Phase I Trial of Multiple Infusions of Autologous Activated T Cells with Anti-CD3 x Anti-CD20 Bispecific Antibody (CD20Bi) after Autologous Peripheral Blood Stem Cell Transplant for NonHodgkins Lymphoma To Improve Graft-vs-Lymphoma Effects.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4488-4488 ◽  
Author(s):  
Lawrence G. Lum ◽  
Joseph P. Uberti ◽  
Zaid Al-Kadhimi ◽  
Cassara Skuba ◽  
Pat Steele ◽  
...  

Abstract Relapse rates after high dose chemotherapy (HDC) autologous peripheral blood stem cell transplant (PBSCT) for high risk refractory or relapsed non-Hodgkin’s lymphoma (NHL) patients remain unacceptably high. In order to augment the anti-lymphoma effect and decrease relapsed rates, our strategy combines 1) the cytotoxicity mediated by anti-CD3 activated autologous T cells (ATC) armed with a bispecific antibody (BiAb) that redirects ATC to kill CD20 + lymphoma cells and circumvents rituximab resistance (Exp Hemat33:452, 2005) and 2) autologous PBSCT after high dose chemotherapy. Arming ATC with anti-CD3 x anti-CD20 (CD20Bi) converts each ATC into a CD20-specific cytotoxic T cell. Our phase I trial tests whether multiple infusions of armed ATC armed given after HDC followed by PBSCT are safe and whether such infusions will provide an anti-lymphoma effect to improve overall survival and disease free survival after PBSCT. On day +4, the patients receive immune consolidation consisting of 3 infusions of CD20Bi-armed ATC per week for 3 weeks and then 1 infusion per week for 6 additional weeks. The dose levels are 5, 10, 15, and 20 x 109 cells/infusion with total armed ATC doses equaling 75, 150, 225, and 300 x 109 CD20Bi-armed ATC. Subcutaneous IL-2 (300,000 IU/m2/day) will be given daily beginning d+4 and ending after the last dose of armed ATC. The dose of CD34+/kg ranged from 1.04 to 3.4 x 106. T cells in the leukopheresis product were activated with anti-CD3 and expanded in low dose IL-2, harvested, armed with CD20Bi, and cryopreserved for infusions after PBSCT. The ATC harvest ranged from 119–140 billion with >94% viability with 96–99% CD3+, 33–67% CD4+, 32–53% CD8+, and <6.5% CD4+CD25+ and CD8+CD25+ cells. All three patients received all of their infusions. Three patients completed the first dose level (total of 70 x 109 CD20Bi armed ATC) without any dose limiting toxicities. All of the patients are alive 780, 650, and 521 days after PBSCT. Two are free of disease and one was transplanted with persistent disease went into remission after PBSCT and relapsed 8 months after PBSCT. He is now in remission following chemotherapy and a MUD PBSCT. The cytotoxicity mediated by the patients’ ex vivo expanded, armed ATC against the B9C cell line was significantly higher than unarmed ATC. CTL activity directed at B cell targets was detected by 3 weeks after PBSCT. These results suggest that large numbers of armed autologous ATC can be infused after PBSCT without major side effects related to the armed targeted ATC. This strategy may provide a unique opportunity to increase the GVL effect without increasing toxicities after autologous PBSCT for CD20+ lymphomas.

2007 ◽  
Vol 16 (5) ◽  
pp. 477-483 ◽  
Author(s):  
Michael W. Schuster ◽  
Tsiporah B. Shore ◽  
John G. Harpel ◽  
June Greenberg ◽  
Bita Jalilizeinali ◽  
...  

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2748-2748
Author(s):  
Lawrence G. Lum ◽  
Zaid Al-Kadhimi ◽  
Cassara Skuba ◽  
Voravit Ratanatharathorn ◽  
Joseph P. Uberti ◽  
...  

Abstract More aggressive treatment strategies are needed for women with metastatic breast cancer(mBrCa). Although peripheral blood stem cell transplant (PBSCT) permits the use of high-dose chemotherapy (HDC) that could not otherwise be given, results for PBSCT have not been encouraging. In order to boost tumor kill, we combined a protocol that targets Her2 using activated T cells (ATC) armed with anti-CD3 × anti-Her2 bispecific antibody (Her2Bi) with a second protocol that involves infusions of ATC after PBSCT to boost anti-tumor immunity. In our phase I trial using ATC with Her2Bi to treat women with mBrCA who have Her2 positve or negative disease, we found that multiple infusions of armed ATC induced cytotoxicity directed at BrCa cells in the peripheral blood mononuclear cells (PBMC) of the patients that develop in 2 weeks and last up to 4 mos (Clin Canc Res12:569,2006). Armed ATC lyse tumors that are Her2 low-expressors (0–1+). Targeting leads to specific cytotoxicity, induces cytokine/chemokine release, and proliferation of the armed ATC. In a second study, ATC were infused after PBSCT into 23 women with mBrCa leading to 70% overall survival and 50% progression free survival at 32 mos after PBSCT. Therefore, the combined strategy involved obtaining T cells by another leukopheresis after the boosting with armed ATC, expanding the immune T cells and infusing the expanded ATC after PBSCT to transfer pre-immune anti-BrCa cytotoxicity to reconstitute cytotoxicity after PBSCT. Two patients (one pt was Her2 3+ ; one pt was Her2 0–1+) underwent treatment with this treatment approach. Both patients were given 8 infusions (20 billion/infusion with a total of 160 billion) of ATC armed with Her2Bi in the first protocol and subsequently leukopheresed and ATC were produced for the second protocol. The expanded ATC at an effector:target ratio (E/T) of 25:1, exhibited cytotoxicity at BrCa tumor cells (SK-BR-3) at 71% and 75% for pts 1 and 2, respectively. The cell product for pt 1 contained 68% CD3+, 32% CD4+, 39% CD8+, 29% CD16+56+, 12% CD4+CD25+, and 15% CD8+CD25+ cells and the cell product for pt 2 contained 35% CD3+, 25% CD4+, 8.5% CD8+, 23.3% CD16+56+, 10.4% CD4+CD25+, and 4.7% CD8+CD25+ cells. The protocol involves infusing 15 doses of ATC after PBSCT with 3 doses of ATC/week for 3 weeks and then ATC once/week for six more weeks. The pt1 and pt 2 received total of 114 and 70 billion ATC, respectively. Pt1 developed anti-BrCA cytotoxicity of 9% 2 weeks after PBSCT. Pt2 exhibited anti-BrCa cytotoxicity at an E:T of 25:1 of 38% and 15% at 3 weeks and 6 months after PBSCT, respectively. Phenotyping of peripheral blood at 6 mos after PBSCT showed 61% CD3+, 36% CD4+, 19.5% CD8+, and 15.5% CD56+ cells. There was no cytotoxicity directed at Daudi cells. These data strongly suggest that transfer of pre-immune cells after PBSCT accelerate immune reconstitution of tumor specific cytotoxicity after PBSCT. The preboost strategy with targeted T cells is being combined in a proof of principle trial to assess whether enhanced cytotoxicity can be consistently enhanced after PBSCT.


Sign in / Sign up

Export Citation Format

Share Document