scholarly journals Targeting CD123 in acute myeloid leukemia using a T-cell–directed dual-affinity retargeting platform

Blood ◽  
2016 ◽  
Vol 127 (1) ◽  
pp. 122-131 ◽  
Author(s):  
Muneera Al-Hussaini ◽  
Michael P. Rettig ◽  
Julie K. Ritchey ◽  
Darja Karpova ◽  
Geoffrey L. Uy ◽  
...  

Key Points A novel CD3×CD123 DART agent induces T-cell-target-specific association, activation, and proliferation. The CD3×CD123 DART induces a dose-dependent killing of AML cell lines and primary AML blasts in vitro and in vivo.

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1383-1383 ◽  
Author(s):  
Tongyuan Xue ◽  
Marissa Del Real ◽  
Emanuela Marcucci ◽  
Candida Toribio ◽  
Sonia Maryam Setayesh ◽  
...  

Acute myeloid leukemia (AML) is the most common acute leukemia in adults. The cure rate for primary AML patients is only 35% and decreases with age. Novel and effective immunotherapies for patients with relapsed and/or refractory (r/r) AML remain an urgent unmet need. CD33 is an attractive immunotherapeutic target for myeloid malignancies given its expression on more than 85% of AML patient samples. We therefore set out to design and test CD33 chimeric antigen receptor (CD33CAR) T cells preclinically as a single agent and in combinational therapy. To assess antileukemic responses of CD33CAR T cells in vitro and in vivo, we enriched CD4/CD8 T cells from peripheral blood mononuclear cells (PBMCs) and genetically modified them to express a second-generation CD33CAR. CD33CAR T cells exhibited potent antigen dependent CD107a degranulation, IFN-γ production and killing activities against AML cells in vitro. Using a NOD-SCID-IL2Rgnull (NSG) xenograft model engrafted with MOLM-14-ffluc, a CD33 expressing AML cell line transduced with lentivirus carrying firefly luciferase (ffluc) and enhanced green fluorescent protein (eGFP), 3 million CD33CAR or mock T cells were introduced intravenously. CD33 CAR T cell-treated group displayed 98.2% leukemic regression 4 days post CAR T infusion, and 99.6% reduction on day 31. Bioluminescent imaging (BLI) and Kaplan-Meier analysis demonstrated that CD33CAR T cells significantly decreased leukemic burden and prolonged overall survival compared to mock T cells in vivo. Decitabine, a DNA hypomethylating agent (HMA), is a main therapeutic agent for treating AML. We observed HMA treatment led to increased CD33 expression on MOLM-14 cells in vitro. We hypothesized that decitabine can potentiate CD33CAR T cell-mediated AML killing by increasing CD33 expression. MOLM-14 cells were treated with either decitabine alone, CD33CAR T cells alone, or sequential treatment using various concentrations of decitabine or DMSO followed by CD33CAR or mock T cells in an E:T ratio of 1:100. We determined the target specific killing activities in each group using flow cytometric based analysis 48 and 96 hours later. The decitabine followed by CD33CAR T cells treatment reproducibly resulted in the most robust antileukemic activity with 80.6% MOLM-14 cells killed. In comparison, CD33CAR T cells or decitabine monotherapy resulted in 11.5% and 50.9% killing, respectively. In vivo testing of the combinational effects of decitabine and CD33CAR T cells are underway and will be updated at the meeting. Finally, checkpoint blockade targeting programmed death-1 (PD-1)/programmed death-ligand 1 (PD-L1) has shown survival benefits, particularly in combination with HMA, for patients with r/r AML (Daver et al. 2019). We observed elevated PD-L1 expression on residual AML blasts that survived the treatment with decitabine in combination with CD33CAR T cells. Therefore, we hypothesized that blockade of PD-1/PD-L1 interaction might further improve the antileukemic effect of CD33CAR T cells against AML cells post antigen induction by decitabine. MOLM-14 cells were treated with decitabine for 2 days and CD33CAR T cells were added in an E:T ratio of 1:75. Anti-PD-1 or IgG4 antibody was added to the culture at various concentrations. The most robust CD33 specific killing was seen in the culture with anti-PD-1 antibody added. Further characterization are underway and will be presented. Taken together, our preclinical findings have demonstrated the potency of the CD33CAR T cell therapy and ways to optimize its efficacy. Our results support clinical translation of CD33CAR T cells for patients with AML. Disclosures Budde: F. Hoffmann-La Roche Ltd: Consultancy.


Blood ◽  
2015 ◽  
Vol 125 (26) ◽  
pp. 4060-4068 ◽  
Author(s):  
Farideh Miraki-Moud ◽  
Essam Ghazaly ◽  
Linda Ariza-McNaughton ◽  
Katharine A. Hodby ◽  
Andrew Clear ◽  
...  

Key Points Most AMLs lack ASS1, which allows synthesis of arginine, and so depend on exogenous sources. Depletion of arginine via ADI-PEG 20 reduces the burden of primary AML in vivo and in vitro.


2012 ◽  
Vol 2012 ◽  
pp. 1-10 ◽  
Author(s):  
A. Dutour ◽  
V. Marin ◽  
I. Pizzitola ◽  
S. Valsesia-Wittmann ◽  
D. Lee ◽  
...  

Genetic engineering of T cells with chimeric T-cell receptors (CARs) is an attractive strategy to treat malignancies. It extends the range of antigens for adoptive T-cell immunotherapy, and major mechanisms of tumor escape are bypassed. With this strategy we redirected immune responses towards the CD33 antigen to target acute myeloid leukemia. To improvein vivoT-cell persistence, we modified human Epstein Barr Virus-(EBV-) specific cytotoxic T cells with an anti-CD33.CAR. Genetically modified T cells displayed EBV and HLA-unrestricted CD33 bispecificityin vitro. In addition, though showing a myeloablative activity, they did not irreversibly impair the clonogenic potential of normal CD34+hematopoietic progenitors. Moreover, after intravenous administration into CD33+human acute myeloid leukemia-bearing NOD-SCID mice, anti-CD33-EBV-specific T cells reached the tumor sites exerting antitumor activityin vivo. In conclusion, targeting CD33 by CAR-modified EBV-specific T cells may provide additional therapeutic benefit to AML patients as compared to conventional chemotherapy or transplantation regimens alone.


2021 ◽  
Vol 9 (5) ◽  
pp. e002509
Author(s):  
Sayed Shahabuddin Hoseini ◽  
Mallika Vadlamudi ◽  
Madelyn Espinosa-Cotton ◽  
Hoa Tran ◽  
Yi Feng ◽  
...  

BackgroundAcute myeloid leukemia (AML) remains one of the most challenging hematological malignancies. Despite progress in therapeutics, majority of patients succumb to this neoplasm. CD33 is a proven therapeutic target, given its expression on most AML cells. Almost all anti-CD33 antibodies target the membrane distal immunoglobulin V (IgV) domain of the CD33 extracellular domain.MethodsIn this manuscript, we present data on three bispecific antibodies (BsAbs) against the CD33 IgV and membrane proximal immunoglobulin C (IgC) domains. We use in vitro binding and cytotoxicity assays to show the effect of these BsAbs on AML cell lines. We also use immunodeficient mice-bearing leukemias from cell lines and patient-derived xenografts to show the effect of these BsAbs in vivo.ResultsIn vitro, the IgV-targeting BsAb had higher binding to AML cell lines using flow cytometry and delivered more potent cytotoxicity in T-cell-dependent cytotoxicity assays; importantly, the IgC domain-targeting outperformed the IgV domain-targeting BsAb in medullary and extramedullary leukemia animal models.ConclusionsThese data support further clinical development of this BsAb for first-in-human phase I clinical trial.


Blood ◽  
2016 ◽  
Vol 128 (3) ◽  
pp. 410-414 ◽  
Author(s):  
Ewa M. Kosciuczuk ◽  
Diana Saleiro ◽  
Barbara Kroczynska ◽  
Elspeth M. Beauchamp ◽  
Frank Eckerdt ◽  
...  

Key Points Merestinib blocks Mnk kinase activity in acute myeloid leukemia cells. Merestinib suppresses human leukemic progenitors and exhibits potent antileukemic effects in a xenograft mouse model.


Blood ◽  
2013 ◽  
Vol 121 (18) ◽  
pp. 3675-3681 ◽  
Author(s):  
Jessica K. Altman ◽  
Amy Szilard ◽  
Bruce W. Konicek ◽  
Philip W. Iversen ◽  
Barbara Kroczynska ◽  
...  

Key Points The Mnk inhibitor cercosporamide suppresses human leukemic progenitors and exhibits antileukemic effects in a xenograft mouse model. Cercosporamide enhances the antileukemic effects of cytarabine in vitro and in vivo.


Blood ◽  
2015 ◽  
Vol 125 (17) ◽  
pp. 2689-2692 ◽  
Author(s):  
Parvathi Ranganathan ◽  
Xueyan Yu ◽  
Ramasamy Santhanam ◽  
Jessica Hofstetter ◽  
Alison Walker ◽  
...  

Key Points Decitabine priming increases antileukemic effects of selinexor in AML in vitro and in vivo. Decitabine priming allows for decreasing the dose of selinexor in patients, thus increasing tolerability without affecting antileukemic activity.


Blood ◽  
2018 ◽  
Vol 131 (15) ◽  
pp. 1639-1653 ◽  
Author(s):  
Paolo Gallipoli ◽  
George Giotopoulos ◽  
Konstantinos Tzelepis ◽  
Ana S. H. Costa ◽  
Shabana Vohra ◽  
...  

Key PointsFLT3ITD TK inhibition impairs glycolysis and glucose utilization without equally affecting glutamine metabolism. Combined targeting of FLT3 TK activity and glutamine metabolism decreases FLT3ITD mutant cells leukemogenic potential in vitro and in vivo.


Blood ◽  
2016 ◽  
Vol 128 (7) ◽  
pp. 971-981 ◽  
Author(s):  
Rachel E. Rau ◽  
Benjamin A. Rodriguez ◽  
Min Luo ◽  
Mira Jeong ◽  
Allison Rosen ◽  
...  

Key Points Data from Dnmt3a−/− mice implicate Dot1l as a critical mediator of the malignant gene expression program of Dnmt3a-mediated leukemia. Pharmacologic inhibition of DOT1L exerts potent antileukemic activity in DNMT3A-mutant human acute myeloid leukemia in vitro and in vivo.


Cancers ◽  
2021 ◽  
Vol 13 (14) ◽  
pp. 3385
Author(s):  
Axel H. Schönthal ◽  
Steve Swenson ◽  
Radu O. Minea ◽  
Hye Na Kim ◽  
Heeyeon Cho ◽  
...  

Despite progress in the treatment of acute myeloid leukemia (AML), the clinical outcome remains suboptimal and many patients are still dying from this disease. First-line treatment consists of chemotherapy, which typically includes cytarabine (AraC), either alone or in combination with anthracyclines, but drug resistance can develop and significantly worsen prognosis. Better treatments are needed. We are developing a novel anticancer compound, NEO212, that was created by covalent conjugation of two different molecules with already established anticancer activity, the alkylating agent temozolomide (TMZ) and the natural monoterpene perillyl alcohol (POH). We investigated the anticancer activity of NEO212 in several in vitro and in vivo models of AML. Human HL60 and U937 AML cell lines, as well as different AraC-resistant AML cell lines, were treated with NEO212 and effects on cell proliferation, cell cycle, and cell death were investigated. Mice with implanted AraC-sensitive or AraC-resistant AML cells were dosed with oral NEO212, and animal survival was monitored. Our in vitro experiments show that treatment of cells with NEO212 results in growth inhibition via potent G2 arrest, which is followed by apoptotic cell death. Intriguingly, NEO212 was equally potent in highly AraC-resistant cells. In vivo, NEO212 treatment strikingly extended survival of AML mice and the majority of treated mice continued to thrive and survive without any signs of illness. At the same time, we were unable to detect toxic side effects of NEO212 treatment. All in all, the absence of side effects, combined with striking therapeutic activity even in an AraC-resistant context, suggests that NEO212 should be developed further toward clinical testing.


Sign in / Sign up

Export Citation Format

Share Document