CHEK2 PV was observed in both patient populations. PV in PDAC-predisposition genes were more frequent in patients with (i) multiple primary cancers (12/38, 32%), (ii) relatives with PDAC (15/56, 27%), (iii) relatives with breast/ovarian/colorectal cancer or melanoma (15/86, 17%) but more rare in sporadic PDAC (5/149, 3.4%). PV in homologous recombination genes were associated with improved overall survival (HR = 0.51, 95% CI 0.34–0.77). (4) Conclusions: Our analysis emphasizes the value of multigene panel testing in PDAC patients, especially in individuals with a positive family cancer history, and underlines the importance of population-matched controls for risk assessment.
- Published
- 2021
- Full Text
- View/download PDF
9. Prevention and management of cetuximab-related hypomagnesemia
- Author
-
Sabine Tejpar, Hiroya Taniguchi, Agustín Falco, Erica Velthuis, Christian Valencia Pineda, and Ricard Mesia
- Subjects
Cancer Research ,Oncology - Abstract
321 Background: Cetuximab is an epidermal growth factor receptor monoclonal antibody indicated for the treatment of squamous cell carcinoma of the head and neck and RAS wild-type metastatic colorectal cancer. Hypomagnesemia is a reversible adverse event associated with cetuximab treatment. Here, we provide the key considerations in prevention and management of cetuximab-related hypomagnesemia events in clinical practice. Methods: A PubMed literature search was conducted for articles published between 18 March 2012 and 18 March 2022 using cetuximab and hypomagnesemia as keywords. Of the 75 articles retrieved, 20 were considered relevant and are included here.Articles were excluded if they focused on hypomagnesemia-associated clinical benefits. Data were summarized and grouped into the following categories: risk and risk factors; monitoring; prevention; and management. Results: The relative risk of grade 3/4 hypomagnesemia following cetuximab therapy appears to be approximately 1.5 times higher for colorectal cancer compared with head and neck cancer. Several risk factors have been identified including age, comorbidities, treatment duration, baseline electrolyte levels, concomitant platinum-based chemotherapy, and polypharmacy. Although the frequencies of hypomagnesemia events vary between the studies identified, data from the included meta-analyses suggest that the incidence of grade 3/4 hypomagnesemia is around 4% among patients receiving cetuximab therapy. Early and regular monitoring of magnesium levels is recommended ideally every 4 to 8 weeks, especially in patients at high risk of serious complications, and up to 8 weeks following treatment cessation. Proposed proactive prophylactic interventions include magnesium supplementation for patients with baseline magnesium levels < 1.8 mg/dL, to prevent severe hypomagnesemia. The management approach depends on the degree of hypomagnesemia, the symptoms and risk factors, and may include supplementation with oral magnesium salts and/or parenteral forms; intravenous forms may be preferred due to low bioavailability and gastrointestinal adverse events related to oral supplements. While recommendations for initiating magnesium replacement in grade 1 (> 1.2 mg/dL) and grade 2 (0.91.2 mg/dL) hypomagnesemia events vary, magnesium supplementation should be given to patients with grade 3 (0.70.9 mg/dL) or grade 4 (< 0.7 mg/dL) hypomagnesemia, due to high risk of serious adverse cardiac events. If magnesium levels do not improve or continue to decrease despite supplementation, a pause in cetuximab administration may be considered. Conclusions: Hypomagnesemia is a reversible and manageable cetuximab-related adverse event and should not interfere with treatment outcomes. A periodic check of magnesium levels during treatment with cetuximab is recommended.
- Published
- 2022
10. Consensus molecular subgroups (CMS) of colorectal cancer (CRC) and first-line efficacy of FOLFIRI plus cetuximab or bevacizumab in the FIRE3 (AIO KRK-0306) trial
- Author
-
Sebastian Stintzing, Dominik Paul Modest, Volker Heinemann, T. Kirchner, Alexander Kiani, Andreas Jung, Dan Aderka, Thomas Decker, Frank Kullmann, L. Fischer von Weikersthal, Werner Scheithauer, Pratyaksha Wirapati, Tobias Heintges, Christian A. Lerchenmuller, H. J. Lenz, Daniel Neureiter, Markus Moehler, G. Seipelt, Sabine Tejpar, Christoph Kahl, S-E. Al-Batran, Swantje Held, and Florian Kaiser
- Subjects
Male ,0301 basic medicine ,Oncology ,Colorectal cancer ,DNA Mutational Analysis ,Leucovorin ,Cetuximab ,Kaplan-Meier Estimate ,medicine.disease_cause ,0302 clinical medicine ,Antineoplastic Combined Chemotherapy Protocols ,cetuximab ,education.field_of_study ,CMS ,Hazard ratio ,Hematology ,Middle Aged ,Prognosis ,Progression-Free Survival ,030220 oncology & carcinogenesis ,FOLFIRI ,Female ,Fluorouracil ,KRAS ,Colorectal Neoplasms ,medicine.drug ,medicine.medical_specialty ,Bevacizumab ,Colon ,First line ,Clinical Decision-Making ,Population ,colorectal cancer ,bevacizumab ,03 medical and health sciences ,Internal medicine ,Gastrointestinal Tumors ,Biomarkers, Tumor ,medicine ,Humans ,education ,Aged ,business.industry ,Gene Expression Profiling ,Rectum ,Original Articles ,medicine.disease ,030104 developmental biology ,Mutation ,Camptothecin ,business - Abstract
BACKGROUND: FIRE-3 compared first-line therapy with FOLFIRI plus either cetuximab or bevacizumab in 592 KRAS exon 2 wild-type metastatic colorectal cancer (mCRC) patients. The consensus molecular subgroups (CMS) are grouping CRC samples according to their gene-signature in four different subtypes. Relevance of CMS for the treatment of mCRC has yet to be defined. PATIENTS AND METHODS: In this exploratory analysis, patients were grouped according to the previously published tumor CRC-CMSs. Objective response rates (ORR) were compared using chi-square test. Overall survival (OS) and progression-free survival (PFS) times were compared using Kaplan-Meier estimation, log-rank tests. Hazard ratios (HR) were estimated according to the Cox proportional hazard method. RESULTS: CMS classification could be determined in 438 out of 514 specimens available from the intent-to-treat (ITT) population (n = 592). Frequencies for the remaining 438 samples were as follows: CMS1 (14%), CMS2 (37%), CMS3 (15%), CMS4 (34%). For the 315 RAS wild-type tumors, frequencies were as follows: CMS1 (12%), CMS2 (41%), CMS3 (11%), CMS4 (34%). CMS distribution in right- versus (vs) left-sided primary tumors was as follows: CMS1 (27% versus 11%), CMS2 (28% versus 45%), CMS3 (10% versus 12%), CMS4 (35% versus 32%). Independent of the treatment, CMS was a strong prognostic factor for ORR (P = 0.051), PFS (P
- Published
- 2019
11. Addressing the dichotomy between individual and societal approaches to personalised medicine in oncology
- Author
-
Roberto Salgado, David B. Solit, David L. Rimm, Jan Bogaerts, Renzo Canetta, Tracy Lively, Kim Lyerly, Paul N. Span, Alison Bateman-House, Amr Makady, L. Bergmann, Sumimasa Nagai, Chris Smith, Mark Robson, Mary Savage, Emile Voest, Christopher Sweeney, Philippe Lambin, Marlene Thomas, Lyndsay Harris, Denis Lacombe, Chistophe Massard, Rene Bernards, Richard Sullivan, Sabine Tejpar, Nina Lukinova, Herbert K. Lyerly, Helen Moore, Malcolm A. Smith, Laura Yee, Ray DuBois, William C. Hahn, Pasi Janne, Cheryl L. Willman, David Rimm, Lothar Bergmann, Ian A. Cree, Priti Hegde, Shirley Hopper, Marc Robson, Magnus Ingelman-Sundberg, Gwen Nichols, Francois Maignen, Benjamin Besse, Rafal Swierzewski, Astrid Kiermaier, Christophe Massard, Michael Caliguri, Victor Velculescu, Paolo Foggi, Willem C. Hahn, Vassilis Golfinopoulos, Precision Medicine, and RS: GROW - R3 - Innovative Cancer Diagnostics & Therapy
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Emerging technologies ,media_common.quotation_subject ,Cancer drugs ,Psychological intervention ,Medical Oncology ,Societal level ,GUIDELINES ,03 medical and health sciences ,0302 clinical medicine ,Clinical trials ,Internal medicine ,medicine ,Humans ,Profiling (information science) ,Precision Medicine ,Health technology assessment ,Evidence-driven optimal health-care delivery ,media_common ,Women's cancers Radboud Institute for Molecular Life Sciences [Radboudumc 17] ,Clinical trial ,030104 developmental biology ,Clinical research ,030220 oncology & carcinogenesis ,Molecular and immunologic profiling ,Psychology ,ACCESS ,Autonomy ,Biomarkers - Abstract
Academic, industry, regulatory leaders and patient advocates in cancer clinical research met in November 2018 at the Innovation and Biomarkers in Cancer Drug Development meeting in Brussels to address the existing dichotomy between increasing calls for personalised oncology approaches based on individual molecular profiles and the need to make resource and regulatory decisions at the societal level in differing health-care delivery systems around the globe. Novel clinical trial designs, the utility and limitations of real-world evidence (RWE) and emerging technologies for profiling patient tumours and tumour-derived DNA in plasma were discussed. While randomised clinical trials remain the gold standard approach to defining clinical utility of local and systemic therapeutic interventions, the broader adoption of comprehensive tumour profiling and novel trial designs coupled with RWE may allow patient and physician autonomy to be appropriately balanced with broader assessments of safety and overall societal benefit. (C) 2019 Published by Elsevier Ltd.
- Published
- 2019
12. Cetuximab Alone or With Irinotecan for Resistant KRAS-, NRAS-, BRAF- and PIK3CA-wild-type Metastatic Colorectal Cancer: The AGITG Randomized Phase II ICECREAM Study
- Author
-
Niall C. Tebbutt, Val Gebski, Louise M. Nott, Timothy J. Price, Jayesh Desai, Sabine Tejpar, Subotheni Thavaneswaran, Kristy P. Robledo, John Simes, Lorraine A. Chantrill, Fiona Day, Jeremy Shapiro, Paul Waring, Guy van Hazel, Mustafa Khasraw, Eva Segelov, Christos S. Karapetis, Michael Jefford, Craig Underhill, and Nick Pavlakis
- Subjects
Adult ,Male ,Proto-Oncogene Proteins B-raf ,Oncology ,medicine.medical_specialty ,Class I Phosphatidylinositol 3-Kinases ,Colorectal cancer ,Cetuximab ,Irinotecan ,medicine.disease_cause ,GTP Phosphohydrolases ,Proto-Oncogene Proteins p21(ras) ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Biomarkers, Tumor ,medicine ,Humans ,Panitumumab ,030212 general & internal medicine ,neoplasms ,Survival rate ,Aged ,business.industry ,Liver Neoplasms ,Gastroenterology ,Membrane Proteins ,Cancer ,Middle Aged ,Prognosis ,medicine.disease ,digestive system diseases ,Survival Rate ,Fluorouracil ,030220 oncology & carcinogenesis ,Mutation ,Female ,KRAS ,Colorectal Neoplasms ,business ,Follow-Up Studies ,medicine.drug - Abstract
Background The Irinotecan Cetuximab Evaluation and Cetuximab Response Evaluation (ICECREAM) study assessed the efficacy of cetuximab monotherapy compared with cetuximab combined with chemotherapy for quadruple wild-type (KRAS, NRAS, BRAF, or P13KCA exon 20) metastatic colorectal cancer. Patients and Methods Patients were enrolled in an open-label, multicenter, phase II trial and randomly assigned to cetuximab 400 mg/m2, then 250 mg/m2 cetuximab weekly, with or without irinotecan 180 mg/m2 every 2 weeks. The primary endpoint was 6-month progression-free survival; secondary endpoints were response rate, overall survival, toxicity, and quality of life. Results From 2012 to 2016, 48 patients were recruited. Two were ineligible, and 2 were not evaluable for response. Characteristics were balanced, except gender (male, 62% vs. 72%) and primary sidedness (left, 95% vs. 68%). For cetuximab compared with cetuximab-irinotecan, progression-free survival was 14% versus 41% (hazard ratio, 0.39; 95% confidence interval, 0.20-0.78; P = .008); response rate was 10% (2 partial responses) versus 38% (1 complete, 8 partial); P = .04. Grade 3 to 4 toxicities were less with cetuximab monotherapy (23% vs. 50%); global and specific quality of life scores did not differ. Conclusion In comparison with cetuximab alone, cetuximab plus irinotecan increases the response rate and delays progression in irinotecan-resistant RAS wild-type colorectal cancer. This echoes data from molecularly unselected patients.
- Published
- 2018
13. A fatal wound complication following sequential anti-angiogenesis, immune checkpoint inhibition and ultra-hypofractionated radiotherapy
- Author
-
Karin Haustermans, Miha Orazem, Eric Van Cutsem, Cédric Draulans, Marina Debecker, Gert De Meerleer, Mathieu Spaas, Sabine Tejpar, and Jeroen Dekervel
- Subjects
Oncology ,medicine.medical_specialty ,Chemotherapy ,business.industry ,medicine.medical_treatment ,Gastroenterology ,General Medicine ,Immunotherapy ,Combined Modality Therapy ,Immune checkpoint ,Targeted therapy ,Clinical trial ,Radiation therapy ,Surgical oncology ,Internal medicine ,medicine ,Humans ,business ,Immune Checkpoint Inhibitors - Abstract
Multimodality treatments combining radiotherapy, immune therapy and/or targeted therapy are under heavy investigation. Promising data from clinical trials are emerging, nevertheless unexpected interactions and adverse events should not be overlooked. Here we present a case study of a patient with metastatic colon adenocarcinoma treated sequentially with a chemotherapy/targeted therapy combination, immune checkpoint inhibitors and ultra-hypofractionated radiotherapy. After radiation treatment, the patient developed extensive posterior abdominal wall wounds coinciding with regression of the irradiated metastatic tumour mass and marked elevation of the inflammation parameters. This case represents an unusual fatal wound complication after palliative ultra-hypofractionated radiotherapy. Further research into synergistic effects of sequential radiotherapy and anti-angiogenesis therapy may provide an advantage in anticipating severe sequelae.
- Published
- 2021
14. Copy number and transcriptome alterations associated with metastatic lesion response to treatment in colorectal cancer
- Author
-
Mathilde Couetoux du Tertre, Eve St-Hilaire, Archana Srivastava, Steven Hébert, Lucas Sideris, Sabine Tejpar, Bernard Lespérance, Petr Kavan, Claudia L. Kleinman, Yoo-Joung Ko, Richard Dalfen, Karen Gambaro, Adrian Gologan, Gerald Batist, Maud Marques, Celia M. T. Greenwood, André Constantin, Mohammed Harb, Ronald Burkes, Benoit Samson, Suzan McNamara, Vincent Pelsser, Errol Camlioglu, Cyrla Hoffert, Zuanel Diaz, and Félix Couture
- Subjects
0301 basic medicine ,Oncology ,Male ,Candidate gene ,Medicine (General) ,Colorectal cancer ,Medicine (miscellaneous) ,Research & Experimental Medicine ,Metastasis ,Transcriptome ,0302 clinical medicine ,Medicine ,Exome sequencing ,Research Articles ,Cause of death ,Aged, 80 and over ,Liver Neoplasms ,Middle Aged ,Progression-Free Survival ,Bevacizumab ,Gene Expression Regulation, Neoplastic ,Exact test ,Medicine, Research & Experimental ,030220 oncology & carcinogenesis ,Molecular Medicine ,Female ,medicine.symptom ,Colorectal Neoplasms ,Life Sciences & Biomedicine ,Research Article ,Adult ,medicine.medical_specialty ,DNA Copy Number Variations ,Antineoplastic Agents ,colorectal cancer ,Lesion ,03 medical and health sciences ,R5-920 ,Internal medicine ,Exome Sequencing ,Humans ,metastasis ,Aged ,Science & Technology ,business.industry ,copy number aberrations ,treatment response ,medicine.disease ,030104 developmental biology ,Drug Resistance, Neoplasm ,business - Abstract
Background Therapeutic resistance is the main cause of death in metastatic colorectal cancer. To investigate genomic plasticity, most specifically of metastatic lesions, associated with response to first‐line systemic therapy, we collected longitudinal liver metastatic samples and characterized the copy number aberration (CNA) landscape and its effect on the transcriptome. Methods Liver metastatic biopsies were collected prior to treatment (pre, n = 97) and when clinical imaging demonstrated therapeutic resistance (post, n = 43). CNAs were inferred from whole exome sequencing and were correlated with both the status of the lesion and overall patient progression‐free survival (PFS). We used RNA sequencing data from the same sample set to validate aberrations as well as independent datasets to prioritize candidate genes. Results We identified a significantly increased frequency gain of a unique CN, in liver metastatic lesions after first‐line treatment, on chr18p11.32 harboring 10 genes, including TYMS, which has not been reported in primary tumors (GISTIC method and test of equal proportions, FDR‐adjusted p = 0.0023). CNA lesion profiles exhibiting different treatment responses were compared and we detected focal genomic divergences in post‐treatment resistant lesions but not in responder lesions (two‐tailed Fisher's Exact test, unadjusted p ≤ 0.005). The importance of examining metastatic lesions is highlighted by the fact that 15 out of 18 independently validated CNA regions found to be associated with PFS in this study were only identified in the metastatic lesions and not in the primary tumors. Conclusion This investigation of genomic‐phenotype associations in a large colorectal cancer liver metastases cohort identified novel molecular features associated with treatment response, supporting the clinical importance of collecting metastatic samples in a defined clinical setting., Graphical Abstract and Graphical Headlights Liver metastatic lesions from colorectal cancer patients were collected before and after first‐line standard chemotherapy and comprehensively profiled with the objective to assess the genomic impact of systemic therapy and investigate association with response and survival. This study allowed identification of novel CNAs having an impact on the transcriptome with a potential prognostic value in patients with colorectal cancer.
- Published
- 2021
15. 477P Utilisation and predictors of genomic testing prior to first-line (1L) therapy in patients (pts) with metastatic colorectal cancer (mCRC)
- Author
-
Sebastian Stintzing, Chiara Cremolini, Takayuki Yoshino, Janick Weberpals, I. Reyes-Rivera, B. Leutgeb, Heinz-Josef Lenz, H. Nimeiri, J. Seligmann, J. Tabernero, Sami Mahrus, Sabine Tejpar, and Axel Grothey
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Colorectal cancer ,First line ,Internal medicine ,medicine ,In patient ,Hematology ,Personalized medicine ,business ,medicine.disease - Published
- 2021
16. Peripherally-driven myeloid NFkB and IFN/ISG responses predict malignancy risk, survival, and immunotherapy regime in ovarian cancer
- Author
-
Louis Boon, Isaure Vanmeerbeek, Valentina Chiappa, Tom Bourne, Thaïs Baert, Caroline Van Holsbeke, Ignace Vergote, Patrizia Agostinis, Jenny Sprooten, Abhishek D. Garg, Dirk Timmerman, Chiara Landolfo, Ann Vankerckhoven, Raquel S Laureano, Yani Berckmans, Wouter Froyman, Antonia Carla Testa, Sabine Tejpar, Dominique Schols, Daniela Fischerova, Daniel M Borras, Roxanne Wouters, and An Coosemans
- Subjects
Cancer Research ,Myeloid ,medicine.medical_treatment ,SERUM ,Mice ,computational biology ,Immunotherapy Biomarkers ,Immunology and Allergy ,RC254-282 ,Ovarian Neoplasms ,biology ,NF-kappa B ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,TARGET ,medicine.anatomical_structure ,Oncology ,Molecular Medicine ,Biomarker (medicine) ,Female ,HEALTH ,immunotherapy ,Immunotherapy ,Antibody ,Life Sciences & Biomedicine ,immunological techniques ,BIOMARKERS ,BENIGN ,Immunology ,DIAGNOSIS ,Immune system ,INFLAMMATION ,medicine ,tumor microenvironment ,Animals ,Humans ,Pharmacology ,Tumor microenvironment ,Science & Technology ,business.industry ,Cancer ,medicine.disease ,Survival Analysis ,Settore MED/40 - GINECOLOGIA E OSTETRICIA ,tumor biomarkers ,CELLS ,Cancer research ,biology.protein ,Ovarian cancer ,business - Abstract
BackgroundTumors can influence peripheral immune macroenvironment, thereby creating opportunities for non-invasive serum/plasma immunobiomarkers for immunostratification and immunotherapy designing. However, current approaches for immunobiomarkers’ detection are largely quantitative, which is unreliable for assessing functional peripheral immunodynamics of patients with cancer. Hence, we aimed to design a functional biomarker modality for capturing peripheral immune signaling in patients with cancer for reliable immunostratification.MethodsWe used a data-driven in silico framework, integrating existing tumor/blood bulk-RNAseq or single-cell (sc)RNAseq datasets of patients with cancer, to inform the design of an innovative serum-screening modality, that is, serum-functional immunodynamic status (sFIS) assay. Next, we pursued proof-of-concept analyses via multiparametric serum profiling of patients with ovarian cancer (OV) with sFIS assay combined with Luminex (cytokines/soluble immune checkpoints), CA125-antigen detection, and whole-blood immune cell counts. Here, sFIS assay’s ability to determine survival benefit or malignancy risk was validated in a discovery (n=32) and/or validation (n=699) patient cohorts. Lastly, we used an orthotopic murine metastatic OV model, with anti-OV therapy selection via in silico drug–target screening and murine serum screening via sFIS assay, to assess suitable in vivo immunotherapy options.ResultsIn silico data-driven framework predicted that peripheral immunodynamics of patients with cancer might be best captured via analyzing myeloid nuclear factor kappa-light-chain enhancer of activated B cells (NFκB) signaling and interferon-stimulated genes' (ISG) responses. This helped in conceptualization of an ‘in sitro’ (in vitro+in situ) sFIS assay, where human myeloid cells were exposed to patients’ serum in vitro, to assess serum-induced (si)-NFκB or interferon (IFN)/ISG responses (as active signaling reporter activity) within them, thereby ‘mimicking’ patients’ in situ immunodynamic status. Multiparametric serum profiling of patients with OV established that sFIS assay can: decode peripheral immunology (by indicating higher enrichment of si-NFκB over si-IFN/ISG responses), estimate survival trends (si-NFκB or si-IFN/ISG responses associating with negative or positive prognosis, respectively), and coestimate malignancy risk (relative to benign/borderline ovarian lesions). Biologically, we documented dominance of pro-tumorigenic, myeloid si-NFκB responseHIGHsi-IFN/ISG responseLOW inflammation in periphery of patients with OV. Finally, in an orthotopic murine metastatic OV model, sFIS assay predicted the higher capacity of chemo-immunotherapy (paclitaxel–carboplatin plus anti-TNF antibody combination) in achieving a pro-immunogenic peripheral milieu (si-IFN/ISG responseHIGHsi-NFκB responseLOW), which aligned with high antitumor efficacy.ConclusionsWe established sFIS assay as a novel biomarker resource for serum screening in patients with OV to evaluate peripheral immunodynamics, patient survival trends and malignancy risk, and to design preclinical chemo-immunotherapy strategies.
- Published
- 2021
17. Monocyte-driven atypical cytokine storm and aberrant neutrophil activation as key mediators of COVID-19 disease severity
- Author
-
Patrick Matthys, W. De Wever, Lore Vanderbeke, Paul Proost, Pierre Van Mol, Junbin Qian, M. Casaer, Dena Panovska, Kimberly Martinod, Karin Thevissen, J. Filtjens, Ingrid Arijs, T. Van Buyten, Jenny Sprooten, R. Vos, A. Emmaneel, J. Vandenhaute, Bram Boeckx, Asier Antoranz, J. Odent, L. C. Velásquez Pereira, Yvan Saeys, A. Wilmer, N. Lorent, Diether Lambrechts, Sander Jansen, Jeroen Raes, Abhishek D. Garg, Joost Wauters, Adrian Liston, Erwin Dreesen, Francesca Maria Bosisio, Dieter Dauwe, S. Van Gassen, Birgit Weynand, Els Wauters, K. Quintelier, Dries Testelmans, M. Gouwy, Greet Hermans, Philippe Meersseman, Christophe Dooms, Toine Mercier, Johan Neyts, Carine Wouters, Katrien Lagrou, Sabine Tejpar, Stephanie Humblet-Baron, P. A. Penttila, Jonas Yserbyt, S. Rex, E. Pollet, Jan Gunst, F. De Smet, Y. Van Herck, Vanderbeke, L [0000-0002-3364-1213], Van Herck, Y [0000-0003-4604-8010], De Smet, F [0000-0002-6669-3335], Martinod, K [0000-0002-1026-6107], Arijs, I [0000-0002-9415-083X], Dauwe, D [0000-0002-9771-2543], Dreesen, E [0000-0002-0785-2930], Emmaneel, A [0000-0003-4569-2330], Gunst, J [0000-0003-2470-6393], Jansen, S [0000-0002-9344-131X], Liston, A [0000-0002-6272-4085], Mercier, T [0000-0002-1517-6426], Neyts, J [0000-0002-0033-7514], Panovska, D [0000-0003-0906-3860], Penttila, PA [0000-0002-5861-4430], Proost, P [0000-0002-0133-5545], Qian, J [0000-0001-6145-045X], Quintelier, K [0000-0001-5306-5615], Raes, J [0000-0002-1337-041X], Saeys, Y [0000-0002-0415-1506], Thevissen, K [0000-0003-0275-9072], Vandenhaute, J [0000-0001-9485-8806], Van Gassen, S [0000-0002-7119-5330], Vos, R [0000-0002-3468-9251], Garg, AD [0000-0002-9976-9922], Matthys, P [0000-0002-9685-6836], Lambrechts, D [0000-0002-3429-302X], Wauters, E [0000-0002-0115-0030], and Apollo - University of Cambridge Repository
- Subjects
Oncology ,Male ,0301 basic medicine ,animal diseases ,medicine.medical_treatment ,General Physics and Astronomy ,Disease ,Extracellular Traps ,Severity of Illness Index ,Monocytes ,Neutrophil Activation ,0302 clinical medicine ,Informed consent ,Immunopathology ,Medicine and Health Sciences ,Medicine ,health care economics and organizations ,Multidisciplinary ,Middle Aged ,Acquired immune system ,Cytokine release syndrome ,medicine.anatomical_structure ,Cytokine ,030220 oncology & carcinogenesis ,Cytokines ,Female ,Infection ,Cytokine Release Syndrome ,medicine.medical_specialty ,Coronavirus disease 2019 (COVID-19) ,Science ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,education ,Antigen-Presenting Cells ,Article ,General Biochemistry, Genetics and Molecular Biology ,Immunophenotyping ,03 medical and health sciences ,Immune system ,Disease severity ,Antigen ,Internal medicine ,Humans ,Aged ,SARS-CoV-2 ,business.industry ,Monocyte ,Histocompatibility Antigens Class II ,COVID-19 ,Biology and Life Sciences ,General Chemistry ,Neutrophil extracellular traps ,medicine.disease ,030104 developmental biology ,Viral infection ,Case-Control Studies ,Immunology ,CELLS ,business ,Cytokine storm - Abstract
Epidemiological and clinical reports indicate that SARS-CoV-2 virulence hinges upon the triggering of an aberrant host immune response, more so than on direct virus-induced cellular damage. To elucidate the immunopathology underlying COVID-19 severity, we perform cytokine and multiplex immune profiling in COVID-19 patients. We show that hypercytokinemia in COVID-19 differs from the interferon-gamma-driven cytokine storm in macrophage activation syndrome, and is more pronounced in critical versus mild-moderate COVID-19. Systems modelling of cytokine levels paired with deep-immune profiling shows that classical monocytes drive this hyper-inflammatory phenotype and that a reduction in T-lymphocytes correlates with disease severity, with CD8+ cells being disproportionately affected. Antigen presenting machinery expression is also reduced in critical disease. Furthermore, we report that neutrophils contribute to disease severity and local tissue damage by amplification of hypercytokinemia and the formation of neutrophil extracellular traps. Together our findings suggest a myeloid-driven immunopathology, in which hyperactivated neutrophils and an ineffective adaptive immune system act as mediators of COVID-19 disease severity., The host immune response plays a critical role in the immunopathology of SARS-CoV2. Here the authors combine a systems biology approach to implicate monocytes as key drivers of cytokine storm and disturbed neutrophil activation in COVID-19 disease severity.
- Published
- 2021
18. Radiomics Response Signature for Identification of Metastatic Colorectal Cancer Sensitive to Therapies Targeting EGFR Pathway
- Author
-
Lawrence H. Schwartz, Hubert Piessevaux, Min Qian, Laurent Dercle, Lin Lu, Sabine Tejpar, Binsheng Zhao, Peter Eggleton, UCL - SSS/IREC/GAEN - Pôle d'Hépato-gastro-entérologie, and UCL - (SLuc) Service de gastro-entérologie
- Subjects
Oncology ,Male ,Cancer Research ,Artificial intelligence ,Colorectal cancer ,Leucovorin ,Cetuximab ,Biomarkers, Pharmacological ,Cohort Studies ,0302 clinical medicine ,Antineoplastic Combined Chemotherapy Protocols ,Image Processing, Computer-Assisted ,Molecular Targeted Therapy ,Neoplasm Metastasis ,0303 health sciences ,Hazard ratio ,Liver Neoplasms ,machine-learning ,ErbB Receptors ,Gene Expression Regulation, Neoplastic ,Response Evaluation Criteria in Solid Tumors ,radiomics ,030220 oncology & carcinogenesis ,FOLFIRI ,Female ,Fluorouracil ,Colorectal Neoplasms ,medicine.drug ,Signal Transduction ,medicine.medical_specialty ,Irinotecan ,deep-learning ,03 medical and health sciences ,Internal medicine ,medicine ,Biomarkers, Tumor ,Humans ,030304 developmental biology ,Retrospective Studies ,Colorectal Cancer ,Receiver operating characteristic ,Proportional hazards model ,business.industry ,Editorials ,medicine.disease ,Drug Resistance, Neoplasm ,business ,Tomography, X-Ray Computed ,Transcriptome ,RAS - Abstract
Background The authors sought to forecast survival and enhance treatment decisions for patients with liver metastatic colorectal cancer by using on-treatment radiomics signature to predict tumor sensitiveness to irinotecan, 5-fluorouracil, and leucovorin (FOLFIRI) alone (F) or in combination with cetuximab (FC). Methods We retrospectively analyzed 667 metastatic colorectal cancer patients treated with F or FC. Computed tomography quality was classified as high (HQ) or standard (SD). Four datasets were created using the nomenclature (treatment) – (quality). Patients were randomly assigned (2:1) to training or validation sets: FCHQ: 78:38, FCSD: 124:62, FHQ: 78:51, FSD: 158:78. Four tumor-imaging biomarkers measured quantitative radiomics changes between standard of care computed tomography scans at baseline and 8 weeks. Using machine learning, the performance of the signature to classify tumors as treatment sensitive or treatment insensitive was trained and validated using receiver operating characteristic (ROC) curves. Hazard ratio and Cox regression models evaluated association with overall survival (OS). Results The signature (area under the ROC curve [95% confidence interval (CI)]) used temporal decrease in tumor spatial heterogeneity plus boundary infiltration to successfully predict sensitivity to antiepidermal growth factor receptor therapy (FCHQ: 0.80 [95% CI = 0.69 to 0.94], FCSD: 0.72 [95% CI = 0.59 to 0.83]) but failed with chemotherapy (FHQ: 0.59 [95% CI = 0.44 to 0.72], FSD: 0.55 [95% CI = 0.43 to 0.66]). In cetuximab-containing sets, radiomics signature outperformed existing biomarkers (KRAS-mutational status, and tumor shrinkage by RECIST 1.1) for detection of treatment sensitivity and was strongly associated with OS (two-sided P Conclusions Radiomics response signature can serve as an intermediate surrogate marker of OS. The signature outperformed known biomarkers in providing an early prediction of treatment sensitivity and could be used to guide cetuximab treatment continuation decisions.
- Published
- 2020
19. Association of Consensus Molecular Subtypes and Molecular Markers With Clinical Outcomes in Patients With Metastatic Colorectal Cancer: Biomarker Analyses From LUME-Colon 1
- Author
-
Ogsen Gabrielyan, Heinz-Josef Lenz, Eric Van Cutsem, Alexey V. Salnikov, Ramona Schmid, T. Kitzing, Fortunato Ciardiello, Jürgen Braunger, Sabine Tejpar, Takayuki Yoshino, Guillem Argiles, Josef Höfler, Lenz, H. -J., Argiles, G., Yoshino, T., Tejpar, S., Ciardiello, F., Braunger, J., Salnikov, A. V., Gabrielyan, O., Schmid, R., Hofler, J., Kitzing, T., and Van Cutsem, E.
- Subjects
Oncology ,Male ,Proteomics ,Indoles ,Circulating biomarkers ,Colorectal cancer ,Predictive marker ,medicine.disease_cause ,Transcriptome ,chemistry.chemical_compound ,Prognostic marker ,0302 clinical medicine ,Tumor Microenvironment ,Medicine ,Randomized Controlled Trials as Topic ,Circulating biomarker ,Aged, 80 and over ,Gastroenterology ,Middle Aged ,Prognosis ,Progression-Free Survival ,030220 oncology & carcinogenesis ,Biomarker (medicine) ,030211 gastroenterology & hepatology ,Nintedanib ,Female ,Microsatellite Instability ,KRAS ,Colorectal Neoplasms ,Adult ,medicine.medical_specialty ,Concordance ,Risk Assessment ,03 medical and health sciences ,Genetic Heterogeneity ,Young Adult ,Internal medicine ,Biomarkers, Tumor ,Humans ,Genomic biomarker ,Aged ,Genomic biomarkers ,business.industry ,Gene Expression Profiling ,medicine.disease ,Clinical trial ,chemistry ,Clinical Trials, Phase III as Topic ,Mutation ,Feasibility Studies ,business - Abstract
INTRODUCTION: LUME-Colon 1 (NCT02149108) was a global, placebo-controlled phase III study of nintedanib in advanced colorectal cancer (CRC). Pre-specified biomarker analyses investigated the association of CRC consensus molecular subtypes (CMS) and tumor genomic and circulating biomarkers with clinical outcomes. MATERIALS AND METHODS: Archival tumor tissue, cell-free DNA (cfDNA), and plasma samples were collected for genomic, transcriptomic, and proteomic analyses to investigate potential associations between CRC CMS and other biomarkers with nintedanib response and clinical outcomes. RESULTS: Of the 765 treated patients, 735, 245, and 192 patient samples were analyzed in the circulating protein, tumor tissue, and cfDNA datasets, respectively. Patients were classified as CMS1 (1.7%), CMS2 (27.7%), CMS3 (0.9%), CMS4 (51.5%), or unclassified (18.2%). Unclassified/mixed CMS was associated with longer overall survival (OS) with nintedanib vs. CMS2 or CMS4 (interaction P-value = .0086); no association was observed for CMS4. Gene expression-based pathway analysis revealed an association between vascular endothelial growth factor-related signaling and OS for nintedanib (P = .0498). The most frequently detected somatic mutations were APC (72.0% [tumor tissue] vs. 56.8% [cfDNA]), TP53 (47.1% vs. 34.9%), KRAS (40.8% vs. 28.6%), and PIK3CA (16.6% vs. 11.5%); concordance rates were > 80%. Median OS differences were observed for APC and TP53 mutations vs. wild-type in cfDNA, indicating a potential prognostic value. Circulating ANG-2, CA-9, CEACAM1, collagen-IV, IGFBP-1, ICAM-1, IL-8, and uPAR were potentially prognostic for both OS and progression-free survival. CONCLUSION: We demonstrated the feasibility of large-scale biomarker analyses and CMS classification within a global clinical trial, and identified signals suggesting a potential for greater nintedanib treatment response in the unclassified/mixed CMS subgroup, despite these tumors showing heterogeneous patterns of CMS mixtures. Our results revealed a high degree of concordance in somatic mutations between tumor tissue and cfDNA. Associations with prognosis for cfDNA somatic mutations, as well as several protein-based biomarkers, may warrant further investigation in future trials. ispartof: CLINICAL COLORECTAL CANCER vol:20 issue:1 pages:84-+ ispartof: location:United States status: published
- Published
- 2020
20. Trial watch: chemotherapy-induced immunogenic cell death in immuno-oncology
- Author
-
Jitka Fucikova, Sabine Tejpar, Lorenzo Galluzzi, Peter Vandenberghe, Laurence Zitvogel, Isaure Vanmeerbeek, Abhishek D. Garg, Guido Kroemer, Dirk De Ruysscher, Radek Spisek, and Jenny Sprooten
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Immunogenic Cell Death ,chemokines ,Review ,Adaptive Immunity ,cytotoxic T lymphocyte ,0302 clinical medicine ,Neoplasms ,ANTITUMOR IMMUNITY ,Immunology and Allergy ,RC254-282 ,CAR T cells ,CHECKPOINT BLOCKADE ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Acquired immune system ,SOFT-TISSUE SARCOMA ,Oncology ,030220 oncology & carcinogenesis ,endoplasmic reticulum stress ,type I interferon ,Immunogenic cell death ,Immunotherapy ,BREAST-CANCER PATIENTS ,Central tolerance ,Life Sciences & Biomedicine ,autophagy ,immune checkpoint blocker ,dendritic cell ,Immunology ,Antineoplastic Agents ,CD8(+) T-CELLS ,DENDRITIC CELLS ,03 medical and health sciences ,Immune system ,Antigen ,Adjuvanticity ,FIND-ME SIGNAL ,medicine ,APOPTOTIC CELLS ,Humans ,Antigen-presenting cell ,CALRETICULIN EXPOSURE ,Science & Technology ,business.industry ,RC581-607 ,RANDOMIZED PHASE-2 TRIAL ,cytokines ,030104 developmental biology ,Immunologic diseases. Allergy ,business - Abstract
The term 'immunogenic cell death' (ICD) denotes an immunologically unique type of regulated cell death that enables, rather than suppresses, T cell-driven immune responses that are specific for antigens derived from the dying cells. The ability of ICD to elicit adaptive immunity heavily relies on the immunogenicity of dying cells, implying that such cells must encode and present antigens not covered by central tolerance (antigenicity), and deliver immunostimulatory molecules such as damage-associated molecular patterns and cytokines (adjuvanticity). Moreover, the host immune system must be equipped to detect the antigenicity and adjuvanticity of dying cells. As cancer (but not normal) cells express several antigens not covered by central tolerance, they can be driven into ICD by some therapeutic agents, including (but not limited to) chemotherapeutics of the anthracycline family, oxaliplatin and bortezomib, as well as radiation therapy. In this Trial Watch, we describe current trends in the preclinical and clinical development of ICD-eliciting chemotherapy as partner for immunotherapy, with a focus on trials assessing efficacy in the context of immunomonitoring. ispartof: ONCOIMMUNOLOGY vol:9 issue:1 ispartof: location:United States status: published
- Published
- 2020
21. The prognostic impact of consensus molecular subtypes (CMS) and its predictive effects for bevacizumab benefit in metastatic colorectal cancer: molecular analysis of the AGITG MAX clinical trial
- Author
-
Niall C. Tebbutt, John M. Mariadason, David S. Williams, Timothy J. Price, Daniel D. Buchanan, Jennifer Mooi, Chee Khoon Lee, Amanda R. Townsend, Jennifer E. Hardingham, Pratyaksha Wirapati, Rebecca Asher, Sabine Tejpar, and P S Savas
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Bevacizumab ,Colorectal cancer ,Mitomycin ,Population ,Kaplan-Meier Estimate ,Capecitabine ,03 medical and health sciences ,0302 clinical medicine ,health services administration ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Progression-free survival ,education ,health care economics and organizations ,Aged ,Aged, 80 and over ,education.field_of_study ,Proportional hazards model ,business.industry ,Gene Expression Profiling ,Hazard ratio ,Hematology ,Middle Aged ,Prognosis ,medicine.disease ,Chemotherapy regimen ,Progression-Free Survival ,030104 developmental biology ,030220 oncology & carcinogenesis ,Female ,Colorectal Neoplasms ,Transcriptome ,business ,medicine.drug - Abstract
Background: The consensus molecular subtypes (CMS) is a transcriptome-based classification of colorectal cancer (CRC) initially described in early-stage cohorts, but the associations of CMS with treatment outcomes in the metastatic setting are yet to be established. This study aimed to evaluate the prognostic impact of CMS classification and its predictive effects for bevacizumab benefit in metastatic CRC by correlative analysis of the AGITG MAX trial. Patients and methods: The MAX trial previously reported improved progression-free survival (PFS) for the addition of bevacizumab (B) to chemotherapy [capecitabine (C)±mitomycin (M)]. Archival primary tumours from 237 patients (50% of trial population) underwent gene expression profiling and classification into CMS groups. CMS groups were correlated to PFS and overall survival (OS). The interaction of CMS with treatment was assessed by proportional hazards model. Results: The distribution of CMS in MAX were CMS1 18%, CMS2 47%, CMS3 12%, CMS4 23%. CMS1 was the predominant subtype in right-sided primary tumours, while CMS2 was the predominant subtype in left-sided. CMS was prognostic of OS (P = 0.008), with CMS2 associated with the best outcome and CMS1 the worst. CMS remained an independent prognostic factor in a multivariate analysis. There was a significant interaction between CMS and treatment (P-interaction = 0.03), for PFS, with hazard ratios (95% CI) for CB+CBM versus C arms in CMS1, 2, 3 and 4: 0.83 (0.43-1.62), 0.50 (0.33-0.76), 0.31 (0.13-0.75) and 1.24 (0.68-2.25), respectively. Conclusions: This exploratory study found that CMS stratified OS outcomes in metastatic CRC regardless of first-line treatment, with prognostic effects of CMS groups distinct from those previously reported in early-stage cohorts. In CMS associations with treatment, CMS2 and possibly CMS3 tumours may preferentially benefit from the addition of bevacizumab to first-line capecitabine-based chemotherapy, compared with other CMS groups. Validation of these findings in additional cohorts is warranted. Clinical trial number: This is a molecular sub-study of MAX clinical trial (NCT00294359). ispartof: Ann Oncol vol:29 issue:11 pages:2240-2246 ispartof: location:England status: published
- Published
- 2018
22. A transatlantic perspective on the integration of immuno-oncology prognostic and predictive biomarkers in innovative clinical trial design
- Author
-
Denis Lacombe, Stephen M. Hewitt, Roberto Salgado, S. Litiere, Sabine Tejpar, M. Thurin, B. Conley, T. Lively, Marie Morfouace, Vassilis Golfinopoulos, and Katherine Hartmann
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Medical Oncology ,03 medical and health sciences ,0302 clinical medicine ,Dosing schedules ,Neoplasms ,Internal medicine ,Biomarkers, Tumor ,Animals ,Humans ,Medicine ,Predictive biomarker ,Response rate (survey) ,Clinical Trials as Topic ,business.industry ,Clinical study design ,Cancer ,Prognosis ,medicine.disease ,Immune therapy ,Clinical trial ,030104 developmental biology ,030220 oncology & carcinogenesis ,Potential biomarkers ,business - Abstract
Immuno-therapeutics aim to activate the body's own immune system against cancer and are one of the most promising cancer treatment strategies, but currently limited by a variable response rate. Biomarkers may help to distinguish those patients most likely to respond to therapy; they may also help guide clinical decision making for combination therapies, dosing schedules, and determining progression versus relapse. However, there is a need to confirm such biomarkers in preferably prospective clinical trials before they can be used in practice. Accordingly, it is essential that clinical trials for immuno-therapeutics incorporate biomarkers. Here, focusing on the specific setting of immune therapies, we discuss both the scientific and logistical hurdles to identifying potential biomarkers and testing them in clinical trials.
- Published
- 2018
23. The proprotein convertase furin is a pro-oncogenic driver in KRAS and BRAF driven colorectal cancer
- Author
-
Sandra Meulemans, John W.M. Creemers, Abdel-Majid Khatib, Zongsheng He, Lieven Thorrez, Serge Evrard, Sabine Tejpar, and Geraldine Siegfried
- Subjects
HUMAN COLON ,0301 basic medicine ,Cancer Research ,endocrine system diseases ,Colorectal cancer ,Angiogenesis ,medicine.disease_cause ,ACTIVATION ,Mice ,0302 clinical medicine ,TUMOR PROGRESSION ,Furin ,Genetics & Heredity ,Mice, Knockout ,PROLIFERATION ,Oncology ,030220 oncology & carcinogenesis ,GROWTH ,KRAS ,Signal transduction ,Colorectal Neoplasms ,Life Sciences & Biomedicine ,HT29 Cells ,EXPRESSION ,Proto-Oncogene Proteins B-raf ,Biochemistry & Molecular Biology ,MAP Kinase Signaling System ,Biology ,Proto-Oncogene Proteins p21(ras) ,03 medical and health sciences ,Downregulation and upregulation ,Genetics ,medicine ,Animals ,Humans ,neoplasms ,Molecular Biology ,Science & Technology ,MUTATIONS ,Cell Biology ,Proprotein convertase ,medicine.disease ,digestive system diseases ,030104 developmental biology ,HEK293 Cells ,Gene Expression Regulation ,CELLS ,Mutation ,Cancer research ,biology.protein ,INHIBITORS ,Carcinogenesis ,RESISTANCE - Abstract
Mutations in KRAS and/or BRAF that activate the ERK kinase are frequently found in colorectal cancer (CRC) and drive resistance to targeted therapies. Therefore, the identification of therapeutic targets that affect multiple signaling pathways simultaneously is crucial for improving the treatment of patients with KRAS or BRAF mutations. The proprotein convertase furin activates several oncogenic protein precursors involved in the ERK-MAPK pathway by endoproteolytic cleavage. Here we show that genetic inactivation of furin suppresses tumorigenic growth, proliferation, and migration in KRAS or BRAF mutant CRC cell lines but not in wild-type KRAS and BRAF cells. In a mouse xenograft model, these KRAS or BRAF mutant cells lacking furin displayed reduced growth and angiogenesis, and increased apoptosis. Mechanistically, furin inactivation prevents the processing of various protein pecursors including proIGF1R, proIR, proc-MET, proTGF-β1 and NOTCH1 leading to potent and durable ERK-MAPK pathway suppression in KRAS or BRAF mutant cells. Furthermore, we identified genes involved in activating the ERK-MAPK pathway, such as PTGS2, which are downregulated in the KRAS or BRAF mutant cells after furin inactivation but upregulated in wild-type KRAS and BRAF cells. Analysis of human colorectal tumor samples reveals a positive correlation between enhanced furin expression and KRAS or BRAF expression. These results indicate that furin plays an important role in KRAS or BRAF-associated ERK-MAPK pathway activation and tumorigenesis, providing a potential target for personalized treatment. ispartof: ONCOGENE vol:39 issue:17 pages:3571-3587 ispartof: location:England status: published
- Published
- 2019
24. Bcl-xL as a poor prognostic biomarker and predictor of response to adjuvant chemotherapy specifically in BRAF-mutant stage II and III colon cancer
- Author
-
Helen G. Coleman, Alan Gilmore, Stephen McQuaid, Ronan T. Gray, Federica Di Nicolantonio, Philip D Dunne, Sabine Tejpar, Darragh G. McArt, Matthew Alderdice, Patrick G. Johnston, Daniel B. Longley, Mark Lawler, Victoria Bingham, D. Klingbiel, Peter Bankhead, Jacqueline James, Caitriona Holohan, Amy M.B. McCorry, and Maurice B Loughrey
- Subjects
Oncology ,medicine.medical_specialty ,education.field_of_study ,molecular stratification ,Bcl-xL ,Colorectal cancer ,Proportional hazards model ,business.industry ,Population ,Disease ,relapse risk ,medicine.disease ,Colon cancer ,Gene expression profiling ,Molecular stratification ,Relapse risk ,colon cancer ,Internal medicine ,Genotype ,Gene expression ,Cohort ,medicine ,gene expression profiling ,Immunohistochemistry ,education ,business ,Research Paper - Abstract
BackgroundBRAF mutation occurs in 8-15% of colon cancers (CC), and is associated with poor prognosis in metastatic disease. Compared to wild-type BRAF (BRAFWT) disease, stage II/III CC patients with BRAF mutant (BRAFMT) tumors have shorter overall survival after relapse; however, time-to-relapse is not significantly different. The aim of this investigation was to identify, and validate, novel predictors of relapse of stage II/III BRAFMT CC.Patients and methodsWe used gene expression data from a cohort of 460 patients (GSE39582) to perform a supervised classification analysis based on risk-of-relapse within BRAFMT stage II/III CC, to identify transcriptomic biomarkers associated with prognosis within this genotype. These findings were validated using immunohistochemistry in an independent population-based cohort of Stage II/III CC (n=691), applying Cox proportional hazards analysis to determine associations with survival.ResultsHigh gene expression levels of Bcl-xL, a key regulator of apoptosis, were associated with increased risk of relapse, specifically in BRAFMT tumors (HR=8.3, 95% CI 1.7-41.7), but not KRASMT/BRAFWT or KRASWT/BRAFWT tumors. High Bcl-xL protein expression in BRAFMT, untreated, stage II/III CC was confirmed to be associated with an increased risk of death in an independent cohort (HR=12.13, 95% CI 2.49-59.13). Additionally, BRAFMT tumors with high levels of Bcl-xL protein expression appeared to benefit from adjuvant chemotherapy (P for interaction =0.006), indicating the potential predictive value of Bcl-xL expression in this setting.ConclusionsThese findings provide evidence that Bcl-xL gene and/or protein expression identifies a poor prognostic subgroup of BRAFMT stage II/III CC patients, who may benefit from adjuvant chemotherapy.Key MessageUsing a combination of computational biology discovery and immunohistochemistry validation in independent patient cohorts, we show that high expression of the apoptosis regulator Bcl-xL is associated with disease relapse specifically within BRAF mutant stage II/III colon cancer.This data could enable tailored disease management to reduce relapse rates in the most aggressive subtype.
- Published
- 2018
25. 457P Circulating tumor DNA (ctDNA) from patients (pts) with advanced colorectal cancer (CRC) is enriched for EGFR extracellular domain (ECD) mutations
- Author
-
Geoffrey R. Oxnard, Liangliang Zhang, Halla Nimeiri, Hanna Tukachinsky, Brennan Decker, Sabine Tejpar, Alexa B. Schrock, Jeffrey M. Venstrom, and Dean Pavlick
- Subjects
Advanced colorectal cancer ,Oncology ,Circulating tumor DNA ,business.industry ,Cancer research ,Extracellular ,Medicine ,Hematology ,business ,Domain (software engineering) - Published
- 2021
26. P-45 An open-label, phase 2 study of patritumab deruxtecan in patients with previously treated advanced/metastatic colorectal cancer
- Author
-
Marwan Fakih, Hiroya Taniguchi, Sabine Tejpar, M. Kanai, J. Beck, Katrina S. Pedersen, G. Pudussery, Yali Liu, Zev A. Wainberg, Kanwal Pratap Singh Raghav, Hisato Kawakami, Arndt Vogel, Kensei Yamaguchi, Sabeen Mekan, Takayuki Yoshino, Scott Kopetz, K. Bando, and Yang Qiu
- Subjects
Oncology ,Patritumab ,medicine.medical_specialty ,business.industry ,Colorectal cancer ,Phases of clinical research ,Hematology ,medicine.disease ,Internal medicine ,medicine ,In patient ,Open label ,Previously treated ,business - Published
- 2021
27. Understanding the role of primary tumour localisation in colorectal cancer treatment and outcomes
- Author
-
Peter Gibbs, Sabine Tejpar, Heinz-Josef Lenz, Sebastian Stintzing, and Lars Thiebach
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Colorectal cancer ,Colon ,Rectum ,Antineoplastic Agents ,Predictive ,Prognostic ,Gastroenterology ,Article ,Descending colon ,03 medical and health sciences ,0302 clinical medicine ,Midgut ,Predictive Value of Tests ,Internal medicine ,medicine ,Biomarkers, Tumor ,Ascending colon ,Humans ,Hindgut ,Neoplasm Metastasis ,Precision Medicine ,Protein Kinase Inhibitors ,Splenic flexure ,business.industry ,Rectal Neoplasms ,Transverse colon ,Sigmoid colon ,medicine.disease ,digestive system diseases ,Gastrointestinal Microbiome ,ErbB Receptors ,Colorectal carcinoma ,030104 developmental biology ,medicine.anatomical_structure ,Treatment Outcome ,Molecular Diagnostic Techniques ,030220 oncology & carcinogenesis ,Colonic Neoplasms ,Mutation ,Metastatic ,business ,Colorectal Neoplasms - Abstract
Metastatic colorectal carcinoma (mCRC) is a heterogeneous disease with differing outcomes and clinical responses and poor prognosis. CRCs can be characterised by their primary tumour location within the colon. The left-sided colon, derived from the hindgut, includes the distal third of the transverse colon, splenic flexure, descending colon, sigmoid colon and rectum. The right-sided colon, derived from the midgut, includes the proximal two-thirds of the transverse colon, ascending colon and caecum. Sometimes, the rectum is described separately, despite originating from the hindgut, and in many clinical series, the left-sided colon includes only tumours within and distal to the splenic flexure. Differences in the microbiome, clinical characteristics and chromosomal and molecular characteristics have been reported between the right and left side of the colon, regardless of how this is defined. There is now strong evidence from clinical studies in patients with mCRC for the prognostic effect of primary tumour location. The impact of primary colonic tumour location on response to treatment is now under investigation in a large number of clinical studies in patients with mCRC. In this review, we summarise the microbiome, clinical, chromosomal and molecular differences associated with the primary location of CRC. We present an overview of the proven prognostic impact of primary tumour location for patients with mCRC and discuss emerging data for the predictive impact of primary tumour location on clinical outcome.
- Published
- 2017
28. Consensus molecular subtypes and the evolution of precision medicine in colorectal cancer
- Author
-
Edmund Scott Kopetz, Louis Vermeulen, Sabine Tejpar, Rodrigo Dienstmann, Justin Guinney, and Josep Tabernero
- Subjects
Epigenomics ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Consensus ,Colorectal cancer ,General Mathematics ,Genomics ,Computational biology ,Disease ,Somatic evolution in cancer ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,Precision Medicine ,Gene ,business.industry ,Applied Mathematics ,Cancer ,medicine.disease ,Precision medicine ,3. Good health ,ErbB Receptors ,030104 developmental biology ,Drug development ,030220 oncology & carcinogenesis ,Mutation ,Colorectal Neoplasms ,Transcriptome ,business - Abstract
Critical driver genomic events in colorectal cancer have been shown to affect the response to targeted agents that were initially developed under the 'one gene, one drug' paradigm of precision medicine. Our current knowledge of the complexity of the cancer genome, clonal evolution patterns under treatment pressure and pharmacodynamic effects of target inhibition support the transition from a one gene, one drug approach to a 'multi-gene, multi-drug' model when making therapeutic decisions. Better characterization of the transcriptomic subtypes of colorectal cancer, encompassing tumour, stromal and immune components, has revealed convergent pathway dependencies that mandate a 'multi-molecular' perspective for the development of therapies to treat this disease.
- Published
- 2017
29. Impact of Consensus Molecular Subtype on Survival in Patients With Metastatic Colorectal Cancer: Results From CALGB/SWOG 80405 (Alliance)
- Author
-
Federico Innocenti, Charles D. Blanke, Richard M. Goldberg, Sabine Tejpar, Monica M. Bertagnolli, Donna Niedzwiecki, Omar Kabbarah, Robert J. Mayer, Pratyaksha Wirapati, Xueping Qu, Tyler Zemla, Fang-Shu Ou, Alan P. Venook, Heinz-Josef Lenz, and Howard S. Hochster
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Disease free survival ,Consensus ,Colorectal cancer ,Treatment outcome ,Clinical Decision-Making ,MODELS ,INHIBITION ,CLASSIFICATION ,Decision Support Techniques ,FIRE-3 ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,In patient ,neoplasms ,FOLFIRI PLUS BEVACIZUMAB ,Science & Technology ,Extramural ,business.industry ,ORIGINAL REPORTS ,CHEMOTHERAPY ,medicine.disease ,OPEN-LABEL ,digestive system diseases ,STATISTICS ,CETUXIMAB ,Clinical trial ,Gene expression profiling ,1ST-LINE TREATMENT ,030104 developmental biology ,030220 oncology & carcinogenesis ,Predictive value of tests ,Colonic Neoplasms ,business ,Colorectal Neoplasms ,Life Sciences & Biomedicine - Abstract
PURPOSE To determine the predictive and prognostic value of the consensus molecular subtypes (CMSs) of colorectal cancer (CRC) that represent a merging of gene expression–based features largely in primary tumors from six independent classification systems and provide a framework for capturing the intrinsic heterogeneity of CRC in patients enrolled in CALGB/SWOG 80405. PATIENTS AND METHODS CALGB/SWOG 80405 is a phase III trial that compared the addition of bevacizumab or cetuximab to infusional fluorouracil, leucovorin, and oxaliplatin or fluorouracil, leucovorin, and irinotecan as first-line treatment of advanced CRC. We characterized the CMS classification using a novel NanoString gene expression panel on primary CRCs from 581 patients enrolled in this study to assess the prognostic and predictive value of CMSs in these patients. RESULTS The CMSs are highly prognostic for overall survival (OS; P < .001) and progression-free survival (PFS; P < .001). Furthermore, CMSs were predictive for both OS ( P for interaction < .001) and PFS ( P for interaction = .0032). In the CMS1 cohort, patients treated with bevacizumab had a significantly longer OS than those treated with cetuximab ( P < .001). In the CMS2 cohort, patients treated with cetuximab had a significantly longer OS than patients treated with bevacizumab ( P = .0046). CONCLUSION These findings highlight the possible clinical utility of CMSs and suggests that refinement of the CMS classification may provide a path toward identifying patients with metastatic CRC who are most likely to benefit from specific targeted therapy as part of the initial treatment.
- Published
- 2019
30. The Interleukin 22 Pathway Interacts with Mutant KRAS to Promote Poor Prognosis in Colon Cancer
- Author
-
Enric Domingo, Lucy C. Garner, Matthias Friedrich, David Barras, Nathan R. West, Sabine Tejpar, Mauro Delorenzi, Elizabeth H. Mann, Tim Maughan, Fiona Powrie, Alina Janney, Samuel J. Bullers, Sarah McCuaig, Viktor H. Koelzer, University of Zurich, and Powrie, Fiona
- Subjects
0301 basic medicine ,Male ,Cancer Research ,Colorectal cancer ,Receptor expression ,Population ,610 Medicine & health ,medicine.disease_cause ,Disease-Free Survival ,Transcriptome ,Proto-Oncogene Proteins p21(ras) ,03 medical and health sciences ,Mice ,0302 clinical medicine ,10049 Institute of Pathology and Molecular Pathology ,medicine ,Biomarkers, Tumor ,Animals ,Humans ,1306 Cancer Research ,education ,Aged ,Mutation ,education.field_of_study ,business.industry ,Interleukins ,Receptors, Interleukin ,Middle Aged ,medicine.disease ,Interleukin-10 Receptor beta Subunit ,Prognosis ,Primary tumor ,3. Good health ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Oncology ,Tumor progression ,030220 oncology & carcinogenesis ,Colonic Neoplasms ,Cancer research ,ras Proteins ,2730 Oncology ,Female ,KRAS ,Neoplasm Recurrence, Local ,business ,Signal Transduction - Abstract
Purpose:The cytokine IL22 promotes tumor progression in murine models of colorectal cancer. However, the clinical significance of IL22 in human colorectal cancer remains unclear. We sought to determine whether the IL22 pathway is associated with prognosis in human colorectal cancer, and to identify mechanisms by which IL22 can influence disease progression.Experimental Design:Transcriptomic data from stage II/III colon cancers in independent discovery (GSE39582 population-based cohort, N = 566) and verification (PETACC3 clinical trial, N = 752) datasets were used to investigate the association between IL22 receptor expression (encoded by the genes IL22RA1 and IL10RB), tumor mutation status, and clinical outcome using Cox proportional hazard models. Functional interactions between IL22 and mutant KRAS were elucidated using human colorectal cancer cell lines and primary tumor organoids.Results:Transcriptomic analysis revealed a poor-prognosis subset of tumors characterized by high expression of IL22RA1, the alpha subunit of the heterodimeric IL22 receptor, and KRAS mutation [relapse-free survival (RFS): HR = 2.93, P = 0.0006; overall survival (OS): HR = 2.45, P = 0.0023]. KRAS mutations showed a similar interaction with IL10RB and conferred the worst prognosis in tumors with high expression of both IL22RA1 and IL10RB (RFS: HR = 3.81, P = 0.0036; OS: HR = 3.90, P = 0.0050). Analysis of human colorectal cancer cell lines and primary tumor organoids, including an isogenic cell line pair that differed only in KRAS mutation status, showed that IL22 and mutant KRAS cooperatively enhance cancer cell proliferation, in part through augmentation of the Myc pathway.Conclusions:Interactions between KRAS and IL22 signaling may underlie a previously unrecognized subset of clinically aggressive colorectal cancer that could benefit from therapeutic modulation of the IL22 pathway.
- Published
- 2019
31. An open-label, phase II study of patritumab deruxtecan (HER3-DXd, U3-1402) in patients (pts) with previously treated advanced/metastatic colorectal cancer (CRC)
- Author
-
Masayuki Kanai, Sabeen Mekan, Takayuki Yoshino, Yali Liu, Scott Kopetz, Hiroya Taniguchi, Kensei Yamaguchi, Sabine Tejpar, Geetha Pudussery, Arndt Vogel, Zev A. Wainberg, Yang Qiu, and Kanwal Pratap Singh Raghav
- Subjects
Cancer Research ,Antibody-drug conjugate ,Patritumab ,Tetrapeptide ,Colorectal cancer ,medicine.drug_class ,business.industry ,Phases of clinical research ,medicine.disease ,Monoclonal antibody ,body regions ,Oncology ,medicine ,Cancer research ,In patient ,skin and connective tissue diseases ,business ,Linker - Abstract
TPS157 Background: Patritumab deruxtecan (HER3-DXd; U3-1402) is a novel, investigational antibody drug conjugate comprising an anti-HER3 monoclonal antibody, a tetrapeptide-based linker, and a topoisomerase I inhibitor payload. Ongoing clinical trials of HER3-DXd in pts with metastatic breast cancer or non-small cell lung cancer have shown promising clinical activity and acceptable safety. HER3 (human epidermal growth receptor 3), a member of the tyrosine kinase receptor family, is overexpressed in most CRC tumors and associated with an adverse prognosis. Significant tumor regression with HER3-DXd has been observed in CRC murine xenograft models, regardless of KRAS mutation status. Here we introduce the design of a phase 2 study (U31402-A-U202) that is evaluating HER3-DXd in previously treated pts with advanced/metastatic CRC. Methods: U31402-A-U202 (NCT04479436) is an open-label, multicenter phase 2 study that will enroll 80 pts in the USA, Europe and Asia. Pts are enrolled who are aged ≥ 18 years with advanced/metastatic colorectal adenocarcinoma that is resistant/refractory/intolerant to ≥ 2 prior lines of therapy including a fluoropyrimidine, irinotecan, a platinum agent, an anti-EGFR agent (if clinically indicated), an anti-VEGF agent (unless contraindicated [CI]), and an immune checkpoint inhibitor (unless CI) for microsatellite instability-high CRC. Pts with current/previous interstitial lung disease or clinically severe pulmonary compromise are excluded. Archival tumor biopsy and pre-treatment tumor biopsy are collected from all pts at screening, with HER3 protein expression measured by immunohistochemistry (IHC). In part 1, results of the HER3 IHC assay from the pre-treatment tumor biopsy are used to assign pts into 1 of 2 cohorts (C). C1: HER3 high (IHC 3+, 2+), n = 24; C2: HER3 low/negative (IHC 1+, 0), n = 12. Pts receive 5.6 mg/kg HER3-DXd IV every 3 weeks. An interim futility analysis will be conducted separately for C1 and C2 and will determine enrollment in part 2, with 2 potential scenarios: enrollment continues irrespective of HER3 IHC status, or enrollment continues in HER3 high pts only. The primary objective is the evaluation of the antitumor activity of HER3-DXd as measured by objective response rate (ORR) (assessed by BICR according to RECIST v1.1). ORR will be summarized with the 2-sided 95% confidence interval. Secondary objectives include the evaluation of efficacy as measured by ORR (assessed by investigator according to RECIST v1.1), duration of response, time to tumor response, disease control rate, progression-free survival (assessed by investigator and BICR according to RECIST v1.1), overall survival, safety and tolerability, HER3 protein expression in tumor tissue and relationship with efficacy, and pharmacokinetic properties. Clinical trial information: NCT04479436.
- Published
- 2021
32. Somatic POLE proofreading domain mutation, immune response, and prognosis in colorectal cancer: a retrospective, pooled biomarker study
- Author
-
Evelyn Fessler, Sarah Briggs, Anita Sveen, Jan Paul Medema, Ian Tomlinson, Viktor H. Koelzer, Peter Dutton, Mauro Delorenzi, Martin D. Berger, Marco de Bruyn, Alessandro Lugli, Kay Lawson, Inti Zlobec, Dahmane Oukrif, Enric Domingo, Marco Novelli, Sergi Castellví-Bel, Rachel Kerr, Mark A. Glaire, Louis Vermeulen, Hans Morreau, Stine A. Danielsen, Sabine Tejpar, Luke Freeman-Mills, Jenifer Muñoz, Gerrit-Jan Liefers, Eleni Frangou, Hans W. Nijman, Arild Nesbakken, Emily Rayner, Tom van Wezel, David J. Kerr, Ragnhild A. Lothe, David N. Church, Arnoud Boot, Targeted Gynaecologic Oncology (TARGON), Translational Immunology Groningen (TRIGR), Center of Experimental and Molecular Medicine, Other departments, and Radiotherapy
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Colorectal cancer ,DNA polymerase epsilon ,Bioinformatics ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Biomarkers, Tumor ,Journal Article ,medicine ,Humans ,Poly-ADP-Ribose Binding Proteins ,Survival analysis ,Aged ,Retrospective Studies ,Aged, 80 and over ,Hepatology ,business.industry ,Proportional hazards model ,Gastroenterology ,Cancer ,Retrospective cohort study ,DNA Polymerase II ,Middle Aged ,Prognosis ,medicine.disease ,Survival Analysis ,030104 developmental biology ,030220 oncology & carcinogenesis ,Mutation ,Biomarker (medicine) ,Female ,DNA mismatch repair ,Colorectal Neoplasms ,business - Abstract
BACKGROUND: Precision cancer medicine depends on defining distinct tumour subgroups using biomarkers that may occur at very modest frequencies. One such subgroup comprises patients with exceptionally mutated (ultramutated) cancers caused by mutations that impair DNA polymerase epsilon (POLE) proofreading.METHODS: We examined the association of POLE proofreading domain mutation with clinicopathological variables and immune response in colorectal cancers from clinical trials (VICTOR, QUASAR2, and PETACC-3) and colorectal cancer cohorts (Leiden University Medical Centre 1 and 2, Oslo 1 and 2, Bern, AMC-AJCC-II, and Epicolon-1). We subsequently investigated its association with prognosis in stage II/III colorectal cancer by Cox regression of pooled individual patient data from more than 4500 cases from these studies.FINDINGS: Pathogenic somatic POLE mutations were detected in 66 (1·0%) of 6517 colorectal cancers, and were mutually exclusive with mismatch repair deficiency (MMR-D) in the 6277 cases for whom both markers were determined (none of 66 vs 833 [13·4%] of 6211; pINTERPRETATION: POLE proofreading domain mutations identify a subset of immunogenic colorectal cancers with excellent prognosis. This association underscores the importance of rare biomarkers in precision cancer medicine, but also raises important questions about how to identify and implement them in practice.FUNDING: Cancer Research UK, Academy of Medical Sciences, Health Foundation, EU, ERC, NIHR, Wellcome Trust, Dutch Cancer Society, Dutch Digestive Foundation.
- Published
- 2016
33. Publisher Correction: Zeb2 drives invasive and microbiota-dependent colon carcinoma
- Author
-
Joachim Taminau, Chris Callewaert, Esther Hoste, Geert Berx, Ioanna Petta, Steven Goossens, Andy Wullaert, Niels Vandamme, Emre Etlioglu, Louis Boon, Sabine Tejpar, Pratyaksha Wirapati, Jody J. Haigh, Gert De Hertogh, Gillian Blancke, Mozes Sze, Lars Vereecke, Bart N. Lambrecht, Hanna-Kaisa Vikkula, Geert van Loo, Sven Jonckheere, Pamela Baldin, Pieter Van Vlierberghe, Karolina Slowicka, Emilie Dumas, Eugene Tulchinsky, David Nittner, Enrico Radaelli, UCL - (SLuc) Centre du cancer, and UCL - (SLuc) Service d'anatomie pathologique
- Subjects
Cancer Research ,Oncology ,Colon carcinoma ,business.industry ,Cancer research ,Medicine ,business - Abstract
No abstract available
- Published
- 2020
34. A phase III study of nivolumab (NIVO), NIVO + ipilimumab (IPI), or chemotherapy (CT) for microsatellite instability-high (MSI-H)/mismatch repair-deficient (dMMR) metastatic colorectal cancer (mCRC): Checkmate 8HW
- Author
-
Ming Lei, James Novotny, Jean-Marie Ledeine, Kentaro Yamazaki, Sandzhar Abdullaev, Heinz-Josef Lenz, Thierry André, Sabine Tejpar, and Andrew Scott Paulson
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Chemotherapy ,Colorectal cancer ,business.industry ,medicine.medical_treatment ,Microsatellite instability ,Ipilimumab ,medicine.disease ,digestive system diseases ,03 medical and health sciences ,0302 clinical medicine ,Microsatellite Stable ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,DNA mismatch repair ,Nivolumab ,business ,030215 immunology ,medicine.drug - Abstract
TPS266 Background: Patients (pts) with MSI-H/dMMR mCRC treated with CT have poorer outcomes than pts with microsatellite stable/MMR proficient mCRC. NIVO (anti–programmed death [PD]-1) and IPI (anti–cytotoxic T lymphocyte antigen-4 [CTLA-4]) are immune checkpoint inhibitors that act synergistically and promote antitumor immune response by complementary mechanisms. NIVO±IPI received accelerated US FDA approval for MSI-H/dMMR mCRC that progressed after fluoropyrimidine, oxaliplatin, and irinotecan treatment based on the phase 2, non-randomized, multicohort CheckMate 142 study. Indirect comparisons suggest that NIVO (3 mg/kg) + low-dose IPI (1 mg/kg) provides improved clinical benefit vs NIVO (investigator-assessed [INV] objective response rate [ORR] 55% vs 31%; 12-month [mo] INV progression-free survival [PFS] rate 71% vs 50%; 12-mo overall survival [OS] rate 85% vs 73%) with a favorable benefit-risk profile for previously treated MSI-H/dMMR mCRC (Overman et al. JCO 2018). NIVO+low-dose IPI also demonstrated robust and durable clinical benefit in first-line MSI-H/dMMR mCRC (INV ORR 64%; 12-mo INV PFS rate 77%; 12-mo OS rate 84%; Lenz et al. ASCO 2019, #3521). To date, no prospective phase 3 studies have reported results for anti–PD-1, anti–PD-1 + anti–CTLA-4, or CT in MSI-H/dMMR mCRC; these treatments will be evaluated in the international, multicenter, open-label, randomized, phase 3 CheckMate 8HW (NCT04008030) study. Methods: Approximately 494 pts aged ≥18 years with histologically confirmed recurrent or mCRC, irrespective of prior treatment with CT and/or targeted agents, not amenable to surgery, and with known tumor MSI-H or dMMR status, and Eastern Cooperative Oncology Group performance status ≤1 will be randomized to receive NIVO, NIVO+IPI, or investigator’s choice CT (pts in the CT arm can receive NIVO+IPI upon progression). The primary endpoint is PFS, assessed by blinded independent central review (BICR). Secondary endpoints include PFS by INV, ORR and disease control rate by BICR and INV, OS, time to and duration of response. Exploratory endpoints include safety. Clinical trial information: NCT04008030.
- Published
- 2020
35. The potential in artificial intelligence-driven radiomic signature to predict survival in patients with metastatic colorectal cancer treated with cetuximab-based therapy
- Author
-
Binsheng Zhao, Peter Eggleton, Hubert Piessevaux, Lawrence H. Schwartz, Lin Lu, Laurent Dercle, Sabine Tejpar, and Min Qian
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Cetuximab ,Colorectal cancer ,business.industry ,medicine.disease ,Folinic acid ,Internal medicine ,medicine ,In patient ,Treatment decision making ,business ,medicine.drug - Abstract
247 Background: This analysis was undertaken to forecast survival and enhance treatment decisions for patients (pts) with colorectal cancer (CRC) with liver metastases sensitive to folinic acid, fluorouracil and irinotecan (FOLFIRI) alone [F] or in combination with cetuximab [FC] using simple quantitative radiomic changes between CT scans at baseline and 8 weeks. Methods: We retrospectively analyzed 667 pts with KRAS-unselected metastatic CRC in NCT00154102 treated with F and FC. CT quality was classified as high (HQ) or standard (SQ), and four data sets were created and named by treatment quality. Pts were randomly assigned 1:2 to training or validation sets: FCHQ, 38/78 pts; FCSQ, 62/124 pts; FHQ, 51/78 pts; FSQ, 78/158 pts. A machine-learning signature was trained using data set FCHQ to classify pts as treatment-sensitive or treatment-insensitive using just 4 of 3,499 potential radiomic imaging features. Performance was calibrated/validated using ROC curves. Hazard ratios (HRs) and Cox regression models were used to evaluate association with overall survival (OS). Results: The signature used decrease in tumor heterogeneity plus boundary infiltration to successfully predict sensitivity to FC (FCHQ: AUC, 0.80; FCSQ: AUC, 0.72) but failed with non-cetuximab regimens (FHQ: AUC, 0.59; FSQ: AUC, 0.55). The radiomic signature outperformed existing biomarkers ( KRAS mutational status and tumor shrinkage by RECIST 1.1) for sensitivity to cetuximab-based therapy and was strongly associated with OS in the cetuximab-containing sets FCHQ (HR, 44.3; p = 0.0001) and FCSQ (HR, 6.5; p = 0.005). Conclusions: This signature, derived from simple radiomic analysis of tumor imaging phenotype using only standard-of-care CT scans, appeared to be treatment-specific and was superior to all tested prognostic biomarkers. The signature provided early prediction of sensitivity and survival and could be used to guide treatment continuation decisions.
- Published
- 2020
36. Abstract A075: SPECTA: The EORTC translational research platform for Europe
- Author
-
Marie Morfouace, Aleksandra Stevovic, Jean-Yves Blay, Teresa de Rojas, Marie Vinches, Martin G. McCabe, Sabine Tejpar, and Vassilis Golfinopoulos
- Subjects
Cancer Research ,education.field_of_study ,medicine.medical_specialty ,business.industry ,Population ,Translational research ,Precision medicine ,medicine.disease ,Central Pathology Review ,Biobank ,Clinical trial ,Oncology ,Informed consent ,Medicine ,Medical physics ,Sarcoma ,education ,business - Abstract
SPECTA is the translational research platform from the European Organisation for Research and Treatment of Cancer (EORTC), which facilitates pan-European research projects by providing a comprehensive infrastructure: a single protocol and Patient Informed Consent to enrol patients with cancer, a centralised database for clinical data collection (including long-term follow-up and quality of life), a centralised biobank and quality control sample workflow, expert molecular laboratories dedicated to the needs of the translational research projects. So far, 11 countries with 36 sites are open for recruitment with 43 authorized investigators. Two SPECTA projects focus on rare populations. The first, AYA (Adolescent and Young Adults) enrols patients aged between 12 and 29 years old, diagnosed with high grade glioma or sarcoma. AYA is a pilot study with a strong collaboration, between DKFZ and EORTC, aiming at diagnosis confirmation (central pathology review by pathologist experts and methylation diagnosis), and extensive molecular analysis (WES, RNAseq and methylation array). The aim is not only to have a direct impact on patients through the clinical advice provided to the treating physician by the molecular tumor board, but also to build knowledge for future clinical trials in this population with unmet needs. The second, Arcagen, is a collaborative project with the EUropean network for RAre CANcer (EURACAN), the European network for rare cancer. This project, with the support of Roche, aims at building a clinically annotated genomic database (NGS panels for FMI) of 1,000 rare cancer patients from the 10 EURACAN domains. So far, 81 patients have been recruited: sarcoma (n=43), thymic malignancies (n=16), germ cell tumor (n=10), and rare head and neck tumor (n=15). Druggable alterations were found in about 15% of patients. Molecular reports will be sent to local clinicians, the study results will be used for designing for the next generation of clinical trials in adult rare cancers. The last project currently enrolling in SPECTA is IMMUcan, an Innovative Medicine Initiative project. IMMUcan will generate broad molecular (WES and RNAseq) and cellular profiling data (multiplex immunofluorescence and imaging mass cytometry) of tumor and its microenvironment. Integration of this information with clinical data, will promote understanding on how the immune system and tumors interact, and the impact of current therapeutic interventions. For this purpose, we will enrol 3,000 patients with NSCLC, HNSCC, breast, colorectal and renal cell cancers. For all projects, we faced recurrent issues regarding tumor and peripheral sample quality. We overcomed those issues by performing optimization studies for pan European collection of biological material (such as PBMCs, ctDNA, vesicles or FFPE slides stability for imaging). SPECTA is an academic solution for next level precision medicine, with all the clinical, operational and scientific challenges that it implies. Citation Format: Marie Vinches, Teresa de Rojas, Aleksandra Stevovic, Martin Mccabe, Jean Yves Blay, Vassilis Golfinopoulos, Sabine Tejpar, Marie Morfouace. SPECTA: The EORTC translational research platform for Europe [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics; 2019 Oct 26-30; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2019;18(12 Suppl):Abstract nr A075. doi:10.1158/1535-7163.TARG-19-A075
- Published
- 2019
37. Managing synchronous liver metastases from colorectal cancer: A multidisciplinary international consensus
- Author
-
Alberto Sobrero, Norihiro Kokudo, Evelyne M. Loyer, Lars Påhlman, Aimery de Gramont, Eric Van Cutsem, René Adam, Jean Nicolas Vauthey, Laura Rubbia-Brandt, Francis Kunstlinger, Sabine Tejpar, Catherine Teh, Philippe Rougier, Graeme J. Poston, and Joan Figueras
- Subjects
Synchronous colorectal liver metastases ,medicine.medical_specialty ,Consensus ,Colorectal cancer ,medicine.medical_treatment ,ddc:616.07 ,Asymptomatic ,Remnant liver ,Multidisciplinary team management ,Surgery ,Systemic therapy ,Meta-Analysis as Topic ,Multidisciplinary approach ,medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,Pathological ,Randomized Controlled Trials as Topic ,Chemotherapy ,Cancer och onkologi ,medicine.diagnostic_test ,business.industry ,General surgery ,Liver Neoplasms ,Magnetic resonance imaging ,General Medicine ,medicine.disease ,Oncology ,Radiology Nuclear Medicine and imaging ,Cancer and Oncology ,Treatment strategy ,medicine.symptom ,business ,Colorectal Neoplasms - Abstract
An international panel of multidisciplinary experts convened to develop recommendations for managing patients with colorectal cancer (CRC) and synchronous liver metastases (CRCLM). A modified Delphi method was used. CRCLM is defined as liver metastases detected at or before diagnosis of the primary CRC. Early and late metachronous metastases are defined as those detected ⩽12months and >12months after surgery, respectively. To provide information on potential curability, use of high-quality contrast-enhanced computed tomography (CT) before chemotherapy is recommended. Magnetic resonance imaging is increasingly being used preoperatively to aid detection of subcentimetric metastases, and alongside CT in difficult situations. To evaluate operability, radiology should provide information on: nodule size and number, segmental localization and relationship with major vessels, response after neoadjuvant chemotherapy, non-tumoral liver condition and anticipated remnant liver volume. Pathological evaluation should assess response to preoperative chemotherapy for both the primary tumour and metastases, and provide information on the tumour, margin size and micrometastases. Although the treatment strategy depends on the clinical scenario, the consensus was for chemotherapy before surgery in most cases. When the primary CRC is asymptomatic, liver surgery may be performed first (reverse approach). When CRCLM are unresectable, the goal of preoperative chemotherapy is to downsize tumours to allow resection. Hepatic resection should not be denied to patients with stable disease after optimal chemotherapy, provided an adequate liver remnant with inflow and outflow preservation remains. All patients with synchronous CRCLM should be evaluated by a hepatobiliary multidisciplinary team. publisher: Elsevier articletitle: Managing synchronous liver metastases from colorectal cancer: A multidisciplinary international consensus journaltitle: Cancer Treatment Reviews articlelink: http://dx.doi.org/10.1016/j.ctrv.2015.06.006 content_type: article copyright: Copyright © 2015 The Authors. Published by Elsevier Ltd. ispartof: Cancer Treatment Reviews vol:41 issue:9 pages:729-41 ispartof: location:Netherlands status: published
- Published
- 2015
- Full Text
- View/download PDF
38. KRAS, NRAS, BRAF mutation comparison of endoscopic and surgically removed primary CRC paired samples: is endoscopy biopsy material adequate for molecular evaluation?
- Author
-
X Saegart, V. De Vriendt, Z Saridaki, G. De Hertogh, Sabine Tejpar, Dora Hatzidaki, L. De Smedt, and Sofie Palmans
- Subjects
Adult ,Male ,Proto-Oncogene Proteins B-raf ,Neuroblastoma RAS viral oncogene homolog ,Cancer Research ,medicine.medical_specialty ,Pathology ,Class I Phosphatidylinositol 3-Kinases ,Colorectal cancer ,Biopsy ,DNA Mutational Analysis ,Colonoscopy ,colorectal cancer ,Adenocarcinoma ,Biology ,Real-Time Polymerase Chain Reaction ,medicine.disease_cause ,GTP Phosphohydrolases ,Phosphatidylinositol 3-Kinases ,medicine ,Humans ,Molecular Diagnostics ,Aged ,Aged, 80 and over ,Mutation ,medicine.diagnostic_test ,Gene Expression Profiling ,endoscopic/primary samples ,Membrane Proteins ,Middle Aged ,medicine.disease ,Genes, ras ,Oncology ,RAS/BRAF ,Female ,Histopathology ,KRAS ,Radiology ,Colorectal Neoplasms - Abstract
Background: An everyday clinical practice dilemma in the 20–30% of metastatic colorectal cancer (CRC) patients that have not been operated on their primary tumour, is, under which specific histopathology and molecular circumstances, an endoscopic biopsy could be considered adequate to provide a representative RAS/BRAF molecular status to guide treatment. Methods: A consecutive series of 193 paired biopsy and primary CRC tumour samples between August 2008 and 2010 available in the Department of Pathology archives, University Hospitals, KU Leuven were retrieved. For a pair to be included, in the endoscopic biopsy, 20% of invasive adenocarcinoma cells should be present and enough slides to yield an extracted DNA concentration of ⩾5 ng μl−1, and no
- Published
- 2015
39. Personalized treatment is better than one treatment fits all in the management of patients with mCRC: a consensus statement
- Author
-
Claus-Henning Köhne, Adam Nosworthy, Yasser Abdel Kader, Fadi Nasr, Osman Gokhan Demir, Serdar Turhal, Riaz Mall, Assel Tumanova, Sabine Tejpar, Suayib Yalcin, Dmitry Osinsky, Hafez Halawani, Diaeddine Trad, and Andrey Meshcheryakov
- Subjects
Proto-Oncogene Proteins B-raf ,Vascular Endothelial Growth Factor A ,Oncology ,Cancer Research ,medicine.medical_specialty ,Bevacizumab ,Colorectal cancer ,Central asia ,Personalized treatment ,Cetuximab ,Oncogene Protein p21(ras) ,Antibodies, Monoclonal, Humanized ,Internal medicine ,medicine ,Humans ,Mutational status ,Panitumumab ,Neoplasm Metastasis ,Precision Medicine ,business.industry ,Antibodies, Monoclonal ,Combination chemotherapy ,General Medicine ,medicine.disease ,digestive system diseases ,ErbB Receptors ,Colorectal Neoplasms ,business ,medicine.drug - Abstract
ABSTRACT The VEGF- (bevacizumab) and EGFR- (cetuximab and panitumumab) targeting monoclonal antibodies have become integral components of the first-line treatment strategies for patients with metastatic colorectal cancer (mCRC). Increasingly combination chemotherapy, with or without a targeted agent, is being used to facilitate curative liver resection and improve survival rates in patients with initially unresectable but potentially resectable mCRC. Currently, the only selective marker for the treatment of patients with mCRC is tumor RAS mutational status. BRAF status is a strong prognostic indicator. Medical and clinical oncologists from Central Asia, Russia, the Middle East, Africa and Turkey reviewed data for the use of targeted agents in the treatment of patients with mCRC and have formed recommendations for the biological of choice first-line for patients with mCRC.
- Published
- 2014
40. Prognosis of stage II and III colon cancer treated with adjuvant 5-fluorouracil or FOLFIRI in relation to microsatellite status: results of the PETACC-3 trial†
- Author
-
Arnaud Roth, Z Saridaki, Mauro Delorenzi, Dirk Klingbiel, F. Bosman, and Sabine Tejpar
- Subjects
medicine.medical_specialty ,Pathology ,congenital, hereditary, and neonatal diseases and abnormalities ,Colorectal cancer ,Leucovorin ,Gastroenterology ,Descending colon ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Ascending colon ,neoplasms ,030304 developmental biology ,ddc:616 ,0303 health sciences ,business.industry ,Microsatellite instability ,nutritional and metabolic diseases ,Hematology ,medicine.disease ,digestive system diseases ,3. Good health ,Irinotecan ,medicine.anatomical_structure ,Oncology ,Chemotherapy, Adjuvant ,Fluorouracil ,030220 oncology & carcinogenesis ,Colonic Neoplasms ,Cohort ,FOLFIRI ,Camptothecin ,Microsatellite Instability ,Neoplasm Recurrence, Local ,business ,Microsatellite Repeats ,medicine.drug - Abstract
BACKGROUND: Although colon cancer (CC) with microsatellite instability (MSI) has a more favorable prognosis than microsatellite stable (MSS) CC, the impact varies according to clinicopathological parameters. We studied how MSI status affects prognosis in a trial-based cohort of stage II and III CC patients treated with 5-fluorouracil (5-FU)/leucovorin or FOLFIRI. MATERIALS AND METHODS: Tissue specimens of 1254 patients were tested for 10 different loci and were classified as MSI-high (MSI-H) when three or more loci were unstable and MSS otherwise. Study end points were overall survival (OS) and relapse-free survival (RFS). RESULTS: In stage II, RFS and OS were better for patients with MSI-H than with MSS CC [hazard ratio (HR) 0.26, 95% CI 0.10-0.65, P = 0.004 and 0.16, 95% CI 0.04-0.64, P = 0.01). In stage III, RFS was slightly better for patients with MSI-H CC (HR 0.67, 95% CI 0.46-0.99, P = 0.04), but the difference was not statistically significant for OS (HR 0.70, 95% CI 0.44-1.09, P = 0.11). Outcomes for patients with MSI-H CC were not different between the two treatment arms. RFS was better for patients with MSI-H than with MSS CC in the right and left colon, whereas for OS this was significant only in the right colon. For patients with KRAS- and BRAF-mutated CC, but not for double wild-type patients, RFS and OS were significantly better when the tumors were also MSI-H. An interaction test was statistically significant for KRAS and MSI status (P = 0.005), but not for BRAF status (P = 0.14). CONCLUSIONS: Our results confirm that for patients with stage II CC but less so for those with stage III MSI-H is strongly prognostic for RFS and OS. In the presence of 5-FU treatment, stage II patients with MSI-H tumors maintain their survival advantage in comparison with MSS patients and adding irinotecan has no added benefit. CLINICALTRIALSGOV IDENTIFIER: NCT00026273.
- Published
- 2017
41. Prediction of overall survival in stage II and III colon cancer beyond TNM system: a retrospective, pooled biomarker study
- Author
-
Frank A. Sinicrope, Christophe Rosty, Polly A. Newcomb, Steven R. Alberts, Sabine Tejpar, Daniel D. Buchanan, Ragnhild A. Lothe, Michael Mason, J. Milburn Jessup, Mark Clendenning, Daniel J. Sargent, Anita Sveen, Mauro Delorenzi, Justin Guinney, Amanda I. Phipps, Arild Nesbakken, Brian M. Bot, Rodrigo Dienstmann, and Stine A. Danielsen
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Colorectal cancer ,Concordance ,Population ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Gastrointestinal Tumors ,Tumor Microenvironment ,medicine ,Humans ,education ,education.field_of_study ,business.industry ,Proportional hazards model ,Microsatellite instability ,KRAS mutation ,Original Articles ,Hematology ,Adult ,Aged ,Aged, 80 and over ,Biomarkers, Tumor/metabolism ,Colonic Neoplasms/metabolism ,Colonic Neoplasms/mortality ,Colonic Neoplasms/pathology ,Female ,Kaplan-Meier Estimate ,Male ,Middle Aged ,Neoplasm Staging ,Proportional Hazards Models ,Retrospective Studies ,Treatment Outcome ,Young Adult ,BRAF mutation ,colon cancer ,microsatellite instability ,prognosis ,medicine.disease ,Prognosis ,Chemotherapy regimen ,digestive system diseases ,3. Good health ,Colon cancer ,030104 developmental biology ,030220 oncology & carcinogenesis ,Biomarker (medicine) ,KRAS ,Colorectal Neoplasms ,business - Abstract
Altres ajuts: This work was partially supported by grant UM1 CA167551 from the National Cancer Institute (NCI), National Institutes of Health (NIH), and through cooperative agreements with members of the Colon Cancer Family Registry (CCFR) and Principal Investigators. Centers contributing to this analysis include the Seattle Colorectal Cancer Family Registry (U01/U24CA074794), Australasian Colorectal Cancer Family Registry (U01 CA074778 and U01/U24 CA097735), Mayo Clinic Cooperative Family Registry for Colon Cancer Studies (U01/U24 CA074800), and Ontario Familial Colorectal Cancer Registry (U01/U24 CA074783). This work was also supported by NCI/NIH grant K07CA172298 and K05CA152715 (to AIP). The content of this article does not necessarily reflect the views or policies of the NIH or any of the collaborating centers in the CCFR, nor does mention of trade names, commercial products, or organizations imply endorsement by the US Government. This study was partly supported by the "Norwegian Cancer Society" and the "Research Council of Norway" (no grants apply). TNM staging alone does not accurately predict outcome in colon cancer (CC) patients who may be eligible for adjuvant chemotherapy. It is unknown to what extent the molecular markers microsatellite instability (MSI) and mutations in BRAF or KRAS improve prognostic estimation in multivariable models that include detailed clinicopathological annotation. After imputation of missing at random data, a subset of patients accrued in phase 3 trials with adjuvant chemotherapy (n = 3016)-N0147 (NCT00079274) and PETACC3 (NCT00026273)-was aggregated to construct multivariable Cox models for 5-year overall survival that were subsequently validated internally in the remaining clinical trial samples (n = 1499), and also externally in different population cohorts of chemotherapy-treated (n = 949) or -untreated (n = 1080) CC patients, and an additional series without treatment annotation (n = 782). TNM staging, MSI and BRAF V600E mutation status remained independent prognostic factors in multivariable models across clinical trials cohorts and observational studies. Concordance indices increased from 0.61-0.68 in the TNM alone model to 0.63-0.71 in models with added molecular markers, 0.65-0.73 with clinicopathological features and 0.66-0.74 with all covariates. In validation cohorts with complete annotation, the integrated time-dependent AUC rose from 0.64 for the TNM alone model to 0.67 for models that included clinicopathological features, with or without molecular markers. In patient cohorts that received adjuvant chemotherapy, the relative proportion of variance explained (R 2) by TNM, clinicopathological features and molecular markers was on an average 65%, 25% and 10%, respectively. Incorporation of MSI, BRAF V600E and KRAS mutation status to overall survival models with TNM staging improves the ability to precisely prognosticate in stage II and III CC patients, but only modestly increases prediction accuracy in multivariable models that include clinicopathological features, particularly in chemotherapy-treated patients.
- Published
- 2017
42. ALK, ROS1, and NTRK Rearrangements in Metastatic Colorectal Cancer
- Author
-
A. Falcone, Salvatore Siena, Alexa B. Schrock, Federica Morano, Seung Tae Kim, Chiara Cremolini, Giovanni Fucà, Filippo de Braud, Siraj M. Ali, Rosa Berenato, Jason Christiansen, Carlotta Antoniotti, Jaclyn F. Hechtman, Vincent A. Miller, Robert H. Shoemaker, Luca Lazzari, Andrea Sartore-Bianchi, Alessio Amatu, Filippo Pietrantonio, James Sun, Luca Novara, Danielle Murphy, Sabine Tejpar, Niall C. Tebbutt, Jeffrey S. Ross, Marissa Chen, Massimo Milione, Philip J. Stephens, Bosun Min, Federica Di Nicolantonio, Alberto Bardelli, and Jeeyun Lee
- Subjects
0301 basic medicine ,Oncology ,Male ,Cancer Research ,Lung Neoplasms ,Colorectal cancer ,neoplasms ,0302 clinical medicine ,Anaplastic lymphoma kinase ,Anaplastic Lymphoma Kinase ,Peritoneal Neoplasms ,Aged, 80 and over ,Gene Rearrangement ,Hazard ratio ,Liver Neoplasms ,ROS ,Middle Aged ,Protein-Tyrosine Kinases ,Prognosis ,Primary tumor ,Combined Modality Therapy ,3. Good health ,Gene Expression Regulation, Neoplastic ,Survival Rate ,030220 oncology & carcinogenesis ,Lymphatic Metastasis ,Female ,Colorectal Neoplasms ,mutation, neoplasms, BRAF, ROS, colorectal cancer ,Adult ,medicine.medical_specialty ,Adolescent ,colorectal cancer ,BRAF ,03 medical and health sciences ,Young Adult ,Internal medicine ,Proto-Oncogene Proteins ,medicine ,ROS1 ,Biomarkers, Tumor ,Humans ,Receptor, trkA ,Survival rate ,Aged ,business.industry ,Cancer ,Gene rearrangement ,medicine.disease ,030104 developmental biology ,Cancer research ,mutation ,business ,Follow-Up Studies - Abstract
Background ALK, ROS1, and NTRK fusions occur in 0.2% to 2.4% of colorectal cancers. Pioneer cases of metastatic colorectal cancer (mCRC) patients bearing rearrangements who benefited from anti-ALK, ROS, and TrkA-B-C therapies have been reported previously. Here we aimed at characterizing the clinical and molecular landscape of ALK, ROS1, and NTRK rearranged mCRC. Methods Clinical features and molecular characteristics of 27 mCRC patients bearing ALK, ROS1, and NTRK rearranged tumors were compared with those of a cohort of 319 patients not bearing rearrangements by means of Fisher's exact, χ2 test, or Mann-Whitney test as appropriate. Overall survival curves were estimated with the Kaplan-Meier method and compared using the log-rank test. A Cox proportional hazard model was adopted in the multivariable analysis. Deep molecular and immunophenotypic characterizations of rearranged cases, including those described in The Cancer Genome Atlas database, were performed. All statistical tests were two-sided. Results Closely recalling the "BRAF history," ALK, ROS1, and NTRK rearrangements more frequently occurred in elderly patients (P = .02) with right-sided tumors (P < .001) and node-spreading (P = .03), RAS wild-type (P < .001), and MSI-high (P < .001) cancers. All patients bearing ALK, ROS1, and NTRK fusions had shorter overall survival (15.6 months, 95% confidence interval [CI] = 0.0 to 20.4 months) than negative patients (33.7 months, 95% CI = 28.3 to 42.1 months), both in the univariate (hazard ratio [HR] = 2.17, 95% CI = 1.03 to 4.57, P < .001) and multivariable models (HR = 2.33, 95% CI = 1.10 to 4.95, P = .02). All four evaluable patients with rearrangements showed primary resistance to anti-epidermal growth factor receptor agents. Frequent association with potentially targetable RNF43 mutations was observed in MSI-high rearranged tumors. Conclusions ALK, ROS1, and NTRK rearrangements define a new rare subtype of mCRC with extremely poor prognosis. Primary tumor site, MSI-high, and RAS and BRAF wild-type status may help to identify patients bearing these alterations. While sensitivity to available treatments is limited, targeted strategies inhibiting ALK, ROS, and TrkA-B-C provided encouraging results.
- Published
- 2017
43. Societal challenges of precision medicine: Bringing order to chaos
- Author
-
Augustinus van Dongen, Jeffrey A. Moscow, Nils Wilking, Daniel Hochhauser, Erasmus Schneider, Jonas Bergh, Adrian Senderowicz, Ultan McDermott, Simon J. Dovedi, Philip A. Beer, John W.M. Martens, Robert L. Becker, Mehdi Mesri, Tawnya C. McKee, Shakun Malik, Denis Lacombe, Roberto Salgado, Thomas Lillie, Tracy Lively, Irene Norstedt, Jan Bogaerts, Kathy Oliver, Moritz Gerstung, Francesco Pignatti, Olli Tenhunen, Hartmut Juhl, Helen M. Moore, Stefan Michiels, Richard L. Schilsky, Kim Lyerly Herbert, Antonio Tito Fojo, Robert J. Kinders, Daniel O’Connor, Christine Vietz, Sabine Tejpar, Stephen M. Hewitt, Eric C. Polley, Nitzan Rosenfeld, Jan H.M. Schellens, Sumimasa Nagai, Shyamala Maheswaran, Vassilis Golfinopoulos, Wim J.G. Oyen, and Medical Oncology
- Subjects
0301 basic medicine ,Cancer Research ,business.industry ,media_common.quotation_subject ,Genomic signature ,Routine practice ,Precision medicine ,Bioinformatics ,Article ,Clinical trial ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Promotion (rank) ,Oncology ,Risk analysis (engineering) ,Drug development ,Assay validation: biomarkers: Preanalytical and analytical validation: precision oncology ,Order (exchange) ,030220 oncology & carcinogenesis ,Medicine ,Biomarker (medicine) ,business ,ta317 ,media_common - Abstract
The increasing number of drugs targeting specific proteins implicated in tumourigenesis and the commercial promotion of relatively affordable genome-wide analyses has led to an increasing expectation among patients with cancer that they can now receive effective personalised treatment based on the often complex genomic signature of their tumour. For such approaches to work in routine practice, the development of correspondingly complex biomarker assays through an appropriate and rigorous regulatory framework will be required. It is becoming increasingly evident that a re-engineering of clinical research is necessary so that regulatory considerations and procedures facilitate the efficient translation of these required biomarker assays from the discovery setting through to clinical application. This article discusses the practical requirements and challenges of developing such new precision medicine strategies, based on leveraging complex genomic profiles, as discussed at the Innovation and Biomarkers in Cancer Drug Development meeting (8th-9th September 2016, Brussels, Belgium).
- Published
- 2017
44. The tyrosine phosphatase PTPRO sensitizes colon cancer cells to anti-EGFR therapy through activation of SRC-mediated EGFR signaling
- Author
-
Eva Budinská, Anna Sablina, Sabine Tejpar, Sharat Singh, Naga-Sailaja Imjeti, Layka Abbasi Asbagh, Maria Francesca Baietti, Loredana Vecchione, Iria Vazquez, Mikhail Steklov, Veerle De Vriendt, Bart Jacobs, Pascale Zimmermann, and Nicholas Hoe
- Subjects
MAP Kinase Signaling System ,EGFR ,Protein tyrosine phosphatase ,phosphatase ,03 medical and health sciences ,0302 clinical medicine ,Epidermal growth factor ,Cell Line, Tumor ,Humans ,Proto-Oncogene Proteins c-cbl ,RNA, Messenger ,Epidermal growth factor receptor ,Phosphorylation ,PTPRO ,Protein Kinase Inhibitors ,030304 developmental biology ,EGFR inhibitors ,0303 health sciences ,Epidermal Growth Factor ,biology ,Kinase ,Chemistry ,Receptor-Like Protein Tyrosine Phosphatases, Class 3 ,Gefitinib ,HCT116 Cells ,3. Good health ,ErbB Receptors ,EGFR inhibitor ,HEK293 Cells ,src-Family Kinases ,colon cancer ,Oncology ,030220 oncology & carcinogenesis ,Colonic Neoplasms ,SRC kinase ,Quinazolines ,Cancer research ,biology.protein ,Caco-2 Cells ,Signal transduction ,HT29 Cells ,Signal Transduction ,Research Paper ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Inappropriate activation of epidermal growth factor receptor (EGFR) plays a causal role in many cancers including colon cancer. The activation of EGFR by phosphorylation is balanced by receptor kinase and protein tyrosine phosphatase activities. However, the mechanisms of negative EGFR regulation by tyrosine phosphatases remain largely unexplored. Our previous results indicate that protein tyrosine phosphatase receptor type O (PTPRO) is down-regulated in a subset of colorectal cancer (CRC) patients with a poor prognosis. Here we identified PTPRO as a phosphatase that negatively regulates SRC by directly dephosphorylating Y416 phosphorylation site. SRC activation triggered by PTPRO down-regulation induces phosphorylation of both EGFR at Y845 and the c-CBL ubiquitin ligase at Y731. Increased EGFR phosphorylation at Y845 promotes its receptor activity, whereas enhanced phosphorylation of c-CBL triggers its degradation promoting EGFR stability. Importantly, hyperactivation of SRC/EGFR signaling triggered by loss of PTPRO leads to high resistance of colon cancer to EGFR inhibitors. Our results not only highlight the PTPRO contribution in negative regulation of SRC/EGFR signaling but also suggest that tumors with low PTPRO expression may be therapeutically targetable by anti-SRC therapies.
- Published
- 2014
45. Personalized medicine in metastatic colorectal cancer treated with anti-epidermal growth factor receptor agents: A future opportunity?
- Author
-
Sabine Tejpar and Hubert Piessevaux
- Subjects
Oncology ,medicine.medical_specialty ,Biological therapies ,biology ,business.industry ,Colorectal cancer ,Treatment options ,General Medicine ,Precision medicine ,medicine.disease ,Treatment success ,Internal medicine ,Anti-Epidermal Growth Factor Receptor ,Immunology ,medicine ,biology.protein ,Epidermal growth factor receptor ,Personalized medicine ,business - Abstract
Treatment options for colorectal cancer have increased substantially in the past decade, with the introduction of novel biological therapies targeting cancer-specific molecules leading to significantly improved outcomes. Despite access to these treatments, we are not yet in an era where we can fully personalize treatment choices for patients with colorectal cancer. A number of prognostic and predictive markers have been identified that appear to be directly related to sensitivity to targeted therapies, such as those against epidermal growth factor receptor. However, the sensitivities of individual tumors toward different biological agents appear to be more complex. It seems that a more complete molecular signature of the tumor must be taken into account when making individual treatment choices. In the absence of having fully elucidated the influence of these prognostic or predictive markers, other surrogate markers of early treatment success may be useful in determining whether to continue treatment with a particular agent. In this review, we discuss the role of molecular markers in choosing appropriate treatment for the individual patient, along with the use of measuring the depth of response to a particular agent to assist decisions on whether to continue therapy in colorectal cancer.
- Published
- 2014
46. Feedback activation of HER3 attenuates response to EGFR inhibitors in colon cancer cells
- Author
-
Sharat Singh, Bart Jacobs, Joe Michel, Albert Bosch-Vilaró, Xinjun Liu, Tony Ng, Phillip Kim, Richard Kirkland, Sabine Tejpar, Gregory Weitsman, Valentina Pomella, Layka Abbasi Asbagh, Borivoj Vojnovic, and Paul R. Barber
- Subjects
0301 basic medicine ,Oncology ,Receptor, ErbB-3 ,Colorectal cancer ,Receptor, ErbB-2 ,Cetuximab ,Tyrosine-kinase inhibitor ,0302 clinical medicine ,Epidermal growth factor ,Phosphorylation ,skin and connective tissue diseases ,EGFR inhibitors ,Feedback, Physiological ,dimerization ,3. Good health ,Up-Regulation ,ErbB Receptors ,030220 oncology & carcinogenesis ,Colonic Neoplasms ,feedback loop ,medicine.drug ,Research Paper ,medicine.medical_specialty ,medicine.drug_class ,Cell Survival ,colorectal cancer ,Lapatinib ,03 medical and health sciences ,Breast cancer ,HER3 ,Internal medicine ,Cell Line, Tumor ,medicine ,Humans ,Protein Kinase Inhibitors ,neoplasms ,Cell Proliferation ,cetuximab resistance ,business.industry ,Cancer ,medicine.disease ,digestive system diseases ,Surgery ,body regions ,030104 developmental biology ,Drug Resistance, Neoplasm ,Quinazolines ,Protein Multimerization ,business - Abstract
// Albert Bosch-Vilaro 1, * , Bart Jacobs 1, * , Valentina Pomella 1 , Layka Abbasi Asbagh 1 , Richard Kirkland 2 , Joe Michel 2 , Sharat Singh 2 , Xinjun Liu 2 , Phillip Kim 2 , Gregory Weitsman 3 , Paul R Barber 4 , Borivoj Vojnovic 3, 4 , Tony Ng 3, 5, 6 , Sabine Tejpar 1 1 Laboratory of Molecular Digestive Oncology, Department of Oncology, KU Leuven, Leuven, Belgium 2 Prometheus Laboratories, San Diego, CA, USA 3 Richard Dimbleby Department of Cancer Research, Randall Division & Division of Cancer Studies, King’s College London, Guy’s Medical School Campus, London, UK 4 Department of Oncology, Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK 5 Breast Cancer Now Research Unit, King’s College London, London, UK 6 UCL Cancer Institute, Paul O'Gorman Building, University College London, London, UK * These authors contributed equally to this work Correspondence to: Sabine Tejpar, email: sabine.tejpar@uzleuven.be Keywords: colorectal cancer, cetuximab resistance, HER3, dimerization, feedback loop Received: September 14, 2016 Accepted: November 30, 2016 Published: December 09, 2016 ABSTRACT The EGFR inhibitor cetuximab is approved for the treatment of colorectal cancer. However, both innate and acquired resistance mechanisms, including compensatory feedback loops, limit its efficacy. Nevertheless, the emergence of these feedback loops has remained largely unexplored to date. Here, we showed feedback upregulation of HER3 and induction of HER3 phosphorylation after cetuximab treatment in colon cancer cells. We also showed that this upregulation occurs, at least partly, through AKT inhibition. Together with this, we observed increased HER2:HER3 dimerization upon cetuximab treatment. Interestingly, lapatinib, a dual EGFR and HER2 tyrosine kinase inhibitor, blocked the increase of cetuximab-induced HER3 phosphorylation. Additionally, we showed that upon HER3 knockdown, cetuximab combined with lapatinib was able to decrease cell viability compared to HER3 expressing cells. These results suggest the existence of a cetuximab-induced feedback HER3 activation that could potentially result in reduced cetuximab efficacy in colorectal cancer patients. Taken together, we provide evidence of the limited effectiveness of cetuximab monotherapy compared to rational combinations.
- Published
- 2016
47. Characterizations of DNA copy number variations and spatio-temporal intra tumor heterogeneity in liver metastasis from colorectal cancer patients
- Author
-
Y-J. Ko, Michael Witcher, Vincent Pelsser, Sabine Tejpar, Maud Marques, M. Couetoux du Tertre, Richard Dalfen, Eve St-Hilaire, B. Samson, Petr Kavan, Errol Camlioglu, Bernard Lespérance, Suzan McNamara, Claudia L. Kleinman, Adrian Gologan, Karen Gambaro, Mohammed Harb, Ronald Burkes, Gerald Batist, Félix Couture, Thierry Alcindor, and Lucas Sideris
- Subjects
Colorectal cancer ,business.industry ,Hematology ,medicine.disease ,Tumor heterogeneity ,Metastasis ,chemistry.chemical_compound ,Oncology ,chemistry ,medicine ,Cancer research ,Copy-number variation ,business ,DNA - Published
- 2018
48. Abstract 579: Immune features and neoantigen recognition in mismatch repair-proficient colorectal cancer liver metastases
- Author
-
Steven Hébert, Mélissa Mathieu, David Henault, Maud Marques, Éric Audemard, Mathieu Courcelles, Scott D. Brown, Pratyaksha Wirapati, Sabine Tejpar, Michael I. D'Angelica, Robert A. Holt, Sylvie Mader, Pierre Thibault, Gerald Batist, Claudia Kleinman, and Simon Turcotte
- Subjects
Cancer Research ,Oncology - Abstract
Immunotherapy for patients with non-highly mutated, mismatch repair-proficient (MMRprof) metastatic colorectal cancer has largely been unsuccessful thus far. Here, we used transcriptomics, genomics and in vitro analyses to study the adaptive immune features of colorectal cancer liver metastases (CRLMs), aiming to provide insights into the development of immunotherapies for this common malignancy. Analysis of the RNAseq data from 63 core biopsies obtained in patients enrolled in the Q-CROC-01 trial (NCT00984048) revealed 17 (27%) immune-reactive (IR) CRLMs, defined by concurrent relative high expression of antigen processing, immune cell, immune checkpoint, interferon-gamma response, cytokine, and chemokine transcripts. More T-cell receptor (TCR) sequences were found in IR vs. non-IR CRLMs (mean 89.8 ± 13.3 vs. 19.5 ± 4.0 CDR3 counts, p Our results support that naturally occurring neoAg-reactive T cells may be therapeutically harnessed in a subgroup of metastatic MMRprof colorectal cancer patients selected based on the level of intratumoral immune reactivity, and potentially enhanced by targeting immune checkpoints more biologically relevant than PD-1/PD-L1 and CTLA-4/CD80. Citation Format: Steven Hébert, Mélissa Mathieu, David Henault, Maud Marques, Éric Audemard, Mathieu Courcelles, Scott D. Brown, Pratyaksha Wirapati, Sabine Tejpar, Michael I. D'Angelica, Robert A. Holt, Sylvie Mader, Pierre Thibault, Gerald Batist, Claudia Kleinman, Simon Turcotte. Immune features and neoantigen recognition in mismatch repair-proficient colorectal cancer liver metastases [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 579.
- Published
- 2019
49. Copy number variation in longitudinal liver metastases biopsies in colorectal cancer identifies biomarker candidates of resistance to standard chemotherapy
- Author
-
Archana Srivastava, Félix Couture, Benoit Samson, Suzan McNamara, Y-J. Ko, Mohammed Harb, Richard Dalfen, Errol Camlioglu, Vincent Pelsser, Ronald L. Burkes, Maud Marques, Adrian Gologan, Karen Gambaro, Sabine Tejpar, Gerald Batist, Eve St-Hilaire, Lucas Sideris, Bernard Lespérance, Claudia L. Kleinman, Cyrla Hoffert, M. Couetoux du Tertre, and Petr Kavan
- Subjects
Oncology ,Chemotherapy ,medicine.medical_specialty ,business.industry ,Colorectal cancer ,medicine.medical_treatment ,Hematology ,medicine.disease ,Internal medicine ,medicine ,Biomarker (medicine) ,Copy-number variation ,business - Published
- 2019
50. Abstract 479: BRAF and KRAS mutation define distinct subtypes of the CpG island methylator phenotype in colorectal cancers
- Author
-
Pratyaksha Wirapati, Catherine Bond, Katia Nones, Ann-Marie Patch, Stephen H. Kazakoff, Nicola Waddell, Sabine Tejpar, Troy Dumenil, Diane McKeone, Lochlan Fennell, Paul Lochead, Shuji Ogino, Barbara A. Leggett, Vicki L. J. Whitehall, John V. Pearson, and Gunter Hartel
- Subjects
Cancer Research ,CpG Island Methylator Phenotype ,Colorectal cancer ,Genome scale ,Methylation ,Biology ,medicine.disease ,digestive system diseases ,Oncology ,Cancer genome ,DNA methylation ,Cancer research ,medicine ,Epigenetics ,neoplasms ,Gene transcript - Abstract
BACKGROUND AND AIMS: Colorectal cancer is an epigenetically heterogeneous disease, however the extent and spectrum of the CpG Island Methylator Phenotype (CIMP) is not clear.METHODS: Genome scale methylation and transcript expression were measured using the Illumina HM450 DNA methylation and HT12 V3 expression microarrays in 216 unselected colorectal cancers. Mutations in epigenetic regulators were assessed using CIMP-classified Cancer Genome Atlas exomes.RESULTS: CIMP-High cancers dichotomised into CIMP-H1 and CIMP-H2 based on methylation profile, which was supported by over-representation of BRAF (74%, P Citation Format: Vicki L. Whitehall, Lochlan Fennell, Troy Dumenil, Gunter Hartel, Katia Nones, Catherine Bond, Diane McKeone, Ann-Marie Patch, Stephen Kazakoff, John Pearson, Nicola Waddell, Pratyaksha Wirapati, Paul Lochead, Shuji Ogino, Sabine Tejpar, Barbara Leggett. BRAF and KRAS mutation define distinct subtypes of the CpG island methylator phenotype in colorectal cancers [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 479.
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.