scholarly journals Effectively Targeting Burkitt Lymphoma By Anti-CD20 Chimeric Antigen Receptor (CAR) Modified Expanded Natural Killer Cells Combined with a Histone Deacetylase Inhibitor, Romidepsin in Vitro and In Humanized NSG Mice

2015 ◽  
Vol 21 (2) ◽  
pp. S152-S153 ◽  
Author(s):  
Yaya Chu ◽  
Ashlin Yahr ◽  
Janet Ayello ◽  
Mitchell S. Cairo
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.


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.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3085-3085 ◽  
Author(s):  
Yaya Chu ◽  
Fangyu Lee ◽  
Janet Ayello ◽  
Brian Hang ◽  
Melanie Zhang ◽  
...  

Abstract Background: The outcome for children with Burkitt lymphoma (BL)has improved significantly but for patients who relapse, the prognosis is dismal due to chemo-immunotherapy resistance (Cairo et al, JCO, 2012, Cairo et al, Blood, 2007). NK cells are bone marrow-derived cytotoxic lymphocytes that play a major role in the rejection of tumors. A variety of activating and inhibitory receptors on the NK cell surface are engaged to regulate NK cell activities and to discriminate target cells from other healthy 'self' cells. However, NK therapy is limited by several factors, including small numbers of active NK cells in unmodified peripheral blood, lack of tumor targeting specificity, and multiple mechanisms of tumor escape of NK cell immunosurveillance. Our group has successfully modified expanded peripheral blood Natural Killer cells (exPBNK) with an anti-CD20 CAR to target rituximab sensitive/resistant CD20+ BL cells in vitro and in NSG mice (Chu/Cairo, et al, Can Imm Res 2015). However, the short lifespan/persistence of adoptively transferred NK cells has limited the therapeutic efficacy. ALT-803 (Altor BioScience Corporation) is a superagonist of an IL-15 variant bound to an IL-15Rα-Fc fusion with enhanced IL-15 biological activity (Zhu et al. 2009 J Immunol), longer half-life and increased potency (Han, et al. Cytokine. 2011). It is currently in several clinical trials in patients with variety of cancers such as refractory indolent non-Hodgkin's lymphoma (NCT02384954). Objective: We hypothesize that ALT-803, IL-15 superagonist complex, promotes exPBNK persistence and significantly enhances the cytotoxicity of anti-CD20 CAR exPBNK against CD20+ BL. Method: PBMCs were expanded with lethally irradiated K562-mbIL21-41BBL cells (Dean Lee et al, PLoS One, 2012). CD56+ CD3- exPBNK cells were isolated using Miltenyi NK cell isolation kit. Anti-CD20-4-1BB-CD3 ζ mRNA (CAR mRNA) was producedin vitro and nucleofected into exPBNK as we have previously described (Chu/Cairo, et al, Can Imm Res 2015). ALT-803 was provided by Altor BioScience Corporation. ExPBNK cells were cultured with 0.35ng/ml or 3.5ng/ml ALT-803. NK proliferation was monitored with MTS assays. NK receptors expression and cytotoxicity were examined by flow cytometry (Chu/Cairo, et al, ASH 2014). NK resistant BL cells Raji and Daudi were used as target cells. Results: % CD56+ CD3- PBNK cells were significantly increased compared to media alone at day 14 (mean 81.85% vs 14.91%, n=3, p<0.001) when co-cultured with the irradiated feeder cell K562-mbIL21-41BBL. The absolute NK numbers were enhanced with irradiated K562-mbIL21-41BBL cells as feeders compared to IL-2 alone after normalized to the INPUT NK cell numbers (mean 2247 fold±293.7 vs 0.516 fold±0.225, n=3, p<0.001) at day 14. Different doses of ALT-803 or IgG were added to the culture medium of purified expanded exPBNK. Proliferation assays were performed at day 3, 7,11, and 17. ALT-803 significantly promoted exPBNK proliferation and persistence compared to IgG in vitro in a dose-dependent manner (A490 reading at 3.5ng/ml dose: ALT803 vs IgG=0.3383+0.009 vs 0.0987+0.0007, P<0.0001 at d17). And ALT-803 significantly enhanced exPBNK cytotoxicity against NK resistant BL cells: Raji (ALT803 vs IgG= 49.54%+2.7% vs 5.99+0.34%, p<0.001, E:T=10:1) and Daudi (ALT803 vs IgG= 63.73%+3.09% vs 2.58+1.96%, p<0.001, E:T=10:1). It also maintained the highcytoxicity of exPBNK at d4, d10 and d18 against Raji (E:T=10:1, d4 vs d10 vs d18=62.07% vs 49.54% vs 61.47%) and against Daudi (E:T=10:1, d4 vs d10 vs d18=76.02% vs 63.73% vs 55%) by maintaining the activating receptors expression such as NKp30, NKp44, and NKp46. Further-more, we demonstrated ALT-803 significantly enhanced the cytotoxicity of anti-CD20 CAR modified exPBNK against Raji (CAR vs MOCK= 81.19%+0.35% vs 66.19+0.94%, p<0.001, E:T=10:1) and Daudi (CAR vs MOCK= 91.41%+0.45% vs 80.56+1.07%, p<0.001, E:T=10:1) compared to mock modified exPBNK. ALT-803 also significantly enhanced the cytotoxicity of anti-CD20 CAR modified exPBNK against NK resistant BL cells: Raji and Daudi compared to anti-CD20 CAR modified exPBNK maintained in medium without ALT803 (Fig.1). Conclusions: ALT-803 maintained the cytotoxicity of exPBNK and in vitro persistence and significantly enhanced anti-CD20 CAR exPBNK cytotoxicity against pediatric NK resistant BL. The in vivo effect of ALT-803 on CAR exPBNK using humanized NSG models is under investigation. Disclosures Wong: Altor BioScience Corporation: Employment, Other: stockholder of Altor Bioscience Corporation. Lee:Intrexon, Ziopharm, Cyto-Sen: Equity Ownership.


2014 ◽  
Vol 3 (4) ◽  
pp. 333-344 ◽  
Author(s):  
Yaya Chu ◽  
Jessica Hochberg ◽  
Ashlin Yahr ◽  
Janet Ayello ◽  
Carmella van de Ven ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document