Cooperative Enhancer Activation by TLX1 and STAT5 Drives Leukemia Development in NUP214-ABL1/TLX1-positive T-cell Acute Lymphoblastic Leukemia

2018 ◽  
Vol 64 ◽  
pp. S42
Author(s):  
Jan Cools
2016 ◽  
Vol 271 (1) ◽  
pp. 156-172 ◽  
Author(s):  
Diana Passaro ◽  
Christine Tran Quang ◽  
Jacques Ghysdael

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1421-1421
Author(s):  
Padma Akkapeddi ◽  
Ana Rita Fragoso ◽  
Julie Hixon ◽  
Mariana Oliveira ◽  
Tânia Carvalho ◽  
...  

Abstract T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological cancer that is treated with intensive multi-agent chemotherapy, often leading to long-term side-effects impacting the quality of life of survivors. Despite the therapeutic success in children, relapses still occur in 10-20% of the cases, and adults face a considerably poorer prognosis. Novel, more selective treatments that contribute to reducing toxicities and improving outcome are thus in need. Interleukin 7 (IL-7) and its receptor IL-7Rα promote leukemia development in a majority of T-ALL patients and mutational activation of IL-7Rα, which occurs in around 10% of the cases, associates with very high risk in relapsed disease. Using combinatorial scFv phage display libraries and antibody reformatting we have now generated a fully human IgG1 monoclonal antibody (named B12) against human IL-7Rα. B12 does not display cross-reactivity against the mouse receptor and recognizes both wild type and mutant forms of IL-7Rα naturally expressed in T-ALL cell lines and patient samples, as well as in Ba/F3 cells stably transduced with human, but not mouse, IL-7Rα. Interestingly, molecular dynamics simulations suggest that B12 forms a stable complex with IL-7Rα at a different site from IL-7. Nonetheless, B12 inhibits IL-7/IL-7R-mediated signaling and induces cell death per se in at least some IL-7/IL-7R-reliant T-ALL cell lines (e.g. IL-7-dependent TAIL7 cells and mutant IL7R DND4.1 cells) and patient samples. Using patient-derived xenograft (PDX) samples, HPB-ALL cells and D1 cells overexpressing a mutated gain-of-function form of IL-7Rα, we show that the antibody also promotes antibody-dependent NK-mediated leukemia cytotoxicity in vitro and delays T-cell leukemia development in vivo, reducing tumor burden and promoting mouse survival. Moreover, B12 cooperates with dexamethasone in promoting the death of both dexamethasone-resistant HPB-ALL cells and a dexamethasone-sensitive PDX sample. Notably, B12 is rapidly internalized via clathrin-coated pits to the early endosome, eventually trafficking to the lysosome - an effect that is slightly accelerated in the presence of IL-7. These characteristics render B12 an attractive vehicle for targeted intracellular delivery of a highly cytotoxic warhead. As such, we engineered a B12-mono-methyl auristatin E (MMAE) antibody-drug conjugate (ADC) in which site-specific conjugation of B12 was carried out by reducing inter-chain disulfide bonds and reacting the thiol group of the free cysteines with a Michael acceptor (carbonyl acrylic derivate) linked to a cleavable linker (valine-citrulline) and the drug (MMAE). Tested against different cell lines, primary patient cells and PDX samples, B12-MMAE ADC demonstrates increased leukemia cell killing ability in vitro as compared to the naked antibody. Altogether, our studies serve as a stepping stone towards the development of novel targeted therapeutic strategies in T-ALL and other diseases where IL-7Rα was shown to play a pathological role. Disclosures Akkapeddi: Instituto de Medicina Molecular João Lobo Antunes: Patents & Royalties: Patents. Neri:Philochem AG: Equity Ownership. Bernardes:Instituto de Medicina Molecular João Lobo Antunes: Patents & Royalties: Patents. Barata:Instituto de Medicina Molecular João Lobo Antunes: Patents & Royalties: Patents.


Oncotarget ◽  
2016 ◽  
Vol 7 (27) ◽  
pp. 41599-41611 ◽  
Author(s):  
Sandrine Poglio ◽  
Daniel Lewandowski ◽  
Julien Calvo ◽  
Aurélie Caye ◽  
Audrey Gros ◽  
...  

Cancers ◽  
2019 ◽  
Vol 11 (5) ◽  
pp. 629 ◽  
Author(s):  
Alberto M. Martelli ◽  
Francesca Paganelli ◽  
Antonietta Fazio ◽  
Chiara Bazzichetto ◽  
Fabiana Conciatori ◽  
...  

T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive blood cancer that comprises 10–15% of pediatric and ~25% of adult ALL cases. Although the curative rates have significantly improved over the past 10 years, especially in pediatric patients, T-ALL remains a challenge from a therapeutic point of view, due to the high number of early relapses that are for the most part resistant to further treatment. Considerable advances in the understanding of the genes, signaling networks, and mechanisms that play crucial roles in the pathobiology of T-ALL have led to the identification of the key drivers of the disease, thereby paving the way for new therapeutic approaches. PTEN is critical to prevent the malignant transformation of T-cells. However, its expression and functions are altered in human T-ALL. PTEN is frequently deleted or mutated, while PTEN protein is often phosphorylated and functionally inactivated by casein kinase 2. Different murine knockout models recapitulating the development of T-ALL have demonstrated that PTEN abnormalities are at the hub of an intricate oncogenic network sustaining and driving leukemia development by activating several signaling cascades associated with drug-resistance and poor outcome. These aspects and their possible therapeutic implications are highlighted in this review.


2017 ◽  
Vol 6 (3) ◽  
pp. e1274478 ◽  
Author(s):  
Margherita Boieri ◽  
Aina Ulvmoen ◽  
Amanda Sudworth ◽  
Clare Lendrem ◽  
Matthew Collin ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document