#4169 cremec initiative: characterization of patient ... · cric002p (wt kras) cric013m (mutated...

1
CRIC013M (mutated KRAS) CRIC002P (wt KRAS) 0 200 400 600 800 1000 1200 1400 10 30 50 70 90 1 Tumor volume (mm 3 ) 0 200 400 600 800 1000 1200 1400 20 30 40 50 60 70 Days post implantation Tumor volume (mm 3 ) Cetuximab (12.5 mg/kg/adm) Days post implantation CRLRB022P in mice 0 500 1000 1500 15 20 25 30 35 Days post implantation Median Tumor volume (mm 3 ) Control Cetuximab (12.5 mg/kg/ adm) 5-FU (50.0 mg/kg/ adm) Oxaliplatin (5.0mg/kg/ adm) Cetuximab IP treatment 5-FU IV treatment Oxaliplatin IV treatment 0 500 1000 1500 15 20 25 30 35 Dayspost implantation MedianTumorvolume (mm 3 ) Control Cetuximab (12.5 mg/kg/adm) 5-FU (50.0 mg/kg/ adm) Oxaliplatin (5.0mg/kg/ adm) Cetuximab IP treatment 5-FU IV treatment Oxaliplatin IV treatment 0 500 1000 1500 15 20 25 30 35 Dayspost implantation MedianTumorvolume (mm 3 ) Control CPT-11 (22mg/kg/ Adm) CPT-11 IV treatment 3 CRLRB022P in rats Control Oxaliplatin (4 mg/kg/ adm) CPT-11 (40 mg/kg/ adm) Cetuximab (10 mg/kg/ adm) 5-FU (30 mg/kg/ adm) Days post implantation 70 60 50 40 30 20 10 Median tumour volume (mm 3 ) 10000 9000 8000 7000 6000 5000 4000 3000 2000 1000 0 Treatment start (D23) Control Oxaliplatin (4 mg/kg/ adm) CPT-11 (40 mg/kg/ adm) Cetuximab (10 mg/kg/ adm) 5-FU (30 mg/kg/ adm) (4 mg/kg/ adm) CPT- adm) (10 mg/kg/ 5- adm ) Days post implantation 70 60 50 40 30 20 10 Median tumour volume (mm 3 ) 10000 9000 8000 7000 6000 5000 4000 3000 2000 1000 0 Days post implantation 70 60 50 40 30 20 10 ) 10000 9000 8000 7000 6000 5000 4000 3000 2000 1000 0 Treatment start (D23) - adm) 5- ) - 5- ) - - ) - - ) - CRLRB022P in mice 0 500 1000 1500 15 20 25 30 35 Days post implantation Median Tumor volume (mm 3 ) Control Cetuximab (12.5 mg/kg/ adm) 5-FU (50.0 mg/kg/ adm) Oxaliplatin (5.0mg/kg/ adm) Cetuximab IP treatment 5-FU IV treatment Oxaliplatin IV treatment 0 500 1000 1500 15 20 25 30 35 Dayspost implantation MedianTumorvolume (mm 3 ) Control Cetuximab (12.5 mg/kg/adm) 5-FU (50.0 mg/kg/ adm) Oxaliplatin (5.0mg/kg/ adm) Cetuximab IP treatment 5-FU IV treatment Oxaliplatin IV treatment CRLRB022P in mice 0 500 1000 1500 15 20 25 30 35 Days post implantation Median Tumor volume (mm 3 ) Control Cetuximab (12.5 mg/kg/ adm) 5-FU (50.0 mg/kg/ adm) Oxaliplatin (5.0mg/kg/ adm) Cetuximab IP treatment 5-FU IV treatment Oxaliplatin IV treatment 0 500 1000 1500 15 20 25 30 35 Dayspost implantation MedianTumorvolume (mm 3 ) Control Cetuximab (12.5 mg/kg/adm) 5-FU (50.0 mg/kg/ adm) Oxaliplatin (5.0mg/kg/ adm) Cetuximab IP treatment 5-FU IV treatment Oxaliplatin IV treatment 0 500 1000 1500 15 20 25 30 35 Dayspost implantation MedianTumorvolume (mm 3 ) Control CPT-11 (22mg/kg/ Adm) CPT-11 IV treatment 3 0 500 1000 1500 15 20 25 30 35 Dayspost implantation MedianTumorvolume (mm 3 ) Control CPT-11 (22mg/kg/ Adm) CPT-11 IV treatment 3 CRLRB022P in rats Control Oxaliplatin (4 mg/kg/ adm) CPT-11 (40 mg/kg/ adm) Cetuximab (10 mg/kg/ adm) 5-FU (30 mg/kg/ adm) Days post implantation 70 60 50 40 30 20 10 Median tumour volume (mm 3 ) 10000 9000 8000 7000 6000 5000 4000 3000 2000 1000 0 Treatment start (D23) Control Oxaliplatin (4 mg/kg/ adm) CPT-11 (40 mg/kg/ adm) Cetuximab (10 mg/kg/ adm) 5-FU (30 mg/kg/ adm) (4 mg/kg/ adm) CPT- adm) (10 mg/kg/ 5- adm ) Days post implantation 70 60 50 40 30 20 10 Median tumour volume (mm 3 ) 10000 9000 8000 7000 6000 5000 4000 3000 2000 1000 0 Days post implantation 70 60 50 40 30 20 10 ) 10000 9000 8000 7000 6000 5000 4000 3000 2000 1000 0 Treatment start (D23) - adm) 5- ) - 5- ) - - ) - - ) - CRLRB022P in rats Control Oxaliplatin (4 mg/kg/ adm) CPT-11 (40 mg/kg/ adm) Cetuximab (10 mg/kg/ adm) 5-FU (30 mg/kg/ adm) Days post implantation 70 60 50 40 30 20 10 Median tumour volume (mm 3 ) 10000 9000 8000 7000 6000 5000 4000 3000 2000 1000 0 Days post implantation 70 60 50 40 30 20 10 Median tumour volume (mm 3 ) 10000 9000 8000 7000 6000 5000 4000 3000 2000 1000 0 Treatment start (D23) Control Oxaliplatin (4 mg/kg/ adm) CPT-11 (40 mg/kg/ adm) Cetuximab (10 mg/kg/ adm) 5-FU (30 mg/kg/ adm) (4 mg/kg/ adm) CPT- adm) (10 mg/kg/ 5- adm ) Days post implantation 70 60 50 40 30 20 10 Median tumour volume (mm 3 ) 10000 9000 8000 7000 6000 5000 4000 3000 2000 1000 0 Days post implantation 70 60 50 40 30 20 10 ) 10000 9000 8000 7000 6000 5000 4000 3000 2000 1000 0 Treatment start (D23) - adm) 5- ) - 5- ) - - ) - - ) - INTRODUCTION MATERIAL AND METHODS CReMEC initiative: Characterization of patient-derived colorectal tumor models and correlation with patient profile Sylvia Julien 1 , Ana Merino-Trigo 2 , Ludovic Lacroix 7,5 , Marc Pocard 3,5 , Diane Goéré 7,5 , Pascale Mariani 4,5 , Sophie Landron 8 , Ludovic Bigot 7,5 , Fariba Nemati 4,5 , Louis-Bastien Weiswald 3,5 , Denis Lantuas 3,5 , Loïc Morgand 9 , Grégoire Prévost 1 , Patrick Gonin 7,5 , Virginie Dangles-Marie 3,4,5 , Alain Pierré 8 , Alain Bruno 8 , Hugues De Thé 5,6 , Hany Soliman 5,6 , Manoel Nunes 2 , Loreley Calvet 2 , Patricia Vrignaud 2 , Olivier Duchamp 9 , Cyril Berthet 9 . 1 Ipsen, 2 sanofi-aventis, 3 Hôpital Lariboisière , 4 Institut Curie, 5 Cancéropole d’Ile de France, 6 Institut Universitaire d'Hématologie, Paris, 7 Institut Gustave Roussy, Villejuif, 8 Institut de recherche Servier, Croissy sur Seine, 9 Oncodesign, Dijon, France. RESULTS CONCLUSION #4169 Well characterized models representing the heterogeneity of human colorectal cancers (CRC) are needed to develop effective therapeutic agents. Establishment of such tools will allow a better prediction of the clinical outcome, taking into account the diversity of each patient tumor phenotype and genotype. For this purpose, we have associated efforts from hospitals, academic groups, biotech and pharmaceutical companies. The goal of this consortium is to create an experimental tumor model resource center to improve or strengthen drug development. From May 2007 to January 2009, 86 surgical specimens [59 primary (P) tumors, 19 metastasis (M), and 8 peritoneal carcinomatosis (C)] were collected from CRC patients (with informed consents and negative HBV, HCV, and HIVs serologies). Tumor samples were subcutaneously xenografted in nude mice and characterized as described below. We report here the results on our panel of models. Patients were informed and gave their consent for providing surgical tumor samples to CReMEC and for HIV1, HIV2, HBV and HCV serological status testing. Tumor samples were collected in 3 medical centers of the Paris-Ile-de-France area: Institut Curie, Institut Gustave Roussy, Hôpital Lariboisière. Fresh tumor material was conditioned into RPMI 1640 with 200 U/mL penicillin, 200 μg/mL streptomycin and 2.5 μg/mL fungizone. Initial xenografting was carried out at the site of sampling, within 12 hours after specimen collection. Procedures were performed according to ethical guidelines for animal care and handling. 20-40 mg fragments were xenografted subcutaneously either in the flank or in the interscapular area, in 3-5 immunodeficient SWISS nude mice Grafted mice were kept for a maximum of 4 months without tumor growth. Clinical data collection Relevant clinical information was collected by the attending physician and included in a standardized data sheet. Identification of the data sheet is anonymous. Histological characterization Samples were fixed for a maximum of 48 hours in alcohol-formalin-acetic acid (AFA) and embedded in paraffin. 5 μm sections were stained with hematoxylin-eosin-saffron. Molecular characterization – sequencing CGH array analysis: Evaluation of genome-wide, gene copy number was evaluated using a 244k CGH array Agilent technology. It was carried out on DNA from the patient sample or at an early passages (P0/P1), and at the passage used for the pharmacological evaluation (P8-9). DNA sequencing: The following genetic markers, relevant in CRC, were selected for sequencing: APC (exons 9 & 16), KRAS (exons 2 & 3), BRAF (exons 11 & 15), TP53 (exons 2 to 11), CTNNB1 (exon 3), PIK3CA (exons 10 and 21), FBXW7 (exons 4 to 11), EGFR (exons 18 to 21) and AKT1 (exon 4). Sanger direct sequencing was performed after PCR amplification of exons of genes harboring hotspot mutations as described in Catalogue Of Somatic Mutations In Cancer (COSMIC: http://www.sanger.ac.uk/genetics/CGP/cosmic/ ). Determination of Microsatellite Instability (MSI) status: The MSI status was determined according to the Consensus Conference (or Revised Bethesda Guidelines) (J. Natl Cancer Inst 2004, 96, 261-268) recommendations using the following five quasimonomorphic markers: NR21, BAT26, BAT25, NR24 and NR22. Pharmacological characterization Tested agents: 5-fluorouracil (5-FU), oxaliplatin (L-OHP), irinotecan (CPT-11) or cetuximab, were tested. In vivo determination of antitumor activity: Tumor fragments were subcutaneously xenografted in SCID mice or nude rats. Tumor-bearing animals were randomized when the mean tumor volume reached 100-200 mm 3 in mice or 500-700 mm 3 in rats. Compounds were formulated in glucose 5% in water. 3 cytotoxic drugs were IV administered, and tested at 70% of their respective highest non toxic dose and using the following regimen in mice: 5-FU at 56 mg/kg/adm Q4Dx2 (30 mg/kg/adm Q7Dx3 in rats) , L-OHP at 5 mg/kg/adm Q4Dx2 (4 mg/kg/adm Q4Dx3 in rats), CPT-11 at 22 mg/kg/adm, Q2Dx3 (40 mg/kg/adm, Q7Dx3in rats) ; 1 targeting therapy was IP administrated: cetuximab at 12.5 mg/kg/adm, (Q3Dx2)x2 (10 mg/kg/adm, IV, Q7Dx3 in rats). Antitumor activity was evaluated by calculating the (T/C) ratio: (T/C) (%) = X 100 median T VOL- DY – median T VOL-DX median C VOL- DY – median C VOL-DX Preservation of the tumor phenotype and genotype Clinical characteristics and in vivo tumorgraft take rate Scoring criteria: -= (T/C) > 42 % + = 10 < (T/C) 42 % ++ = 0 (T/C) 10 % (stable disease) +++ = (T/C) <0 % (tumor regression) 8 4 12 5 CPT-11 7 3 4 15 Cetuximab 0 0 2 27 L-OHP 0 5 17 7 5-FU +++ ++ + - Activity score 22 4 4 <10% 10-50% >50% Mucus Secretion 22 4 4 <10% 10-50% >50% Mucus Secretion Tumor Models Differentiation 2 12 13 2 1 WD WD, MD WD, MD, PD MD, PD PD, UD Differentiation 2 12 13 2 1 WD WD, MD WD, MD, PD MD, PD PD, UD Mutations 1 0 0 0 KRAS/BRAF +++ ++ + - Activity score 4 1 3 11 EGFR mutated pathway 3 2 1 3 EGFR wild-type pathway* 0 0 0 2 BRAF 0 0 2 1 KRAS/PIK3CA 3 1 1 8 KRAS Fresh tumors Surgery * Patient identification * Patient clinical history * Patient informed consent * Negative HIV, HBV, HCV serology Paraffin Tumor bank Snap-frozen Database Tumor growth Molecular Histological Pharmacological Characterization 26 31 36 41 46 51 0 200 400 600 800 1000 1200 1400 CPT-11 treatment CPT-11 22 mg/kg IV Control (untreated) Days after tumor inoculation Median tumor size (mg +/- IQR) Tumor grafting P0 P2 – P6 Establishment of in vivo tumorgraft models RPMI/DMSO freezing Fresh tumors Surgery Fresh tumors Surgery * Patient identification * Patient clinical history * Patient informed consent * Negative HIV, HBV, HCV serology * Patient identification * Patient clinical history * Patient informed consent * Negative HIV, HBV, HCV serology Paraffin Tumor bank Snap-frozen Paraffin Tumor bank Snap-frozen Database Tumor growth Molecular Histological Pharmacological Characterization 26 31 36 41 46 51 0 200 400 600 800 1000 1200 1400 CPT-11 treatment CPT-11 22 mg/kg IV Control (untreated) Days after tumor inoculation Median tumor size (mg +/- IQR) Database Database Tumor growth Molecular Histological Pharmacological Characterization Tumor growth Molecular Histological Pharmacological Characterization 26 31 36 41 46 51 0 200 400 600 800 1000 1200 1400 CPT-11 treatment CPT-11 22 mg/kg IV Control (untreated) Days after tumor inoculation Median tumor size (mg +/- IQR) 26 31 36 41 46 51 0 200 400 600 800 1000 1200 1400 CPT-11 treatment CPT-11 22 mg/kg IV Control (untreated) CPT-11 treatment CPT-11 22 mg/kg IV Control (untreated) Days after tumor inoculation Median tumor size (mg +/- IQR) Tumor grafting P0 P2 – P6 Establishment of in vivo tumorgraft models RPMI/DMSO freezing Tumor grafting P0 P2 – P6 Establishment of in vivo tumorgraft models RPMI/DMSO freezing PR: Partial Response CR: Complete Response TFS: Tumor Free Survival Tumor take rate (%) Parameter Class Number Frequency (%) Site of origin Primary 37 66.1 62.7 Peritoneal carcinosis 5 8.9 62.5 Metastasis # 14 25.0 73.7 Sex Female 35 62.5 66.0 Male 21 37.5 63.6 Age 0- 50y 11 20.4 78.6 [ 50y ; 60y] 12 24.5 50.0 [ 60y ; 70y] 12 20.4 60.0 [ 70y ; 80y] 15 24.5 68.2 >80y 6 10.2 100 Stage 0 - I 2 3.7 22.2 II 8 14.8 57.1 III 13 24.1 92.9 IV 31 57.4 70.4 Patient characteristics in the collection # hepatic, splenic and mesenteric lymph node metastasis Tumor take rate (%) Parameter Class Number Frequency (%) Site of origin Primary 37 66.1 62.7 Peritoneal carcinosis 5 8.9 62.5 Metastasis # 14 25.0 73.7 Sex Female 35 62.5 66.0 Male 21 37.5 63.6 Age 0- 50y 11 20.4 78.6 [ 50y ; 60y] 12 24.5 50.0 [ 60y ; 70y] 12 20.4 60.0 [ 70y ; 80y] 15 24.5 68.2 >80y 6 10.2 100 Stage 0 - I 2 3.7 22.2 II 8 14.8 57.1 III 13 24.1 92.9 IV 31 57.4 70.4 Patient characteristics in the collection # hepatic, splenic and mesenteric lymph node metastasis Significant correlation* between CEA serum level of patient and tumor take of all types Significant distribution* of primary tumors in regards of lymph node status (panel A, N1 = 1 to 6, N2 = 7 to 15 positive regional lymph nodes), stages (panel B) and differentiation status (panel C). No other significant correlation were found among the following parameters: gender, age, resection extent, lymphatic embolies, perinervous invasion, initial treatment, genotype * p values were calculated with logical regression test Gene mutation profile and response to chemotherapy Histopathological analyses completed for 30 colon tumor models were in concordance to those observed in the corresponding patient’s tumor. CGH analysis showed very similar profile between early and advance passages The expression and localization of β β β-catenin, CEA, CAIX, EGFR, CD105 and LYVE-1 is ongoing. The majority of tumor models were non-mucinous adenocarcinomas Splenic metastasis (1/86) Mesenteric lymph node (1/86) All models collected (86) Left Right Primary tumor (59/86) Carcinosis (8/86) Hepatic Metastasis (16/86) Ovarian metastasis (1/86) Left Rect. Left Right Left Right Rect. Splenic metastasis (1/86) Mesenteric lymph node (1/86) All models collected (86) Left Right Primary tumor (59/86) Carcinosis (8/86) Hepatic Metastasis (16/86) Ovarian metastasis (1/86) Left Rect. Left Right Left Right Rect. Right colon Left colon Rectum Models growing on nude mice (56) Primary tumor (37/56) Ca r c i n o s i s ( 5 / 5 6 ) Hepatic Metastasis (12/56) Splenic metastasis (1/56) Mesenteric lymph node (1/56) Left Right Rect.Left Right Left Right Rect. Right colon Left colon Rectum Right colon Left colon Rectum Models growing on nude mice (56) Primary tumor (37/56) Ca r c i n o s i s ( 5 / 5 6 ) Hepatic Metastasis (12/56) Splenic metastasis (1/56) Mesenteric lymph node (1/56) Left Right Rect.Left Right Left Right Rect. Only 1 model showed a poor differentiated (PD) to undifferentiated (UD) characteristic, as well as its correspondent patient Similar CGH profiles between early and advance passages Loss in chromosomes 3p and 4q in advance passage Most models with EGFR mutated pathway are resistant to Cetuximab, nevertheless several Cetuximab sensitive KRAS-mutated models have been identified. Patient: CR-IGR-034P Patient: CR-LRB-010P Patient: CR-LRB-008M P10: CR-LRB-010P P11: CR-LRB-008M P11: CR-IGR-034P Patient Tumor Model Well Differentiated Well to Moderate Differentiated Poor to UnDifferentiated Patient: CR-IGR-034P Patient: CR-LRB-010P Patient: CR-LRB-008M P10: CR-LRB-010P P11: CR-LRB-008M P11: CR-IGR-034P Patient: CR-IGR-034P Patient: CR-LRB-010P Patient: CR-LRB-008M Patient: CR-IGR-034P Patient: CR-IGR-034P Patient: CR-LRB-010P Patient: CR-LRB-010P Patient: CR-LRB-008M Patient: CR-LRB-008M P10: CR-LRB-010P P11: CR-LRB-008M P11: CR-IGR-034P P10: CR-LRB-010P P10: CR-LRB-010P P11: CR-LRB-008M P11: CR-LRB-008M P11: CR-IGR-034P P11: CR-IGR-034P Patient Tumor Model Patient Tumor Model Well Differentiated Well to Moderate Differentiated Poor to UnDifferentiated Sensitivity to Cetuximab versus mutated genes involved in EGFR pathway T/C = -41% (Day 31), 9/10 PR & 4/10 CR CRIGR023M GAIN LOSS CRIGR002C GAIN LOSS * Mutations of PTEN or other genes related to EGFR pathway have not been yet characterized Multiple mutation profiles are observed in our tumor panel (n=60), matching the human tumor genetic heterogeneity Pharmacological studies exhibit diversity in the response to chemotherapy 13/29 tumorgrafts show tumor regression after treatment with at least one standard monotherapy Diversity of colorectal cancers is fully addressed in this collection of patient-derived tumor models. Genotype and phenotype are largely preserved throughout the model establishment process. Difference of gene mutation and drug sensitivity profiles are observed between the models. Plan to complete the full correlation analysis between clinical data, gene mutations, transcriptome profile, ex-vivo and in vivo drug sensitivity. Perspectives to fully exploit this new collection for new drug candidate selection. T/C = < 0 % (Day 36), 10/10 CR + 100% TFS 0 CEA (ng/mL) Positive xenograft take 5 10 15 20 25 30 35 40 45 50 170 175 180 4 1 11 2.3 8 27 p = 0.0292 (*) All types of tumor samples / CEA dosing Negative xenograft take 0 5 10 15 20 25 30 35 40 45 50 170 180 4 1 11 2.3 8 27 p = 0.0292 (*) 0 CEA (ng/mL) Positive xenograft take 5 10 15 20 25 30 35 40 45 50 170 175 180 4 1 11 2.3 8 27 p = 0.0292 (*) All types of tumor samples / CEA dosing Negative xenograft take 0 5 10 15 20 25 30 35 40 45 50 170 180 4 1 11 2.3 8 27 p = 0.0292 (*) A Percentage (%) N0 N0 N1 + N2 N1 + N2 0 20 40 60 80 100 120 (n=11) (n=26) (n=5) (n=17) Primary tumor samples / Lymph node status Positive xenograft take Negative xenograft take p = 0.0038 (**) N0 N0 N1 + N2 N1 + N2 0 20 40 60 80 100 120 (n=11) (n=26) (n=5) (n=17) p = 0.0038 (**) A Percentage (%) N0 N0 N1 + N2 N1 + N2 0 20 40 60 80 100 120 (n=11) (n=26) (n=5) (n=17) Primary tumor samples / Lymph node status Positive xenograft take Negative xenograft take p = 0.0038 (**) N0 N0 N1 + N2 N1 + N2 0 20 40 60 80 100 120 (n=11) (n=26) (n=5) (n=17) p = 0.0038 (**) B Stages 0-I Stages 0-I Stage II Stage II Stage III Stage III Stage IV Stage IV 0 20 40 60 80 100 120 (n=7) (n=6) (n=7) (n=6) (n=13) (n=14) Primary tumor samples / Stages Negative xenograft take Positive xenograft take p = 0.0101 (*) Percentage (%) Stages 0-I Stages 0-I Stage II Stage II Stage III Stage III Stage IV Stage IV 0 20 40 60 80 100 120 (n=7) (n=6) (n=7) (n=6) (n=13) (n=14) p = 0.0101 (*) B Stages 0-I Stages 0-I Stage II Stage II Stage III Stage III Stage IV Stage IV 0 20 40 60 80 100 120 (n=7) (n=6) (n=7) (n=6) (n=13) (n=14) Primary tumor samples / Stages Negative xenograft take Positive xenograft take p = 0.0101 (*) Percentage (%) Stages 0-I Stages 0-I Stage II Stage II Stage III Stage III Stage IV Stage IV 0 20 40 60 80 100 120 (n=7) (n=6) (n=7) (n=6) (n=13) (n=14) p = 0.0101 (*) C Primary tumor samples / Differentiation 0 Well Well Fairly Fairly Weakly 20 40 60 80 100 120 (n=10) (n=11) (n=9) (n=25) Percentage (%) p = 0.0287 (*) Negative xenograft take Positive xenograft take 0 Well Well Fairly Fairly Weakly 20 40 60 80 100 120 (n=10) (n=11) (n=9) (n=25) p = 0.0287 (*) C Primary tumor samples / Differentiation 0 Well Well Fairly Fairly Weakly 20 40 60 80 100 120 (n=10) (n=11) (n=9) (n=25) Percentage (%) p = 0.0287 (*) Negative xenograft take Positive xenograft take 0 Well Well Fairly Fairly Weakly 20 40 60 80 100 120 (n=10) (n=11) (n=9) (n=25) p = 0.0287 (*) Similar tumor response ranges have been observed in mice and rats T/C in mice L-OHP: 45% 5-FU: 45% Cetuximab: 78% T/C in rat L-OHP: 104% 5-FU: 63% Cetuximab: 19% CPT-11: 1% T/C in mice CPT-11 < 0% 70% 80% 80% 50% 70% 90% 60% 80% 80% 50% 90% 90% 70% 60% 90% 60% 40% 100% 40% 70% 40% 80% 50% 80% 50% 80% 80% 80% 80% 50% 90% 40% 80% 30% 90% 80% 90% 90% 85% 60% 85% 50% 60% 70% 40% 40% 80% 50% 30% 50% 70% 90% 70% 80% 80% 50% 50% 50% 50% 80% CRIGR023M CRIC004M CRIC007M CRIGR007P CRIGR025P CRLRB010P CRLRB011M CRLRB013P CRLRB014P CRIC018P CRIC002P CRIC003P CRIC025M CRIGR004P CRIGR008P CRLRB007P CRIGR015P CRLRB019C CRIGR021P CRIC020P CRIC021M CRIGR032P CRIGR029P CRIGR012P CRIGR014P CRIC012P CRIC013M CRLRB008M CRLRB009C CRIGR003P CRIGR009P CRIGR016P CRIGR020P CRIGR001M CRIGR038C CRLRB017P CRIGR034P CRLRB018P CRLRB003P CRLRB004P CRLRB015P CRIC006M CRIC019P CRIC029P CRIC028M CRIGR002P CRIGR002C CRIGR052M CRLRB005P CRIGR047P CRIGR048M CRLRB022P CRIC008P CRIC009M CRIC010P CRIGR011C CRIGR039P CRIGR043P CRIC005P CRIC014P TP53 62% 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 0 0 0 0 0 0 0 0 0 0 1 1 1 1 1 1 1 1 1 0 0 0 1 1 1 1 0 1 1 1 0 1 1 0 0 0 0 0 0 0 0 APC 53% 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 0 1 1 1 1 1 1 1 1 0 0 0 0 0 0 0 0 0 0 0 1 1 1 0 0 0 0 0 0 0 0 0 0 0 1 1 1 0 0 0 0 0 KRAS 45% 1 1 1 1 1 1 1 1 1 1 0 0 0 0 0 0 0 0 0 0 0 1 1 1 1 0 1 1 1 1 1 1 1 1 1 1 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 1 1 1 0 0 PIK3CA 10% 1 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 1 1 0 0 0 0 1 1 0 0 0 0 0 0 0 0 0 0 0 1 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 FBXW7 8% 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 1 1 1 1 0 1 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 BRAF 5% 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 * 0 0 0 0 0 0 0 0 0 1 1 0 1 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 CTNNB1 3% 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 1 1 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 EGFR 2% 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 1 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 AKT1 0% 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 MSI status MSS MSS MSS MSS MSS MSS MSS MSS MSS MSS MSS MSS MSS MSI-L MSI-L MSS MSS MSS MSS MSS MSS MSS MSS MSS MSI-L MSS MSS MSS MSI-L MSS MSS MSS MSS MSI-L MSI-H MSI-H MSS MSI-L MSI-L MSS MSS MSI-L MSI-L MSI-L MSS MSS MSS MSS MSS MSS MSS MSS MSS MSS Xenograft 1 1 1 1 1 1 1 1 1 1 1 1 1 0 1 1 0 1 0 1 1 1 1 1 1 0 1 1 1 1 1 1 0 0 1 1 1 1 1 1 1 1 1 1 1 1 1 1 0 1 1 1 1 1 1 1 1 1 1 0 CGH Analysis 0 1 1 0 0 1 0 0 0 0 0 1 0 1 1 0 0 0 0 0 0 1 0 1 1 0 0 0 1 0 0 1 1 0 0 0 1 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 1 80 0 0 0 1 0 Other ERCC1-A138T FCGR2A-F196Y * BRAF-N594D 5-FU + - - - - + + + ++ + + ++ + + + + + ++ + - + ++ + ++ + + - + - l-OHP - - - - - - - - - - - - + - - - + - - - - - - - - - - - - CPT-11 + + ++ + ++ +++ - +++ - + - - +++ - + ++ + +++ + + + + +++ ++ + +++ +++ + +++ CETUXIMAB + ++ +++ + - - +++ +++ +++ - - - +++ + - +++ - - - - - - ++ ++ +++ + - - - Mutation MSI-L Low (MSI-L) or High (MSI-H) microsatellite instability Wild-type No data available Model from the same patient Xenograft model established CGH analysis % above sample name: % of tumor cells in the analyzed sample Scoring criteria: - = (T/C) > 42 % + = 10 < (T/C) 42 % ++ = 0 (T/C) 10 % (stable desease) +++ = (T/C) <0 % (tumor regression) Characterization in progress % beside gene name: % of mutations among all sequenced samples

Upload: others

Post on 12-Mar-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: #4169 CReMEC initiative: Characterization of patient ... · CRIC002P (wt KRAS) CRIC013M (mutated KRAS) 0 200 400 600 800 1000 1200 1400 10 30 50 70 90 110 Tumor volume (mm 3) 0 200

CRIC013M (mutated KRAS) CRIC002P (wt KRAS)

0

200

400

600

800

1000

1200

1400

10 30 50 70 90 110

Tum

or v

olum

e (m

m3 )

0

200

400

600

800

1000

1200

1400

20 30 40 50 60 70

Days post implantation

Tum

or v

olum

e (m

m3 )

Cetuximab(12.5 mg/kg/adm)Days post implantation

CRLRB022P in mice

0

500

1000

1500

15 20 25 30 35

Days post implantation

Med

ian

Tum

orvo

lum

e (m

m3 )

ControlCetuximab (12.5 mg/kg/ adm)5-FU (50.0 mg/kg/ adm)Oxaliplatin (5.0mg/kg/ adm)Cetuximab IP treatment5-FU IV treatmentOxaliplatin IV treatment

0

500

1000

1500

15 20 25 30 35

Dayspost implantation

Med

ianT

umor

volu

me

(mm

3)

ControlCetuximab (12.5 mg/kg/adm)5-FU (50.0 mg/kg/ adm)Oxaliplatin (5.0mg/kg/ adm)Cetuximab IP treatment5-FU IV treatmentOxaliplatin IV treatment

0

500

1000

1500

15 20 25 30 35Dayspost implantation

Med

ianT

umor

volu

me

(mm

3 )

Control

CPT-11 (22mg/kg/ Adm)

CPT-11 IV treatment

3

CRLRB022P in rats

ControlOxaliplatin (4 mg/kg/ adm)CPT-11 (40 mg/kg/ adm)Cetuximab (10 mg/kg/ adm)5-FU (30 mg/kg/ adm)

Days post implantation70605040302010

Med

ian

tum

our

volu

me

(mm

3 ) 10000

9000

8000

7000

6000

5000

4000

3000

2000

1000

0

Treatment start(D23)

ControlOxaliplatin (4 mg/kg/ adm)CPT-11 (40 mg/kg/ adm)Cetuximab (10 mg/kg/ adm)5-FU (30 mg/kg/ adm)

(4 mg/kg/ adm)CPT- adm)

(10 mg/kg/5- adm)

Days post implantation70605040302010

Med

ian

tum

our

volu

me

(mm

3 ) 10000

9000

8000

7000

6000

5000

4000

3000

2000

1000

0

Days post implantation70605040302010

) 10000

9000

8000

7000

6000

5000

4000

3000

2000

1000

0

Treatment start(D23)

- adm)

5- )

-

5- )

-

- )

-

- )

-

CRLRB022P in mice

0

500

1000

1500

15 20 25 30 35

Days post implantation

Med

ian

Tum

orvo

lum

e (m

m3 )

ControlCetuximab (12.5 mg/kg/ adm)5-FU (50.0 mg/kg/ adm)Oxaliplatin (5.0mg/kg/ adm)Cetuximab IP treatment5-FU IV treatmentOxaliplatin IV treatment

0

500

1000

1500

15 20 25 30 35

Dayspost implantation

Med

ianT

umor

volu

me

(mm

3)

ControlCetuximab (12.5 mg/kg/adm)5-FU (50.0 mg/kg/ adm)Oxaliplatin (5.0mg/kg/ adm)Cetuximab IP treatment5-FU IV treatmentOxaliplatin IV treatment

CRLRB022P in mice

0

500

1000

1500

15 20 25 30 35

Days post implantation

Med

ian

Tum

orvo

lum

e (m

m3 )

ControlCetuximab (12.5 mg/kg/ adm)5-FU (50.0 mg/kg/ adm)Oxaliplatin (5.0mg/kg/ adm)Cetuximab IP treatment5-FU IV treatmentOxaliplatin IV treatment

0

500

1000

1500

15 20 25 30 35

Dayspost implantation

Med

ianT

umor

volu

me

(mm

3)

ControlCetuximab (12.5 mg/kg/adm)5-FU (50.0 mg/kg/ adm)Oxaliplatin (5.0mg/kg/ adm)Cetuximab IP treatment5-FU IV treatmentOxaliplatin IV treatment

0

500

1000

1500

15 20 25 30 35Dayspost implantation

Med

ianT

umor

volu

me

(mm

3 )

Control

CPT-11 (22mg/kg/ Adm)

CPT-11 IV treatment

3

0

500

1000

1500

15 20 25 30 35Dayspost implantation

Med

ianT

umor

volu

me

(mm

3 )

Control

CPT-11 (22mg/kg/ Adm)

CPT-11 IV treatment

3

CRLRB022P in rats

ControlOxaliplatin (4 mg/kg/ adm)CPT-11 (40 mg/kg/ adm)Cetuximab (10 mg/kg/ adm)5-FU (30 mg/kg/ adm)

Days post implantation70605040302010

Med

ian

tum

our

volu

me

(mm

3 ) 10000

9000

8000

7000

6000

5000

4000

3000

2000

1000

0

Treatment start(D23)

ControlOxaliplatin (4 mg/kg/ adm)CPT-11 (40 mg/kg/ adm)Cetuximab (10 mg/kg/ adm)5-FU (30 mg/kg/ adm)

(4 mg/kg/ adm)CPT- adm)

(10 mg/kg/5- adm)

Days post implantation70605040302010

Med

ian

tum

our

volu

me

(mm

3 ) 10000

9000

8000

7000

6000

5000

4000

3000

2000

1000

0

Days post implantation70605040302010

) 10000

9000

8000

7000

6000

5000

4000

3000

2000

1000

0

Treatment start(D23)

- adm)

5- )

-

5- )

-

- )

-

- )

-

CRLRB022P in rats

ControlOxaliplatin (4 mg/kg/ adm)CPT-11 (40 mg/kg/ adm)Cetuximab (10 mg/kg/ adm)5-FU (30 mg/kg/ adm)

Days post implantation70605040302010

Med

ian

tum

our

volu

me

(mm

3 ) 10000

9000

8000

7000

6000

5000

4000

3000

2000

1000

0

Days post implantation70605040302010

Med

ian

tum

our

volu

me

(mm

3 ) 10000

9000

8000

7000

6000

5000

4000

3000

2000

1000

0

Treatment start(D23)

ControlOxaliplatin (4 mg/kg/ adm)CPT-11 (40 mg/kg/ adm)Cetuximab (10 mg/kg/ adm)5-FU (30 mg/kg/ adm)

(4 mg/kg/ adm)CPT- adm)

(10 mg/kg/5- adm)

Days post implantation70605040302010

Med

ian

tum

our

volu

me

(mm

3 ) 10000

9000

8000

7000

6000

5000

4000

3000

2000

1000

0

Days post implantation70605040302010

) 10000

9000

8000

7000

6000

5000

4000

3000

2000

1000

0

Treatment start(D23)

- adm)

5- )

-

5- )

-

- )

-

- )

-

INTRODUCTION

MATERIAL AND METHODS

CReMEC initiative: Characterization of patient-deri ved colorectal tumor models and correlation with pa tient profileSylvia Julien 1, Ana Merino-Trigo 2, Ludovic Lacroix 7,5, Marc Pocard 3,5, Diane Goéré 7,5, Pascale Mariani 4,5, Sophie Landron 8, Ludovic Bigot 7,5, Fariba Nemati 4,5, Louis-Bastien Weiswald 3,5, Denis Lantuas 3,5, Loïc Morgand 9,

Grégoire Prévost 1, Patrick Gonin 7,5, Virginie Dangles-Marie 3,4,5, Alain Pierré 8, Alain Bruno 8, Hugues De Thé 5,6, Hany Soliman 5,6, Manoel Nunes 2, Loreley Calvet 2, Patricia Vrignaud 2, Olivier Duchamp 9, Cyril Berthet 9.

1Ipsen, 2sanofi-aventis, 3Hôpital Lariboisière , 4Institut Curie, 5Cancéropole d’Ile de France, 6Institut Universitaire d'Hématologie, Paris, 7Institut Gustave Roussy, Villejuif, 8Institut de recherche Servier, Croissy sur Seine, 9Oncodesign, Dijon, France.

RESULTS

CONCLUSION

#4169

Well characterized models representing the heterogeneity of human colorectal cancers (CRC) are needed to develop effective therapeutic agents. Establishment of such tools will allow a better prediction of the clinical outcome, taking into account the diversity of each patient tumor phenotype and genotype. For this purpose, we have associated efforts from hospitals, academic groups, biotech and pharmaceutical companies. The goal of this consortium is to create an experimental tumor model resource center to improve or strengthen drug development. From May 2007 to January 2009, 86 surgical specimen s [59 primary (P) tumors, 19 metastasis (M), and 8 peritoneal carcinomatosis (C)] were collected from C RC patients (with informed consents and negative HBV, HCV, and HIVs serologies). Tumor samples were subcutaneously xenografted in nude mice and characterized as described below. We report here the results on our panel of models.

� Patients were informed and gave their consent for providing surgical tumor samples to CReMEC and for HIV1, HIV2, HBV and HCV serological status testing.

� Tumor samples were collected in 3 medical centers o f the Paris-Ile-de-France area: Institut Curie, Institut Gu staveRoussy, Hôpital Lariboisière.

� Fresh tumor material was conditioned into RPMI 1640 with 200 U/mL penicillin, 200 µg/mL streptomycin and 2.5 µg/mL fungizone.

� Initial xenografting was carried out at the site of sampling, within 12 hours after specimen collection. Procedu res were performed according to ethical guidelines for animal care and handling.

� 20-40 mg fragments were xenografted subcutaneously either in the flank or in the interscapular area, in 3-5 immunodeficient SWISS nude mice

� Grafted mice were kept for a maximum of 4 months wi thout tumor growth.

Clinical data collectionRelevant clinical information was collected by the attending physician and included in a standardized d ata sheet.Identification of the data sheet is anonymous.

Histological characterizationSamples were fixed for a maximum of 48 hours in alc ohol-formalin-acetic acid (AFA) and embedded in par affin. 5 µm sections were stained with hematoxylin-eosin-saffron.

Molecular characterization – sequencingCGH array analysis: Evaluation of genome-wide, gene copy number was eva luated using a 244k CGH array Agilent technology. It was carried out on DNA from the patient sample o r at an early passages (P0/P1), and at the passage u sed for the pharmacological evaluation (P8-9).

DNA sequencing: The following genetic markers, relevant in CRC, wer e selected for sequencing: APC (exons 9 & 16), KRAS(exons 2 & 3), BRAF (exons 11 & 15), TP53 (exons 2 to 11), CTNNB1 (exon 3), PIK3CA (exons 10 and 21), FBXW7 (exons 4 to 11), EGFR (exons 18 to 21) and AKT1 (exon 4). Sanger direct sequencing was performed a fter PCR amplification of exons of genes harboring hotspot mutations as described in Catalog ue Of Somatic Mutations In Cancer (COSMIC: http://www.sanger.ac.uk/genetics/CGP/cosmic/ ).

Determination of Microsatellite Instability (MSI) status: The MSI status was determined according to the Cons ensus Conference (or Revised Bethesda Guidelines) ( J. Natl Cancer Inst 2004, 96, 261-268) recommendations using the follow ing five quasimonomorphic markers: NR21, BAT26, BAT25, NR24 and NR22.

Pharmacological characterizationTested agents: 5-fluorouracil (5-FU), oxaliplatin (L-OHP), irinotec an (CPT-11) or cetuximab, were tested.

In vivo determination of antitumor activity: Tumor fragments were subcutaneously xenografted in SCID mice or nude rats. Tumor-bearing animals were randomized when the mean tumor volume reached 100-200 mm 3 in mice or 500-700 mm 3 in rats. Compounds were formulated in glucose 5% in water. 3 cytotoxic drugs were IV administered, and tested at 70% of their respective highest non toxic dose and using the fol lowing regimen in mice: 5-FU at 56 mg/kg/adm Q4Dx2 (30 mg/kg/adm Q7Dx3 in rats) , L-OHP at 5 mg/kg/adm Q4Dx2 (4 mg/kg/adm Q4Dx3 in rats), CPT-11 at 22 mg/kg/adm, Q2Dx3 (40 mg/ kg/adm, Q7Dx3in rats) ; 1 targeting therapy was IP administrated: c etuximab at 12.5 mg/kg/adm, (Q3Dx2)x2 (10 mg/kg/adm, IV, Q7Dx3 in rats).Antitumor activity was evaluated by calculating the ∆(T/C) ratio:

∆∆∆∆(T/C) (%) = X 100median T VOL- DY – median T VOL-DX

median C VOL- DY – median C VOL-DX

Preservation of the tumor phenotype and genotype

Clinical characteristics and in vivo tumorgraft take rate

Scoring criteria: - = ∆(T/C) > 42 %+ = 10 < ∆(T/C) ≦ 42 %++ = 0 ≦ ∆(T/C) ≦10 % (stable disease) +++ = ∆(T/C) <0 % (tumor regression)

84125CPT-11

73415Cetuximab

00227L-OHP

051775-FU

++++++-Activity score

22

4

4<10%

10-50%

>50%

Mucus Secretion

22

4

4<10%

10-50%

>50%

Mucus Secretion

Tum

orM

odel

s Differentiation2

1213

2 1WDWD, MDWD, MD, PD

MD, PDPD, UD

Differentiation2

1213

2 1WDWD, MDWD, MD, PD

MD, PDPD, UD

Mutations 1000KRAS/BRAF

++++++-Activity score

41311EGFR mutated pathway

3213EGFR wild-type pathway*

0002BRAF

0021KRAS/PIK3CA

3118KRAS

Fresh

tumors

Surgery

* Patient identification

* Patient clinical history

* Patient informed consent

* Negative HIV, HBV, HCV serology

Paraffin

Tumor bank

Snap-frozen

Database

Tumor growth

Molecular

Histological

Pharmacological

Characterization

26 31 36 41 46 510

200

400

600

800

1000

1200

1400 CPT-11 treatmentCPT-11 22 mg/kg IVControl (untreated)

Days after tumor inoculation

Med

ian

tum

or s

ize

(mg

+/-

IQR

)

Tumor

grafting

P0P2 – P6

Establishment of in vivo tumorgraft models

RPMI/DMSO freezing

Fresh

tumors

Surgery

Fresh

tumors

Surgery

* Patient identification

* Patient clinical history

* Patient informed consent

* Negative HIV, HBV, HCV serology

* Patient identification

* Patient clinical history

* Patient informed consent

* Negative HIV, HBV, HCV serology

Paraffin

Tumor bank

Snap-frozenParaffin

Tumor bank

Snap-frozen

Database

Tumor growth

Molecular

Histological

Pharmacological

Characterization

26 31 36 41 46 510

200

400

600

800

1000

1200

1400 CPT-11 treatmentCPT-11 22 mg/kg IVControl (untreated)

Days after tumor inoculation

Med

ian

tum

or s

ize

(mg

+/-

IQR

)

Database

Database

Tumor growth

Molecular

Histological

Pharmacological

Characterization

Tumor growth

Molecular

Histological

Pharmacological

Characterization

26 31 36 41 46 510

200

400

600

800

1000

1200

1400 CPT-11 treatmentCPT-11 22 mg/kg IVControl (untreated)

Days after tumor inoculation

Med

ian

tum

or s

ize

(mg

+/-

IQR

)

26 31 36 41 46 510

200

400

600

800

1000

1200

1400 CPT-11 treatmentCPT-11 22 mg/kg IVControl (untreated)

CPT-11 treatmentCPT-11 22 mg/kg IVControl (untreated)

Days after tumor inoculation

Med

ian

tum

or s

ize

(mg

+/-

IQR

)

Tumor

grafting

P0P2 – P6

Establishment of in vivo tumorgraft models

RPMI/DMSO freezing

Tumor

grafting

P0P2 – P6

Establishment of in vivo tumorgraft models

RPMI/DMSO freezing

PR: Partial ResponseCR: Complete ResponseTFS: Tumor Free Survival

Tumor takerate (%)

Parameter Class Number Frequency(%)

Site of origin Primary 37 66.1 62.7Peritonealcarcinosis 5 8.9 62.5

Metastasis# 14 25.0 73.7

Sex Female 35 62.5 66.0Male 21 37.5 63.6

Age 0−50y 11 20.4 78.6[ 50y ; 60y] 12 24.5 50.0[ 60y ; 70y] 12 20.4 60.0[ 70y ; 80y] 15 24.5 68.2

>80y 6 10.2 100Stage 0 - I 2 3.7 22.2

II 8 14.8 57.1III 13 24.1 92.9IV 31 57.4 70.4

Patient characteristics in the collection

# hepatic, splenic and mesenteric lymph node metastasis

Tumor takerate (%)

Parameter Class Number Frequency(%)

Site of origin Primary 37 66.1 62.7Peritonealcarcinosis 5 8.9 62.5

Metastasis# 14 25.0 73.7

Sex Female 35 62.5 66.0Male 21 37.5 63.6

Age 0−50y 11 20.4 78.6[ 50y ; 60y] 12 24.5 50.0[ 60y ; 70y] 12 20.4 60.0[ 70y ; 80y] 15 24.5 68.2

>80y 6 10.2 100Stage 0 - I 2 3.7 22.2

II 8 14.8 57.1III 13 24.1 92.9IV 31 57.4 70.4

Patient characteristics in the collection

# hepatic, splenic and mesenteric lymph node metastasis Significant correlation* between CEA serum level of patient and tumor take of all types

Significant distribution* of primary tumors in rega rds of lymph node status (panel A, N1 = 1 to 6, N2 = 7 to 15 positive regional lymph nodes), stages (panel B) an d differentiation status (panel C).No other significant correlation were found among t he following parameters: gender, age, resection ext ent, lymphatic embolies, perinervous invasion, initial tr eatment, genotype

* p values were calculated with logical regression test

Gene mutation profile and response to chemotherapy

Histopathological analyses completed for 30 colon tumor models were in concordance to those observed in the corresponding patient’s tumor.CGH analysis showed very similar profile between early and advance passagesThe expression and localization of ββββ-catenin, CEA, CAIX, EGFR, CD105 and LYVE-1 is ongoing.

The majority of tumor models were non-mucinous adenocarcinomas

Splenicmetastasis

(1/86)

Mesentericlymph node

(1/86)

All models collected (86)

Left (21/59)

Right (20/59)

Primary tumor(59/86)Carcinosis (8/86)

HepaticMetastasis (16/86)

Ovarianmetastasis

(1/86)

Left (4/6)

Rect.(18/59)Left (5/8)

Right (3/8)

Left (9/16)

Right (5/16)

Rect.(2/16)

Splenicmetastasis

(1/86)

Mesentericlymph node

(1/86)

All models collected (86)

Left (21/59)

Right (20/59)

Primary tumor(59/86)Carcinosis (8/86)

HepaticMetastasis (16/86)

Ovarianmetastasis

(1/86)

Left (4/6)

Rect.(18/59)Left (5/8)

Right (3/8)

Left (9/16)

Right (5/16)

Rect.(2/16)

Right colon

Left colon

Rectum

Models growing on nude mice (56)

Primary tumor(37/56)Carcinosis (5/56)

HepaticMetastasis (12/56)

Splenicmetastasis

(1/56)

Mesentericlymph node

(1/56)

Left (15/37)

Right (13/37)Rect.(9/37)

Left (4/5)

Right (1/5 )

Left (6/12) Right (5/12)

Rect. Right colon

Left colon

Rectum

Right colon

Left colon

Rectum

Models growing on nude mice (56)

Primary tumor(37/56)Carcinosis (5/56)

HepaticMetastasis (12/56)

Splenicmetastasis

(1/56)

Mesentericlymph node

(1/56)

Left (15/37)

Right (13/37)Rect.(9/37)

Left (4/5)

Right (1/5 )

Left (6/12) Right (5/12)

Rect.

Only 1 model showed a poor differentiated (PD) to undifferentiated (UD) characteristic, as well as its correspondent patient

Similar CGH profiles between early and advance passage s

Loss in chromosomes 3p and 4q in advance passage

Most models with EGFR mutated pathway are resistant to Cetuximab, nevertheless several Cetuximab sensitive KRAS-mutated models have been identified.

Patient: CR-IGR-034PPatient: CR-LRB-010P Patient: CR-LRB-008M

P10: CR-LRB-010P P11: CR-LRB-008M P11: CR-IGR-034P

Pat

ient

Tum

orM

odel

Well DifferentiatedWell to Moderate

Differentiated Poor to UnDifferentiated

Patient: CR-IGR-034PPatient: CR-LRB-010P Patient: CR-LRB-008M

P10: CR-LRB-010P P11: CR-LRB-008M P11: CR-IGR-034P

Patient: CR-IGR-034PPatient: CR-LRB-010P Patient: CR-LRB-008M Patient: CR-IGR-034PPatient: CR-IGR-034PPatient: CR-LRB-010PPatient: CR-LRB-010P Patient: CR-LRB-008MPatient: CR-LRB-008M

P10: CR-LRB-010P P11: CR-LRB-008M P11: CR-IGR-034PP10: CR-LRB-010PP10: CR-LRB-010P P11: CR-LRB-008MP11: CR-LRB-008M P11: CR-IGR-034PP11: CR-IGR-034P

Pat

ient

Tum

orM

odel

Pat

ient

Tum

orM

odel

Well DifferentiatedWell to Moderate

Differentiated Poor to UnDifferentiated

Sensitivity to Cetuximab versus mutated genes involved in EGFR pathway ∆T/C = -41% (Day 31),

9/10 PR & 4/10 CR

CRIGR023MGAIN

LOSS

CRIGR002CGAIN

LOSS

* Mutations of PTEN or other genes related to EGFR pathway have not been yet characterized

Multiple mutation profiles are observed in our tumo r panel (n=60), matching the human tumor genetic heterogeneity Pharmacological studies exhibit diversity in the response to chemotherapy13/29 tumorgrafts show tumor regression after treat ment with at least one standard monotherapy

Diversity of colorectal cancers is fully addressed in this collection of patient-derived tumor models.

Genotype and phenotype are largely preserved throughout the model establishment process.

Difference of gene mutation and drug sensitivity profiles are observed between the models.

Plan to complete the full correlation analysis between clinical data, gene mutations, transcriptome

profile, ex-vivo and in vivo drug sensitivity.

Perspectives to fully exploit this new collection for new drug candidate selection.

∆T/C = < 0 % (Day 36), 10/10 CR + 100% TFS

0

CE

A (

ng/m

L)

Positive xenograft take

5

10

15

2025

30

35

40

4550

170175180

41

11

2.3

8

27

p = 0.0292 (*)

All types of tumor samples / CEA dosing

Negativexenograft take

0

5

10

15

2025

30

35

40

4550

170

180

41

11

2.3

8

27

p = 0.0292 (*)

0

CE

A (

ng/m

L)

Positive xenograft take

5

10

15

2025

30

35

40

4550

170175180

41

11

2.3

8

27

p = 0.0292 (*)

All types of tumor samples / CEA dosing

Negativexenograft take

0

5

10

15

2025

30

35

40

4550

170

180

41

11

2.3

8

27

p = 0.0292 (*)

AP

erce

ntag

e(%

)

N0

N0

N1 + N2

N1 + N2

0

20

40

60

80

100

120

(n=11)

(n=26)

(n=5)

(n=17)

Primary tumor samples / Lymph node status

Positive xenograft take

Negativexenograft take

p = 0.0038 (**)

N0

N0

N1 + N2

N1 + N2

0

20

40

60

80

100

120

(n=11)

(n=26)

(n=5)

(n=17)

p = 0.0038 (**)

AP

erce

ntag

e(%

)

N0

N0

N1 + N2

N1 + N2

0

20

40

60

80

100

120

(n=11)

(n=26)

(n=5)

(n=17)

Primary tumor samples / Lymph node status

Positive xenograft take

Negativexenograft take

p = 0.0038 (**)

N0

N0

N1 + N2

N1 + N2

0

20

40

60

80

100

120

(n=11)

(n=26)

(n=5)

(n=17)

p = 0.0038 (**)

B

Stages 0-I

Stages 0-I

Stage II

Stage II

Stage III

Stage III

Stage IV Stage IV

0

20

40

60

80

100

120

(n=7)

(n=6)

(n=7) (n=6)

(n=13)

(n=14)

Primary tumor samples / Stages

Negativexenograft take

Positive xenograft take

p = 0.0101 (*)

Per

cent

age

(%)

Stages 0-I

Stages 0-I

Stage II

Stage II

Stage III

Stage III

Stage IV Stage IV

0

20

40

60

80

100

120

(n=7)

(n=6)

(n=7) (n=6)

(n=13)

(n=14)

p = 0.0101 (*)

B

Stages 0-I

Stages 0-I

Stage II

Stage II

Stage III

Stage III

Stage IV Stage IV

0

20

40

60

80

100

120

(n=7)

(n=6)

(n=7) (n=6)

(n=13)

(n=14)

Primary tumor samples / Stages

Negativexenograft take

Positive xenograft take

p = 0.0101 (*)

Per

cent

age

(%)

Stages 0-I

Stages 0-I

Stage II

Stage II

Stage III

Stage III

Stage IV Stage IV

0

20

40

60

80

100

120

(n=7)

(n=6)

(n=7) (n=6)

(n=13)

(n=14)

p = 0.0101 (*)

C Primary tumor samples / Differentiation

0

Well

Well

Fairly

Fairly

Weakly

20

40

60

80

100

120

(n=10)

(n=11)

(n=9)

(n=25)

Per

cent

age

(%)

p = 0.0287 (*)

Negativexenograft take

Positive xenograft take

0

Well

Well

Fairly

Fairly

Weakly

20

40

60

80

100

120

(n=10)

(n=11)

(n=9)

(n=25)

p = 0.0287 (*)

C Primary tumor samples / Differentiation

0

Well

Well

Fairly

Fairly

Weakly

20

40

60

80

100

120

(n=10)

(n=11)

(n=9)

(n=25)

Per

cent

age

(%)

p = 0.0287 (*)

Negativexenograft take

Positive xenograft take

0

Well

Well

Fairly

Fairly

Weakly

20

40

60

80

100

120

(n=10)

(n=11)

(n=9)

(n=25)

p = 0.0287 (*)

Similar tumor response ranges have been observed in mice and rats

∆∆∆∆T/C in miceL-OHP: 45%5-FU: 45%

Cetuximab: 78%

∆∆∆∆T/C in ratL-OHP: 104%

5-FU: 63%Cetuximab: 19%

CPT-11: 1%

∆∆∆∆T/C in miceCPT-11 < 0%

70%

80%

80%

50%

70%

90%

60%

80%

80%

50%

90%

90%

70%

60%

90%

60%

40%

100%

40%

70%

40%

80%

50%

80%

50%

80%

80%

80%

80%

50%

90%

40%

80%

30%

90%

80%

90%

90%

85%

60%

85%

50%

60%

70%

40%

40%

80%

50%

30%

50%

70%

90%

70%

80%

80%

50%

50%

50%

50%

80%

CR

IGR

023M

CR

IC00

4M

CR

IC00

7M

CR

IGR

007P

CR

IGR

025P

CR

LRB

010P

CR

LRB

011M

CR

LRB

013P

CR

LRB

014P

CR

IC01

8P

CR

IC00

2P

CR

IC00

3P

CR

IC02

5M

CR

IGR

004P

CR

IGR

008P

CR

LRB

007P

CR

IGR

015P

CR

LRB

019C

CR

IGR

021P

CR

IC02

0P

CR

IC02

1M

CR

IGR

032P

CR

IGR

029P

CR

IGR

012P

CR

IGR

014P

CR

IC01

2P

CR

IC01

3M

CR

LRB

008M

CR

LRB

009C

CR

IGR

003P

CR

IGR

009P

CR

IGR

016P

CR

IGR

020P

CR

IGR

001M

CR

IGR

038C

CR

LRB

017P

CR

IGR

034P

CR

LRB

018P

CR

LRB

003P

CR

LRB

004P

CR

LRB

015P

CR

IC00

6M

CR

IC01

9P

CR

IC02

9P

CR

IC02

8M

CR

IGR

002P

CR

IGR

002C

CR

IGR

052M

CR

LRB

005P

CR

IGR

047P

CR

IGR

048M

CR

LRB

022P

CR

IC00

8P

CR

IC00

9M

CR

IC01

0P

CR

IGR

011C

CR

IGR

039P

CR

IGR

043P

CR

IC00

5P

CR

IC01

4P

TP53 62%1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 0 0 0 0 0 0 0 0 0 0 1 1 1 1 1 1 1 1 1 0 0 0 1 1 1 1 0 1 1 1 0 1 1 0 0 0 0 0 0 0 0

APC 53%1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 0 1 1 1 1 1 1 1 1 0 0 0 0 0 0 0 0 0 0 0 1 1 1 0 0 0 0 0 0 0 0 0 0 0 1 1 1 0 0 0 0 0

KRAS 45%1 1 1 1 1 1 1 1 1 1 0 0 0 0 0 0 0 0 0 0 0 1 1 1 1 0 1 1 1 1 1 1 1 1 1 1 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 1 1 1 0 0

PIK3CA 10%1 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 1 1 0 0 0 0 1 1 0 0 0 0 0 0 0 0 0 0 0 1 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0

FBXW7 8%0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 1 1 1 1 0 1 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0

BRAF 5%0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 * 0 0 0 0 0 0 0 0 0 1 1 0 1 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0

CTNNB1 3%0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 1 1 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0

EGFR 2%0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 1 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0

AKT1 0%0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0

MSI status MSS MSS MSS MSS MSS MSS MSS MSS MSS MSS MSS MSS MSS MSI-L MSI-L MSS MSS MSS MSS MSS MSS MSS MSS MSS MSI-L MSS MSS MSS MSI-L MSS MSS MSS MSS MSI-L MSI-H MSI-H MSS MSI-L MSI-L MSS MSS MSI-L MSI-L MSI-L MSS MSS MSS MSS MSS MSS MSS MSS MSS MSS

Xenograft 1 1 1 1 1 1 1 1 1 1 1 1 1 0 1 1 0 1 0 1 1 1 1 1 1 0 1 1 1 1 1 1 0 0 1 1 1 1 1 1 1 1 1 1 1 1 1 1 0 1 1 1 1 1 1 1 1 1 1 0

CGH Analysis 0 1 1 0 0 1 0 0 0 0 0 1 0 1 1 0 0 0 0 0 0 1 0 1 1 0 0 0 1 0 0 1 1 0 0 0 1 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 1 80 0 0 0 1 0

Other ERCC1-A138T FCGR2A-F196Y * BRAF-N594D

5-FU + - - - - + + + ++ + + ++ + + + + + ++ + - + ++ + ++ + + - + -

l-OHP - - - - - - - - - - - - + - - - + - - - - - - - - - - - -

CPT-11 + + ++ + ++ +++ - +++ - + - - +++ - + ++ + +++ + + + + +++ ++ + +++ +++ + +++

CETUXIMAB + ++ +++ + - - +++ +++ +++ - - - +++ + - +++ - - - - - - ++ ++ +++ + - - -

Mutation MSI-L Low (MSI-L) or High (MSI-H) microsatellite instability Wild-type No data available Model from the same patient Xenograft model established CGH analysis % above sample name: % of tumor cells in the analyzed sample

Scoring criteria: - = ∆(T/C) > 42 % + = 10 < ∆(T/C) ≦ 42 % ++ = 0 ≦ ∆(T/C) ≦10 % (stable desease) +++ = ∆(T/C) <0 % (tumor regression) Characterization in progress % beside gene name: % of mutations among all sequen ced samples