Development of a living organoid biobank derived from colorectal cancer patients: Towards personalized medicine

Development of a living organoid biobank derived from colorectal cancer patients: Towards personalized medicine

abstracts Conclusions: Our results provide the first evidence that CASC21 is a novel oncogenic regulator and a potential therapeutic target in colon c...

90KB Sizes 1 Downloads 42 Views

abstracts Conclusions: Our results provide the first evidence that CASC21 is a novel oncogenic regulator and a potential therapeutic target in colon cancer. Legal entity responsible for the study: Xiaoping Qian. Funding: Has not received any funding. Disclosure: All authors have declared no conflicts of interest.

636P

Prognostic and predictive impact on FMS-like tyrosine kinase 3 (FLT3) amplification in patients with metastatic colorectal cancer

Background: FMS-like tyrosine kinase 3 (FLT3) plays a key role in hematopoiesis, and FLT3-associated molecular alterations are an established predictor for the treatment with FLT3 inhibitors in acute myeloid leukemia. However, the oncogenic role of FLT3 amplification (amp) in patients (pts) with metastatic colorectal cancer (mCRC) has not yet been well established. Methods: Tumor tissue samples from 2,329 mCRC pts were sequenced using next-generation sequencing (NGS) with Oncomine Comprehensive Assay in the Nationwide Cancer Genome Screening Project in Japan (SCRUM-Japan GI-SCREEN). Clinicopathological features, co-altered genes, prognosis and regorafenib efficacy on FLT3 amp (defined as copy number  7.0) vs. non-FLT3 amp mCRC were investigated. Results: Between Apr 2015 and Jun 2018, a total of 85 pts (3.6%) with mCRC with FLT3 amp were observed. There were no clear differences in baseline characteristics between pts with or without FLT3 amp. The enrichment of TP53 mutation in FLT3 amp mCRC was observed more frequently than non-FLT3 amp (74.1% vs. 64.7%, P ¼ 0.08), but RAS mutation frequency was similar in both (48.2% vs. 42.8%, P ¼ 0.31). In contrast, activating alterations in BRAF (1.1% vs. 7.1%, P ¼ 0.053) and PIK3CA (1.1% vs 7.0%, P ¼ 0.03) mutations were both less frequent in FLT3 amp vs. non-FLT3 amp mCRC. Median OS from 1st-line chemotherapy in FLT3 amp mCRC was significantly shorter than those in non-FLT3 amp (30.2 vs. 43.4 months, P ¼ 0.002). Furthermore, in 20 pts receiving regorafenib, a multikinase inhibitor with a mild inhibitory activity of FLT3, the disease control rate (DCR) was higher in FLT3 amp mCRC pts (n ¼ 7) compared with non-FLT3 amp (57.1% vs. 23.0 %, P ¼ 0.13). Conclusions: FLT3 amp was associated with a significantly worse survival and a higher DCR from regorafenib, suggesting it has a distinct oncogenic role in mCRC. Further investigation of the oncogenic role of FLT3 amp in mCRC is warranted in clinical trials. Clinical trial identification: UMIN000016344. Legal entity responsible for the study: SCRUM-Japan. Funding: 17 SCRUM-Japan Collaborating Pharmaceutical Companies, AMED, NCC. Disclosure: H. Taniguchi: Research grant / Funding (self): Takeda; Advisory / Consultancy: Chugai; Advisory / Consultancy: Taiho. T. Kato: Honoraria (self): Chugai Pharmaceutical; Advisory / Consultancy: Takeda; Advisory / Consultancy: Eli Lilly; Advisory / Consultancy: Bayer; Advisory / Consultancy: Sanofi; Advisory / Consultancy: Yakult Honsya. S. Yuki: Speaker Bureau / Expert testimony: Chugai Pharma; Speaker Bureau / Expert testimony: Bristol-Myers Squibb; Speaker Bureau / Expert testimony: Bayer Yakuhin; Speaker Bureau / Expert testimony: Ono Pharmaceutical; Speaker Bureau / Expert testimony: Pharma International; Speaker Bureau / Expert testimony: Daiichi Sankyo; Speaker Bureau / Expert testimony: Takeda Pharmaceutical; Speaker Bureau / Expert testimony: Eli Lilly Japan; Speaker Bureau / Expert testimony: Taiho Pharmaceutical; Speaker Bureau / Expert testimony: Sanofi; Speaker Bureau / Expert testimony: Yakult Honsha; Speaker Bureau / Expert testimony: Merck Biopharma. T. Masuishi: Honoraria (self): Eli Lilly; Honoraria (self): Chugai Pharma; Honoraria (self): Merck Serono; Honoraria (self): Taiho Pharmaceutical; Honoraria (self): Yakult Honsya. K. Kato: Research grant / Funding (self): Shionogi; Research grant / Funding (self): Ono Pharmaceutical; Research grant / Funding (self): Merck Serono. N. Izawa: Advisory / Consultancy: Taiho Pharmaceutical; Advisory / Consultancy: Sanofi; Advisory / Consultancy: MSD; Advisory / Consultancy: Bayer; Advisory / Consultancy: Bristol-Myers; Advisory / Consultancy: Lilly; Advisory / Consultancy: Merck Serono. T. Moriwaki: Speaker Bureau / Expert testimony, Research grant / Funding (institution): Taiho Pharmaceutical; Research grant / Funding (institution): MSD; Speaker Bureau / Expert testimony, Research grant / Funding (institution): Takeda; Research grant / Funding (institution): Yakult Honsha; Research grant / Funding (institution): Eisai; Speaker Bureau / Expert testimony: Chugai Pharma; Speaker Bureau / Expert testimony: Yakult Honsha; Speaker Bureau / Expert testimony: Merck Serono; Speaker Bureau / Expert testimony: Sanofi; Speaker Bureau / Expert testimony: Liliy Japan; Speaker Bureau / Expert testimony: Bayer; Speaker Bureau / Expert testimony: Ono Pharmaceutical. Y. Kagawa: Speaker Bureau / Expert testimony: Chugai; Speaker Bureau / Expert

v240 | Gastrointestinal Tumours, Colorectal

testimony: Eli Lilly; Advisory / Consultancy: Sanofi; Speaker Bureau / Expert testimony: Taiho. W. Okamoto: Research grant / Funding (institution): MSD. Y. Nakamura: Research grant / Funding (institution): Ono Pharmaceutical; Research grant / Funding (institution): Taiho Pharmaceutical. K. Yamazaki: Honoraria (self): Daiichi Sankyo; Honoraria (self): Chugai Pharmaceutical; Honoraria (self): Eli Lilly; Honoraria (self): Merck Serono; Honoraria (self), Research grant / Funding (institution): Taiho Pharma; Honoraria (self): Bayer Yakuhin. T. Yoshino: Research grant / Funding (institution): Chugai pharma; Research grant / Funding (institution): Sanofi; Research grant / Funding (institution): Sumitomo Dainippon pharma; Research grant / Funding (institution): Glaxo SmithKline. All other authors have declared no conflicts of interest.

637P

RBP-Jj in colon cancer cells facilitates tumour associated macrophages (TAMs)-induced cell metastasis by secreting CXCL11

M.J. Liu, X. Fu, S. Wang, L. Jiang, K. Nan, W. Wang Medical Oncology, First Affiliated Hospital of Xi’an Jiaotong University (School of Medicine), Xi’an, China Background: Metastasis is the main cause of death in colon cancer patients. RBP-Jj, which is the main transcription mediator of Notch signaling, is involved in colon cancer development but its function in colon cancer metastasis is still unclear. Here we aimed to find out the function of RBP-Jj in colon cancer metastasis and its underlying mechanisms of modulating the interaction between colon cancer cells and tumorassociated macrophages (TAMs). Methods: Cell migration, invasion and epithelial to mesenchymal transition (EMT) were used to reflect cell metastasis ability. A oc-culture system was adopted to research the mutual regulation between colon cancer cells and TAMs. Gain- and loss-of-function experiments and TGF-b/Smad3 pathway activator and inhibitor were used to determine the underlying mechanisms of RBP-Jj and TAMs in regulating colon cancer metastasis in vitro and in vivo. RNA sequencing was performed to find out the regulating factor between colon cancer cell and TAMs. RBP-Jj and E-cadherin expression and TAMs infiltration were examined in 201 colon cancer patients by immunohistochemical assay and were analyzed combined with clinical parameters. Results: RBP-Jj and TAMs promoted colon cancer cell metastasis through the TGF-b/ Smad3 pathway and secreting of TGF-b, respectively. RBP-Jj in colon cancer cell facilitated TAMs to secret TGF-b through secreting CXCL11. RBP-Jj and E-cadherin was highly expressed in colon cancer tissues and para-tumor tissues, respectively. And there are more TAMs infiltrated in colon cancer tissues. RBP-Jj expression and TAMs infiltration were negatively associated with E-cadherin expression and overall survival and positively associated with metastasis. RBP-Jj and E-cadherin expression and TAMs infiltration were independent prognostic factors in colon cancer patients. Conclusions: Our research demonstrated that colon cancer cells with high RBP-Jj expression secreted CXCL11 to enhance TGF-b secretion of TAMs which facilitated colon cancer metastasis. RBP-Jj exrpression and TAMs infiltration were associated with colon cancer metastasis and acted as prognostic factors. Legal entity responsible for the study: Wenjuan Wang. Funding: National Natural Science Foundation of China (No. 81502099); Key laboratory of tumor precision medicine open project of Shaanxi province (No. KLTPMSX2018-B3); The scientific research fund of the first affiliated hospital of Xi’an Jiaotong University (2018MS-06). Disclosure: All authors have declared no conflicts of interest.

638P

Development of a living organoid biobank derived from colorectal cancer patients: Towards personalized medicine

F. Papaccio1, M.F. Gutierrez-Bravo1, M. Cabeza-Segura2, V. Gambardella3, M. Huerta3, C. Martinez Ciarpaglini4, S. Rosello Keranen1, F. Gimeno-Valiente1, T. Fleitas1, N. Tarazona Llavero5, D. Roda5, A. Cervantes5, J. Castillo2 1 Medical Oncology, INCLIVA Biomedical Research Institute, Valencia, Spain, 2 Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain, 3Medical Oncology, Hospital Clinico Universitario de Valencia, Valencia, Spain, 4Pathology, INCLIVA Instituto de Investigaci on Sanitaria, Valencia, Spain,5Medical Oncology Department, Biomedical Research Institute INCLIVA, Hospital Clınico Vale`ncia, University of Valencia, Valencia, Spain, Background: Organoids are 3D in vitroprimary culture of great interest for translational research representing an efficient and reproducible cancer model. The aim of this project is to generate a biobank of colorectal cancer (CRC) patients derived organoids (PDOs) that could be used to analyze molecular characteristics and to test different treatments as well as to study the underlying molecular causes of cancer and treatment resistance. Methods: Primary or metastatic CRC tissues have been obtained from patients who underwent surgery. Tissue has been washed and incubated with antibiotics. After mechanical and enzymatic digestion, free cells have been seeded in Matrigel with proper medium. Development of organoids has been observed the day after seeding. Organoids have been passaged, expanded and stored in liquid nitrogen to constitute a biobank. For morphological characterization, after reaching an appropriate volume, organoids have been fixed in 4% paraformaldehyde, stained for hematoxylin and eosin (H&E) and immunohistochemistry (IHC) for ki67, CDX2, CK20, MUC2/5 was performed. Immunofluorescence (IF) for DAPI, Lgr5 and CDH1 has been performed.For genomic characterization, DNA has been extracted with phenol-chlorophorm and

Volume 30 | Supplement 5 | October 2019

Downloaded from https://academic.oup.com/annonc/article-abstract/30/Supplement_5/mdz246.115/5577970 by guest on 24 October 2019

H. Hasegawa1, H. Taniguchi2, T. Kato3, S. Fujii4, H. Ebi5, M. Shiozawa6, S. Yuki7, T. Masuishi8, K. Kato9, N. Izawa10, T. Moriwaki11, Y. Kagawa12, Y. Sakamoto13, W. Okamoto13, Y. Nakamura14, K. Yamazaki15, T. Yoshino14 1 Gastroenterology and Hepatology, National Hospital Organization Osaka National Hospital, Osaka, Japan, 2Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan, 3Surgery, Osaka National Hospital, Osaka, Japan, 4Division of Pathology, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan, 5Molecular Therapeutics, Aichi Cancer Center Research Institute, Nagoya, Japan, 6Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Japan, 7Gastroenterology and Hepatology, Hokkaido University Hospital, Sapporo, Japan, 8Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan, 9 Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan, 10Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan, 11Gastroenterology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan, 12 Surgery, Kansai Rosai Hospital, Amagasaki, Japan, 13Clinical research Support Office, Biobank Translational Research Support Section, National Cancer Center Hospital East, Kashiwa, Japan, 14Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan, 15Gastroenterology, Sizuoka Cancer Center, Shizuoka, Japan

Annals of Oncology

abstracts

Annals of Oncology

grant / Funding (institution): Merck Serono; Advisory / Consultancy, Speaker Bureau / Expert testimony, Research grant / Funding (institution): Roche; Advisory / Consultancy, Research grant / Funding (institution): Beigene; Advisory / Consultancy, Speaker Bureau / Expert testimony, Research grant / Funding (institution): Bayer; Advisory / Consultancy, Speaker Bureau / Expert testimony, Research grant / Funding (institution): Servier; Advisory / Consultancy, Research grant / Funding (institution): Lilly; Advisory / Consultancy, Research grant / Funding (institution): Novartis; Advisory / Consultancy, Research grant / Funding (institution): Takeda; Advisory / Consultancy, Research grant / Funding (institution): Astellas; Advisory / Consultancy: Pierre Fabre; Research grant / Funding (institution): Genentech; Research grant / Funding (institution): Fibrogen; Research grant / Funding (institution): Amcure; Research grant / Funding (institution): Sierra Oncology; Research grant / Funding (institution): AstraZeneca; Research grant / Funding (institution): Medimmune; Research grant / Funding (institution): BMS; Research grant / Funding (institution): MSD; Speaker Bureau / Expert testimony: Amgen; Speaker Bureau / Expert testimony: Foundation Medicine. All other authors have declared no conflicts of interest.

639P

Microsatellite instability detection in colorectal cancer: 44-Center comparison between the Idylla MSI assay and routine molecular and immunohistochemistry tests on formalin-fixed paraffin-embedded tissue

X. Matias-Guiu Pathology, Hospital Universitario Arnau de Vilanova, Lleida, Spain Background: Microsatellite instability (MSI) is observed in 15% of all colorectal cancers (CRC), including Lynch syndrome caused by germline mutation of an MMR gene. Guidelines recommend assessing the MSI status of all CRC patients for screening for Lynch syndrome as well as for prognostic stratification. MSI-high tumors are more vulnerable to the immune system and therefore generally have a better prognosis. Methods: The performance of the IdyllaTM MSI Assay was compared to routine immunohistochemistry (IHC) and molecular tests (including Bethesda panel) on 1,301 archival CRC formalin-fixed, paraffin-embedded (FFPE) tissue sections, at 44 centers worldwide. Results: The failure rate of the IdyllaTM MSI Assay was 0.23%, while the molecular methods had a higher failure rate overall of 0.86% probably due to their use of a longer amplicon. IHC had a much higher failure rate of 4.37%, possibly related to interpretation difficulties of the results. Routine method failure rates might be an underestimation as the current analysis was done retrospectively on samples with known routine results. Compared to IHC, the results of 37 of the 1,075 samples tested were discordant, resulting in an overall concordance agreement of 96.6%. When taking molecular results and further analysis of discordant samples into account, the IdyllaTM MSI result was confirmed in 16 samples. Compared to molecular methods, 16 of the 812 samples tested were discordant, resulting in an overall concordance agreement of 98.0%. Taking IHC results and further analysis of discordant samples into account, the IdyllaTM MSI result was confirmed in 8 samples. Conclusions: Results of the IdyllaTM MSI Assay were highly concordant with results of routine tests and lower failure rates were observed. Further advantages are the lack of need for matched normal tissue, the low dependence on pre-analytical conditions, the simplified workflow, the short turnaround times, the automated result interpretation, and the very limited hands-on work. Editorial acknowledgement: Teams involved in the MSI, Idylla MSI Assay, Colon Rectal Cancer, Multicenter Study. Legal entity responsible for the study: Irblleida. Funding: Biocartis. Disclosure: X. Matias-guiu: Non-remunerated activity/ies: Biocartis.

Volume 30 | Supplement 5 | October 2019

640P

Expression profile of EPHB3 and its prognostic significance in colorectal cancer progression (Running head: Prognostic value of EPHB3 in colorectal cancers)

B. Jang, H. Kim Pathology, Jeju National University School of Medicine, Jeju-si, Republic of Korea Background: A tumour suppressive role for EPHB receptors in various malignancies has been described, however their expression profile and prognostic significance in human colorectal cancers (CRCs) remain unclear. In this study, we aimed to investigate the expression profile of EPHB3 during CRC progression and determined its prognostic impact in a large cohort of CRC samples. Methods: We examined the EPHB3 mRNA levels by real time PCR with fresh frozen CRC samples and evaluated protein expression by immunohistochemistry with tissue microarrays containing various benign tumours and 610 CRC samples. AOM-DSS induced colitis model was used to investigate the EPHB3 expression profile during the colitis-associated carcinogenesis. Results: EPHB3 expression was upregulated in CRCs than in normal colonic mucosa, and showed positive correlations with intestinal stem cell (ISC) markers (EPHB2, OLFM4, LRIG1) and CD44, a candidate cancer stem cell marker. EPHB3 positivity was observed in 24% of 610 CRCs and showed negative correlations with differentiation, lympho-vascular invasions and TNM stages. EPHB3 expression significantly declined during adenoma-carcinoma transition and invasion into deeper layers. In particular, a substantial reduction of EPHB3 was observed in the budding cancer cells at the invasive fronts. In AOM-DSS induced colitis-associated colon cancer model, EPHB3 expression increased along with tumor development. Notably, EPHB3 was positively associated with microsatellite instability (MSI) phenotype but was not associated with CpG island methylator phenotype, KRAS and BRAF mutations. Furthermore, EPHB3 positivity was a prognostic marker for better clinical outcomes in CRC patients. Conclusions: EPHB3 was upregulated in CRCs and showed positive correlations with candidate cancer stem cell marker CD44. EPHB3 expression decreased during adenoma-carcinoma transition and particularly in the budding cancer cells at the invasive fronts. EPHB3 positivity was associated with MSI phenotype and demonstrated to be a good prognostic marker in CRCs. Legal entity responsible for the study: The authors. Funding: Has not received any funding. Disclosure: All authors have declared no conflicts of interest.

641P

A pan-ErbB family inhibitor, AF8c, promotes apoptosis by DR5/Nrf2 activation via ROS in colorectal cancer cells

S. Jeong1, H.K. Yun2, Y.A. Jeong2, D.Y. Kim2, M.J. Jo3, S.H. Park3, B.R. Kim1, J.L. Kim1, Y.J. Na3, D-H. Lee1, S.C. Oh4 1 Oncology/Hematology, Korea University Guro Hospital, Seoul, Republic of Korea, 2 Graduate School of Medicine, College of Medicine, Korea University, Seoul, Republic of Korea, 3Oncology, Korea University Guro Hospital, Seoul, Republic of Korea, 4 Department of Internal Medicine, Korea University Guro Hospital, Seoul, Republic of Korea Background: Multifunctional, antiproliferative small molecules are deemed to offer pharmacodynamic and pharmacokinetic benefits over combination therapy, in addition to reducing toxicity, cancer resistance, and therapy costs. In this study, we conducted an in vitro cellular screen of our recently reported series of EGFR/HER2 inhibitors. Methods: Cytotoxicity induced by AF8c was confirmed using WST-1 assay. An Annexin V-Fluorescein Isothiocyanate (FITC) Apoptosis Detection Kit, TUNEL assay, and Western blotting were carried out to invastigate apoptosis. A human phosphorylated kinase array was used to identify proteins differentially expressed between control cells and AF8c-treated cell lines. ROS generation was monitored by flow cytometry, confocal microscopy using dihydroethidium (DHE) and MitoSOX. For in vivo tumor xenograft study, four-week-old female BALB/c nude mice were injected subcutaneously with either HT29 Lucþ or HCT116 Lucþ cells (1  107 cells in 100 lL PBS). When the tumor had reached approximately 100 mm3 in size, the mice were randomly divided into three groups (n ¼ 8): DMSO-treated, treated with 10 mg/kg AF8c, and treated with 20 mg/kg AF8c. Results: Analysis of the AF8c mode of action in CRC cells revealed that it mediates apoptosis via the generation of endoplasmic reticulum (ER) stress and reactive oxygen species (ROS), as well as selective activation of nuclear respiratory factor 2 alpha subunit (Nrf2) and death receptor 5 (DR5), but not DR4. Silencing of DR5 attenuated the expression levels of Nrf2 and partially inhibited AF8c-induced apoptosis. Additionally, upregulation of Nrf2 by AF8c evoked apoptosis through a decrease in antioxidant levels. Treatment of mice with AF8c also resulted in the upregulation of DR5, Nrf2, and CHOP proteins, subsequently leading to a significant decrease in tumor burden.

doi:10.1093/annonc/mdz246 | v241

Downloaded from https://academic.oup.com/annonc/article-abstract/30/Supplement_5/mdz246.115/5577970 by guest on 24 October 2019

sequenced with an internal customized panel. A comparison statistical test with matched tissue has been performed. Results: The success rate in CRC-PDOs establishment is around 80%. PDOs staining with H&E shows a morphology comparable to the original tissue. IHC shows ki67 staining, nuclear positivity for CDX2, cytoplasmic CK20 and MUC2/5. IF evidences the positivity for the intestinal stem cells marker Lgr5 and CDH1 thus confirms that PDOs faithfully recapitulate original tissue morphology and cell composition.Between 80 and 120 ng of genomic DNA has been sequenced. Spectrum of PDOs mutations and polimorphisms displays a good concordance with that of matched patients with an R of linear concordance of 0.9. Conclusions: We have developed a robust protocol for efficient development of CRC PDOs. Organoids recapitulate morphological and genomic features of the original patient. They work as a solid in vitromodel, suitable for functional studies and drug screening, helping in promoting precision medicine. Legal entity responsible for the study: The authors. Funding: Grants from the Instituto de Salud Carlos III (PI15/02180 to AC and PI18/ 01508 to TF). FP is supported by the ESMO 2018 fellowship programme. VG was supported by the ESMO 2014 fellowship programme and by Rio Ortega contract CM18/ 00241 from the Carlos III Health Institute; TF is supported by Joan Rodes contract 17/ 00026 from the Carlos III Health Institute. NT was supported by Rio Ortega contract CM15/00246 from the Carlos III Health Institute; DR was supported by Joan Rodes contract 16/00040 from the Carlos III Health Institute. MFGB is supported by a Santiago Grisolia fellowship. Disclosure: A. Cervantes: Advisory / Consultancy, Speaker Bureau / Expert testimony, Research