scholarly journals Establishment and Characterization of a Reliable Xenograft Model of Hodgkin Lymphoma Suitable for the Study of Tumor Origin and the Design of New Therapies

Cancers ◽  
2018 ◽  
Vol 10 (11) ◽  
pp. 414 ◽  
Author(s):  
Radhia M’kacher ◽  
Monika Frenzel ◽  
Mustafa Al Jawhari ◽  
Steffen Junker ◽  
Corina Cuceu ◽  
...  

To identify the cells responsible for the initiation and maintenance of Hodgkin lymphoma (HL) cells, we have characterized a subpopulation of HL cells grown in vitro and in vivo with the aim of establishing a reliable and robust animal model for HL. To validate our model, we challenged the tumor cells in vivo by injecting the alkylating histone-deacetylase inhibitor, EDO-S101, a salvage regimen for HL patients, into xenografted mice. Methodology: Blood lymphocytes from 50 HL patients and seven HL cell lines were used. Immunohistochemistry, flow cytometry, and cytogenetics analyses were performed. The in vitro and in vivo effects of EDO-S101 were assessed. Results: We have successfully determined conditions for in vitro amplification and characterization of the HL L428-c subline, containing a higher proportion of CD30−/CD15− cells than the parental L428 cell line. This subline displayed excellent clonogenic potential and reliable reproducibility upon xenografting into immunodeficient NOD-SCID-gamma (−/−)(NSG) mice. Using cell sorting, we demonstrate that CD30−/CD15− subpopulations can gain the phenotype of the L428-c cell line in vitro. Moreover, the human cells recovered from the seventh week after injection of L428-c cells into NSG mice were small cells characterized by a high frequency of CD30−/CD15− cells. Cytogenetic analysis demonstrated that they were diploid and showed high telomere instability and telomerase activity. Accordingly, chromosomal instability emerged, as shown by the formation of dicentric chromosomes, ring chromosomes, and breakage/fusion/bridge cycles. Similarly, high telomerase activity and telomere instability were detected in circulating lymphocytes from HL patients. The beneficial effect of the histone-deacetylase inhibitor EDO-S101 as an anti-tumor drug validated our animal model. Conclusion: Our HL animal model requires only 103 cells and is characterized by a high survival/toxicity ratio and high reproducibility. Moreover, the cells that engraft in mice are characterized by a high frequency of small CD30−/CD15− cells exhibiting high telomerase activity and telomere dysfunction.

2002 ◽  
Vol 64 (7) ◽  
pp. 1079-1090 ◽  
Author(s):  
Yuka Sasakawa ◽  
Yoshinori Naoe ◽  
Takeshi Inoue ◽  
Tatsuya Sasakawa ◽  
Masahiko Matsuo ◽  
...  

2020 ◽  
Vol 16 (16) ◽  
pp. 3184-3199
Author(s):  
Batsaikhan Buyandelger ◽  
Eli E Bar ◽  
Kuo-Sheng Hung ◽  
Ruei-Ming Chen ◽  
Yung-Hsiao Chiang ◽  
...  

Leukemia ◽  
2004 ◽  
Vol 18 (12) ◽  
pp. 1951-1963 ◽  
Author(s):  
E Weisberg ◽  
L Catley ◽  
J Kujawa ◽  
P Atadja ◽  
S Remiszewski ◽  
...  

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1108-1108
Author(s):  
Yaya Chu ◽  
Ashlin Yahr ◽  
Janet Ayello ◽  
Mitchell S. Cairo

Abstract Background: The outcome for patients with Burkitt lymphoma (BL)has improved significantly but for patients who relapse, the prognosis is dismal due to chemo-radiotherapy resistance (Cairo et al, JCO, 2012).Natural Killer (NK) cells play an important role in tumor eradication post allogeneic stem cell transplantation (Ruggeri, Science 2002). Our group has successfully engineered expanded peripheral blood Natural Killer cells (exPBNK) cells with an anti-CD20 chimeric antigen receptor (CAR exPBNK) to target relapsed/resistant CD20+ BL cells in vitro and in NSG mice (Chu & Cairo, ASH, 2013). Romidepsin, a histone deacetylase inhibitor, enhances NKG2D ligands expression (Satwani, Chu/Cairo, Cytotherapy 2014). Objective: We investigated the combined effect of anti-CD20 CAR expanded PBNK cells with Romidepsin against CD20+ BL cells in vitro and in humanized NSG mice. Methods: PBNK cells were expanded with inactivated K562-mbIL15-41BBL cells and purified. Anti-CD20-4-1BB-CD3ζ mRNA was produced in vitro and nucleofected into exPBNK (Chu & Cairo, ASH, 2013). CAR expression was detected by flow cytometry. Raji, Raji-2R and Raji-4RH (provided by Matthew Barth, MD) cells were treated with 10ng/ml romidepsin, provided by Celgene. MICA/B expression was analyzed by flow cytometry. NK cytotoxicity was assessed by europium release assays. Humanized Raji xenografted NSG mice were established as previously described (Chu & Cairo, ASH, 2013). After successful Raji-Luc engraftment in mice at day 7, 2.2mg/kg romidepsin or PBS was i.p. injected to these mice. 5x106 anti-CD20 CAR exPBNK cells or MOCK exPBNK cells (without anti-CD20 CAR expression) were i.p. injected to each mouse 24hrs later after romidepsin injection. Romidepsin followed by exPBNK or CAR exPBNK therapy was given to each mouse once a week for continuous 3 weeks. Tumor regression and/or progression were monitored weekly by tumor volume measurement and by in vivo bioluminescent imaging. Statistical probability of survival and comparison of survival curves were determined by Kaplan Meier Method and the log-rank test. The results with a P value < 0.05 were deemed statistically significant. Results: MICA/B expression was significantly increased in rituximab sensitive Raji (Day0 vs Day1: 0.634+ 0.074% vs 47.7+1.04%; n=3, P<0.001), rituximab resistant Raji-2R (Day0 vs Day1: 0.472+ 0.091% vs 75.6+2.56%; n=3, P<0.001) and Raji-4RH (Day0 vs Day1: 0.187+ 0.0196% vs 69.33+0.841%; n=3, P<0.001) after romidepsin treatment. In vitro cytotoxicity of ex vivo expanded PBNK (exPBNK) was significantly enhanced against romidepsin treated Raji, Raji-2R, and Raji-4RH compared to untreated targets at E:T=3:1 (6.3+ 0.38% vs 16.93+1.2%, P<0.001; 42.82+ 2.2% vs 63.44+2%, P<0.001; 61.78+ 1.01% vs 74.78+1.8%, P<0.001; n=4). Blocking NKG2D in exPBNK significantly reduced in vitro cytotoxicity against romidepsin treated Raji, Raji-2R, and Raji-4RH compared to the unblocked exPBNK (P<0.001), indicating NKG2D and MICA/B are involved in the enhanced cytotoxicity in romidepsin treated BL. In vitro cytotoxicity of anti-CD20 CAR modified exPBNK was further significantly enhanced against romidepsin treated Raji, Raji-2R, and Raji-4RH compared to untreated targets at E:T=3:1 (24.48+ 1.11% vs 6.28+0.85%, P<0.001; 74.75+ 3.34% vs 47.99+0.5%, P<0.001; 80.6+ 0.77% vs 68.9+1.8%, P<0.001; n=4), or compared to the mock exPBNK against romidepsin treated targets (24.48+ 1.11% vs 18.52+1.06%, P<0.01; 74.75+ 3.34% vs 66.65+2.34%, P<0.05; 80.6+ 0.77% vs 74.7+1.01%, P<0.05; n=4). Rs4;11 was used as another control (Fig.1A) In humanized Raji xenograft NSG mice, the romidepsin+CAR exPBNK treated mice had significantly extended surviving time (over 60% mice surviving more than 100 days) compared to the untreated mice (median 28 days, P<0.001), the mock exPBNK treated mice (median 29 days, P<0.001), the CAR exPBNK treated mice (median 42.5 days, P<0.05), the romidepsin treated mice (median 30 days, P<0.001), and the romidepsin+mock exPBNK treated mice (median 34.5 days, P<0.05) (Fig.1B). Conclusion: anti-CD20 CAR modified expanded PBNK cells significantly enhanced cytotoxicity against romidepsin treated rituximab sensitive and resistant BL in vitro and in vivo. These results indicate the therapeutic potential of the combination of anti-CD20 CAR modified exPBNK cells and romidepsin against BL disease in patients with chemo-radio therapy resistance. Figure 1 Figure 1. Disclosures Cairo: Celgene: Research Funding.


Sign in / Sign up

Export Citation Format

Share Document