Use of zebrafish embryos as avatar of patients with pancreatic cancer: A new xenotransplantation model towards personalized medicine

Gregorio Di Franco, Alice Usai, Niccola Funel, Matteo Palmeri, Ida Elena Rosamaria Montesanti, Matteo Bianchini, Desirée Gianardi, Niccolò Furbetta, Simone Guadagni, Enrico Vasile, Alfredo Falcone, Luca Emanuele Pollina, Vittoria Raffa, Luca Morelli, Gregorio Di Franco, Alice Usai, Niccola Funel, Matteo Palmeri, Ida Elena Rosamaria Montesanti, Matteo Bianchini, Desirée Gianardi, Niccolò Furbetta, Simone Guadagni, Enrico Vasile, Alfredo Falcone, Luca Emanuele Pollina, Vittoria Raffa, Luca Morelli

Abstract

Background: The response to chemotherapy treatment of patients with pancreatic ductal adenocarcinoma (PDAC) is difficult to predict and the identification of patients who most likely will benefit from aggressive chemotherapy approaches is crucial. The concept of personalized medicine has emerged in the last years with the objective to tailor the medical treatment to the individual characteristics of each patient, and particularly to the tumor biology of each patient. The need for in-vivo xenotransplantation models for cancer patients has increased exponentially, and for this reason zebrafish avatars have gained popularity. Preliminary studies were conducted also with PDAC tissue.

Aim: To develop a simple, not expensive, diffusible zebrafish embryo model as avatar for patients affected by PDAC.

Methods: Tumor tissue was taken from the surgical specimen by the histopathologist. After its fragmentation into small pieces, they are stained with CM-Dil. Small pieces of stained tissue were transplanted into the yolk of wt AB zebrafish embryos with a glass capillary needle. Embryos were incubated at 35 °C in E3 medium supplemented with 1% Pen/Strep in the presence or absence of drugs for the following days in respect of the treatment plan (Gemcitabine; Gemcitabine and Oxaliplatin; Gemcitabine and nab-Paclitaxel; 5-Fluorouracil and Folinic acid and Oxaliplatin and Irinotecan). The response of zebrafish xenografts to the chemotherapy options has been analyzed by monitoring the fluorescent stained area at 2 h post injection (hpi), 1 d and 2 d post injection (dpi). In each time point, the mean size of the stained area was measured by ImageJ and it was normalized with respect to the 1 dpi time point mean relative tumor area (RTA). We evaluated the effect of the chemotherapy exposition comparing the mean RTA of each treated subgroup and the control group and evaluating the percentage reduction of the mean RTA by comparing each treated subgroup with the control group.

Results: Between July 2018 and October 2019, a total of 15 patients with pancreatic cancer were prospectively enrolled. In all cases, it was possible to take a fragment of the tumor from the surgical specimen for the xenotransplantation in the zebrafish embryos. The histological examination confirmed the presence of a PDAC in all cases. In absence of chemotherapy (control group), over time the Dil-stained area showed a statistically significant increase in all cases. A statistically significant reduction of the mean RTA in the treated subgroups for at least one chemotherapy scheme was reported in 6/15 (40%) cases. The analysis of the percentage reduction of the RTA in treated subgroups in comparison to the control group revealed the presence of a linear relationship in each subgroup between the percentage reduction of the RTA and the number of cases reporting each percentage threshold considered for the analysis.

Conclusion: Our model seems to be effective for the xenotransplantation of PDAC tissue and evaluation of the effect of each chemotherapy scheme on the xenotransplanted tumor tissue.

Keywords: Avatar of oncological patients; Chemotherapy efficacy; Pancreatic ductal adenocarcinoma; Personalized medicine; Xenotransplantation; Zebrafish embryos.

Conflict of interest statement

Conflict-of-interest statement: The authors declare that there is no conflict of interest.

©The Author(s) 2020. Published by Baishideng Publishing Group Inc. All rights reserved.

Figures

Figure 1
Figure 1
Protocol used for the evaluation of the chemotherapy drugs efficacy. A piece of pancreatic ductal adenocarcinoma tumor tissue is injected into the yolk sac of zebrafish embryos 2 dpf. At 2 h post injection (hpi) the embryos are imaged and exposed to chemotherapy for 2 d. A and B: On the right panel a representative image of control embryos after 2 hpi and 2 dpi; C and D: On the right panel a representative image of treated embryos after 2 hpi and 2 dpi. Qualitatively images show the increased of fluorescent area in control group versus the regression in xenotransplanted embryos treated with chemotherapy.
Figure 2
Figure 2
Tumor tissue was successfully engrafted in healthy zebrafish embryos in all cases. A: Surgical specimen was obtained from patient with diagnosed pancreatic ductal adenocarcinoma (PDAC); B: Two fragments of the tumor were taken; C and D: One of them was used for histological evaluation (C) revealing that the percentage of epithelial cells (mean PDAC counterpart) out of the total surface area is 31.8% ± 4.9% (n = 3) (D); E: The second fragment of the tumor was used for xenotransplantation into the yolk of zebrafish embryos 2 dpf. After 2 d post xenotransplantation, the zebrafish patient-derived xenografts (zPDX) are Formalin-Fixed Paraffin Embedded. We performed the hematoxylin and eosin staining and immunohistochemistry using anti-Human Pan-Cytokeratin antibody on zPDX sections, highlighting the presence of epithelial PDAC cells (orange arrows) and the immune-negative counterpart (black arrows) that might be associated with the microenvironment side of PDAC human tissue. PDAC: Pancreatic ductal adenocarcinoma; PDX: Patient-derived xenografts; PanCK: Pan-Cytokeratin antibody.
Figure 3
Figure 3
Representative hematoxylin and eosin stained sections of zebrafish patient-derived xenografts. Visible morphologic alteration of the human pancreatic ductal adenocarcinoma nuclei (orange arrows) in zebrafish patient-derived xenografts exposed to Gemcitabine + nab-Paclitaxel and 5-Fluorouracil + Folinic acid + Oxaliplatin + Irinotecan could be associated with a chemotherapy damage. Control group shows normal nuclei (black arrows). GEM/nab-P: Gemcitabine + nab-Paclitaxel; FOLFOXIRI: 5-Fluorouracil + Folinic acid + Oxaliplatin + Irinotecan.
Figure 4
Figure 4
Cases with a statistically significant reduction of the mean relative tumor area for at least one chemotherapy scheme (the code below each diagram corresponds to the case number). Results are expressed as average ± SEM, aP < 0.05, 1-way ANOVA followed by Dunnett’s multiple comparisons test. Control group shows normal nuclei (black arrows). GEM: Gemcitabine; GEMOX: Gemcitabine + Oxaliplatin; GEM/nab-P: Gemcitabine + nab-Paclitaxel; FOLFOXIRI: 5-Fluorouracil + Folinic acid + Oxaliplatin + Irinotecan.
Figure 5
Figure 5
Percentage of cases with a percentage reduction of mean relative tumor area equal or greater to each threshold value. RTA: relative tumor area; Control group shows normal nuclei (black arrows). k: Control group; GEM: Gemcitabine; GEMOX: Gemcitabine + Oxaliplatin; GEM/nab-P: Gemcitabine + nab-Paclitaxel; FOLFOXIRI: 5-Fluorouracil + Folinic acid + Oxaliplatin + Irinotecan.

References

    1. Ferlay J, Colombet M, Soerjomataram I, Dyba T, Randi G, Bettio M, Gavin A, Visser O, Bray F. Cancer incidence and mortality patterns in Europe: Estimates for 40 countries and 25 major cancers in 2018. Eur J Cancer. 2018;103:356–387.
    1. Rawla P, Sunkara T, Gaduputi V. Epidemiology of Pancreatic Cancer: Global Trends, Etiology and Risk Factors. World J Oncol. 2019;10:10–27.
    1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424.
    1. Terashima T, Yamashita T, Sakai A, Ohta H, Hinoue Y, Toya D, Kawai H, Yonejima M, Urabe T, Noda Y, Mizukoshi E, Kaneko S. Treatment patterns and outcomes of unresectable pancreatic cancer patients in real-life practice: a region-wide analysis. Jpn J Clin Oncol. 2018;48:966–973.
    1. Schlick K, Magnes T, Ratzinger L, Jaud B, Weiss L, Melchardt T, Greil R, Egle A. Novel models for prediction of benefit and toxicity with FOLFIRINOX treatment of pancreatic cancer using clinically available parameters. PLoS One. 2018;13:e0206688.
    1. Goldstein D, El-Maraghi RH, Hammel P, Heinemann V, Kunzmann V, Sastre J, Scheithauer W, Siena S, Tabernero J, Teixeira L, Tortora G, Van Laethem JL, Young R, Penenberg DN, Lu B, Romano A, Von Hoff DD. nab-Paclitaxel plus gemcitabine for metastatic pancreatic cancer: long-term survival from a phase III trial. J Natl Cancer Inst. 2015:107.
    1. Conroy T, Desseigne F, Ychou M, Bouché O, Guimbaud R, Bécouarn Y, Adenis A, Raoul JL, Gourgou-Bourgade S, de la Fouchardière C, Bennouna J, Bachet JB, Khemissa-Akouz F, Péré-Vergé D, Delbaldo C, Assenat E, Chauffert B, Michel P, Montoto-Grillot C, Ducreux M Groupe Tumeurs Digestives of Unicancer; PRODIGE Intergroup. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364:1817–1825.
    1. Tentler JJ, Tan AC, Weekes CD, Jimeno A, Leong S, Pitts TM, Arcaroli JJ, Messersmith WA, Eckhardt SG. Patient-derived tumour xenografts as models for oncology drug development. Nat Rev Clin Oncol. 2012;9:338–350.
    1. Lee LM, Seftor EA, Bonde G, Cornell RA, Hendrix MJ. The fate of human malignant melanoma cells transplanted into zebrafish embryos: assessment of migration and cell division in the absence of tumor formation. Dev Dyn. 2005;233:1560–1570.
    1. Aboulkheyr Es H, Montazeri L, Aref AR, Vosough M, Baharvand H. Personalized Cancer Medicine: An Organoid Approach. Trends Biotechnol. 2018;36:358–371.
    1. Perkhofer L, Frappart PO, Müller M, Kleger A. Importance of organoids for personalized medicine. Per Med. 2018;15:461–465.
    1. Nakagawa H, Fujita M. Whole genome sequencing analysis for cancer genomics and precision medicine. Cancer Sci. 2018;109:513–522.
    1. Prokop JW, May T, Strong K, Bilinovich SM, Bupp C, Rajasekaran S, Worthey EA, Lazar J. Genome sequencing in the clinic: the past, present, and future of genomic medicine. Physiol Genomics. 2018;50:563–579.
    1. Konantz M, Balci TB, Hartwig UF, Dellaire G, André MC, Berman JN, Lengerke C. Zebrafish xenografts as a tool for in vivo studies on human cancer. Ann N Y Acad Sci. 2012;1266:124–137.
    1. Marques IJ, Weiss FU, Vlecken DH, Nitsche C, Bakkers J, Lagendijk AK, Partecke LI, Heidecke CD, Lerch MM, Bagowski CP. Metastatic behaviour of primary human tumours in a zebrafish xenotransplantation model. BMC Cancer. 2009;9:128.
    1. Weiss FU, Marques IJ, Woltering JM, Vlecken DH, Aghdassi A, Partecke LI, Heidecke CD, Lerch MM, Bagowski CP. Retinoic acid receptor antagonists inhibit miR-10a expression and block metastatic behavior of pancreatic cancer. Gastroenterology. 2009;137:2136–45.e1-7.
    1. Guo M, Wei H, Hu J, Sun S, Long J, Wang X. U0126 inhibits pancreatic cancer progression via the KRAS signaling pathway in a zebrafish xenotransplantation model. Oncol Rep. 2015;34:699–706.
    1. Amin MB, Edge SB, American Joint Committee on Cancer. AJCC cancer staging manual. 8th ed. New York: Springer, 2017.
    1. Kimmel CB, Ballard WW, Kimmel SR, Ullmann B, Schilling TF. Stages of embryonic development of the zebrafish. Dev Dyn. 1995;203:253–310.
    1. Usai A, Di Franco G, Colucci P, Pollina LE, Vasile E, Funel N, Palmeri M, Dente L, Falcone A, Morelli L, Raffa V. A Model of a Zebrafish Avatar for Co-Clinical Trials. Cancers (Basel) 2020;12
    1. Watanabe H, Okada M, Kaji Y, Satouchi M, Sato Y, Yamabe Y, Onaya H, Endo M, Sone M, Arai Y. [New response evaluation criteria in solid tumours-revised RECIST guideline (version 1.1)] Gan To Kagaku Ryoho. 2009;36:2495–2501.
    1. Gaudenzi G, Albertelli M, Dicitore A, Würth R, Gatto F, Barbieri F, Cotelli F, Florio T, Ferone D, Persani L, Vitale G. Patient-derived xenograft in zebrafish embryos: a new platform for translational research in neuroendocrine tumors. Endocrine. 2017;57:214–219.
    1. Costa B, Estrada MF, Mendes RV, Fior R. Zebrafish Avatars towards Personalized Medicine-A Comparative Review between Avatar Models. Cells. 2020;9
    1. Ooft SN, Weeber F, Dijkstra KK, McLean CM, Kaing S, van Werkhoven E, Schipper L, Hoes L, Vis DJ, van de Haar J, Prevoo W, Snaebjornsson P, van der Velden D, Klein M, Chalabi M, Boot H, van Leerdam M, Bloemendal HJ, Beerepoot LV, Wessels L, Cuppen E, Clevers H, Voest EE. Patient-derived organoids can predict response to chemotherapy in metastatic colorectal cancer patients. Sci Transl Med. 2019;11
    1. Vlachogiannis G, Hedayat S, Vatsiou A, Jamin Y, Fernández-Mateos J, Khan K, Lampis A, Eason K, Huntingford I, Burke R, Rata M, Koh DM, Tunariu N, Collins D, Hulkki-Wilson S, Ragulan C, Spiteri I, Moorcraft SY, Chau I, Rao S, Watkins D, Fotiadis N, Bali M, Darvish-Damavandi M, Lote H, Eltahir Z, Smyth EC, Begum R, Clarke PA, Hahne JC, Dowsett M, de Bono J, Workman P, Sadanandam A, Fassan M, Sansom OJ, Eccles S, Starling N, Braconi C, Sottoriva A, Robinson SP, Cunningham D, Valeri N. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science. 2018;359:920–926.
    1. Sachs N, de Ligt J, Kopper O, Gogola E, Bounova G, Weeber F, Balgobind AV, Wind K, Gracanin A, Begthel H, Korving J, van Boxtel R, Duarte AA, Lelieveld D, van Hoeck A, Ernst RF, Blokzijl F, Nijman IJ, Hoogstraat M, van de Ven M, Egan DA, Zinzalla V, Moll J, Boj SF, Voest EE, Wessels L, van Diest PJ, Rottenberg S, Vries RGJ, Cuppen E, Clevers H. A Living Biobank of Breast Cancer Organoids Captures Disease Heterogeneity. Cell. 2018;172:373–386.e10.
    1. Kim M, Mun H, Sung CO, Cho EJ, Jeon HJ, Chun SM, Jung DJ, Shin TH, Jeong GS, Kim DK, Choi EK, Jeong SY, Taylor AM, Jain S, Meyerson M, Jang SJ. Patient-derived lung cancer organoids as in vitro cancer models for therapeutic screening. Nat Commun. 2019;10:3991.
    1. Rubio-Viqueira B, Jimeno A, Cusatis G, Zhang X, Iacobuzio-Donahue C, Karikari C, Shi C, Danenberg K, Danenberg PV, Kuramochi H, Tanaka K, Singh S, Salimi-Moosavi H, Bouraoud N, Amador ML, Altiok S, Kulesza P, Yeo C, Messersmith W, Eshleman J, Hruban RH, Maitra A, Hidalgo M. An in vivo platform for translational drug development in pancreatic cancer. Clin Cancer Res. 2006;12:4652–4661.
    1. Malaney P, Nicosia SV, Davé V. One mouse, one patient paradigm: New avatars of personalized cancer therapy. Cancer Lett. 2014;344:1–12.
    1. Inoue T, Terada N, Kobayashi T, Ogawa O. Patient-derived xenografts as in vivo models for research in urological malignancies. Nat Rev Urol. 2017;14:267–283.
    1. Schartl M. Beyond the zebrafish: diverse fish species for modeling human disease. Dis Model Mech. 2014;7:181–192.
    1. Wertman J, Veinotte CJ, Dellaire G, Berman JN. The Zebrafish Xenograft Platform: Evolution of a Novel Cancer Model and Preclinical Screening Tool. Adv Exp Med Biol. 2016;916:289–314.
    1. Wu JQ, Zhai J, Li CY, Tan AM, Wei P, Shen LZ, He MF. Patient-derived xenograft in zebrafish embryos: a new platform for translational research in gastric cancer. J Exp Clin Cancer Res. 2017;36:160.
    1. Fior R, Póvoa V, Mendes RV, Carvalho T, Gomes A, Figueiredo N, Ferreira MG. Single-cell functional and chemosensitive profiling of combinatorial colorectal therapy in zebrafish xenografts. Proc Natl Acad Sci USA. 2017;114:E8234–E8243.
    1. Veinotte CJ, Dellaire G, Berman JN. Hooking the big one: the potential of zebrafish xenotransplantation to reform cancer drug screening in the genomic era. Dis Model Mech. 2014;7:745–754.
    1. Astone M, Dankert EN, Alam SK, Hoeppner LH. Fishing for cures: The alLURE of using zebrafish to develop precision oncology therapies. NPJ Precis Oncol. 2017;1
    1. Bentley VL, Veinotte CJ, Corkery DP, Pinder JB, LeBlanc MA, Bedard K, Weng AP, Berman JN, Dellaire G. Focused chemical genomics using zebrafish xenotransplantation as a pre-clinical therapeutic platform for T-cell acute lymphoblastic leukemia. Haematologica. 2015;100:70–76.
    1. Barriuso J, Nagaraju R, Hurlstone A. Zebrafish: a new companion for translational research in oncology. Clin Cancer Res. 2015;21:969–975.
    1. Lin J, Zhang W, Zhao JJ, Kwart AH, Yang C, Ma D, Ren X, Tai YT, Anderson KC, Handin RI, Munshi NC. A clinically relevant in vivo zebrafish model of human multiple myeloma to study preclinical therapeutic efficacy. Blood. 2016;128:249–252.
    1. Tong H, Fan Z, Liu B, Lu T. The benefits of modified FOLFIRINOX for advanced pancreatic cancer and its induced adverse events: a systematic review and meta-analysis. Sci Rep. 2018;8:8666.
    1. Lakatos G, Petranyi A, Szűcs A, Nehéz L, Harsanyi L, Hegyi P, Bodoky G. Efficacy and Safety of FOLFIRINOX in Locally Advanced Pancreatic Cancer. A Single Center Experience. Pathol Oncol Res. 2017;23:753–759.
    1. Nitsche U, Wenzel P, Siveke JT, Braren R, Holzapfel K, Schlitter AM, Stöß C, Kong B, Esposito I, Erkan M, Michalski CW, Friess H, Kleeff J. Resectability After First-Line FOLFIRINOX in Initially Unresectable Locally Advanced Pancreatic Cancer: A Single-Center Experience. Ann Surg Oncol. 2015;22 Suppl 3:S1212–S1220.
    1. Sadot E, Doussot A, O'Reilly EM, Lowery MA, Goodman KA, Do RK, Tang LH, Gönen M, D'Angelica MI, DeMatteo RP, Kingham TP, Jarnagin WR, Allen PJ. FOLFIRINOX Induction Therapy for Stage 3 Pancreatic Adenocarcinoma. Ann Surg Oncol. 2015;22:3512–3521.
    1. Wang ZQ, Zhang F, Deng T, Zhang L, Feng F, Wang FH, Wang W, Wang DS, Luo HY, Xu RH, Ba Y, Li YH. The efficacy and safety of modified FOLFIRINOX as first-line chemotherapy for Chinese patients with metastatic pancreatic cancer. Cancer Commun (Lond) 2019;39:26.
    1. Kræmer PC, Schmidt HH, Ladekarl M. Danish experiences with FOLFIRINOX as first-line therapy in patients with inoperable pancreatic cancer. Dan Med J. 2014;61:A4819.
    1. Shi YX, Xu RH, Jiang WQ, Zhang L, Lin TY, Li YH, Xia ZJ, Luo HY, Han B, Wang F, He YJ, Guan ZZ. [Efficacy of gemcitabine combined oxaliplatin on advanced pancreatic cancer] Ai Zheng. 2007;26:1381–1384.
    1. Li J, Merl M, Lee MX, Kaley K, Saif MW. Safety and efficacy of single-day GemOx regimen in patients with pancreatobiliary cancer: a single institution experience. Expert Opin Drug Saf. 2010;9:207–213.
    1. Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, Seay T, Tjulandin SA, Ma WW, Saleh MN, Harris M, Reni M, Dowden S, Laheru D, Bahary N, Ramanathan RK, Tabernero J, Hidalgo M, Goldstein D, Van Cutsem E, Wei X, Iglesias J, Renschler MF. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369:1691–1703.
    1. Casanova-Martinez C, Romero-Ventosa EY, González-Costas S, Arroyo-Conde C, Piñeiro-Corrales G. Evaluation of the use of nab-paclitaxel and gemcitabine in clinical practice. J Cancer Res Ther. 2018;14:S730–S735.
    1. Karasic TB, O'Hara MH, Loaiza-Bonilla A, Reiss KA, Teitelbaum UR, Borazanci E, De Jesus-Acosta A, Redlinger C, Burrell JA, Laheru DA, Von Hoff DD, Amaravadi RK, Drebin JA, O'Dwyer PJ. Effect of Gemcitabine and nab-Paclitaxel With or Without Hydroxychloroquine on Patients With Advanced Pancreatic Cancer: A Phase 2 Randomized Clinical Trial. JAMA Oncol. 2019;5:993–998.
    1. Montes AF, Villarroel PG, Ayerbes MV, Gómez JC, Aldana GQ, Tuñas LV, Fernández MS, Fernández MJ. Prognostic and predictive markers of response to treatment in patients with locally advanced unresectable and metastatic pancreatic adenocarcinoma treated with gemcitabine/nab-paclitaxel: Results of a retrospective analysis. J Cancer Res Ther. 2017;13:240–245.
    1. Louvet C, Labianca R, Hammel P, Lledo G, Zampino MG, André T, Zaniboni A, Ducreux M, Aitini E, Taïeb J, Faroux R, Lepere C, de Gramont A GERCOR; GISCAD. Gemcitabine in combination with oxaliplatin compared with gemcitabine alone in locally advanced or metastatic pancreatic cancer: results of a GERCOR and GISCAD phase III trial. J Clin Oncol. 2005;23:3509–3516.
    1. Ergun Y, Ozdemir NY, Guner EK, Esin E, Sendur MA, Koksoy EB, Demirci NS, Eren T, Dede I, Sezer A, Engin H, Oksuzoglu B, Yalcin B, Utkan G, Zengin N, Urun Y. Comparison of Gemcitabine monotherapy with Gemcitabine and Cisplatin combination in metastatic pancreatic cancer: a retrospective analysis. J BUON. 2018;23:116–121.

Source: PubMed

3
Sottoscrivi