scholarly journals Kinase domain point mutations in Philadelphia chromosome-positive acute lymphoblastic leukemia emerge after therapy with BCR-ABL kinase inhibitors

Cancer ◽  
2008 ◽  
Vol 113 (5) ◽  
pp. 985-994 ◽  
Author(s):  
Dan Jones ◽  
Deborah Thomas ◽  
C. Cameron Yin ◽  
Susan O'Brien ◽  
Jorge E. Cortes ◽  
...  
Blood ◽  
2011 ◽  
Vol 117 (13) ◽  
pp. 3585-3595 ◽  
Author(s):  
Nidal Boulos ◽  
Heather L. Mulder ◽  
Christopher R. Calabrese ◽  
Jeffrey B. Morrison ◽  
Jerold E. Rehg ◽  
...  

Abstract The introduction of cultured p185BCR-ABL-expressing (p185+) Arf−/− pre-B cells into healthy syngeneic mice induces aggressive acute lymphoblastic leukemia (ALL) that genetically and phenotypically mimics the human disease. We adapted this high-throughput Philadelphia chromosome–positive (Ph+) ALL animal model for in vivo luminescent imaging to investigate disease progression, targeted therapeutic response, and ALL relapse in living mice. Mice bearing high leukemic burdens (simulating human Ph+ ALL at diagnosis) entered remission on maximally intensive, twice-daily dasatinib therapy, but invariably relapsed with disseminated and/or central nervous system disease. Although relapse was frequently accompanied by the eventual appearance of leukemic clones harboring BCR-ABL kinase domain (KD) mutations that confer drug resistance, their clonal emergence required prolonged dasatinib exposure. KD P-loop mutations predominated in mice receiving less intensive therapy, whereas high-dose treatment selected for T315I “gatekeeper” mutations resistant to all 3 Food and Drug Administration–approved BCR-ABL kinase inhibitors. The addition of dexamethasone and/or L-asparaginase to reduced-intensity dasatinib therapy improved long-term survival of the majority of mice that received all 3 drugs. Although non–tumor-cell–autonomous mechanisms can prevent full eradication of dasatinib-refractory ALL in this clinically relevant model, the emergence of resistance to BCR-ABL kinase inhibitors can be effectively circumvented by the addition of “conventional” chemotherapeutic agents with alternate antileukemic mechanisms of action.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1831-1831
Author(s):  
Dan Jones ◽  
Rayjalakshmi Luthra ◽  
Hagop M. Kantarjian ◽  
Megan Breeden ◽  
Susan O’Brien ◽  
...  

Abstract Bcr-Abl kinase domain (KD) point mutations are detected in the dominant clone(s) in approximately 45% of CML at the time of disease resistance, developing after an average of 20–35 months of imatinib therapy. However, low numbers of Philadelphia chromosome (Ph)+ tumor cells with KD mutations could be present at earlier timepoints providing a pool of potential resistant subclones. Since current therapy of Ph+ ALL relies on imatinib maintenance therapy, the pattern of Bcr-Abl KD mutations in this tumor is an important and understudied phenomenon. We assessed the frequency and levels of Bcr-Abl KD mutations at different points in ALL, including at diagnosis, upon relapse and following salvage therapy with kinase inhibitors. We performed Bcr-Abl KD mutational analysis by direct sequencing in 25 cases of Ph+ ALL at the time of diagnosis and 25 cases upon disease persistence/relapse. For comparison, we analyzed 22 cases of lymphoid blast crisis of CML (LyBC), most of which transformed following long-term imatinib monotherapy. To track the emergence of mutated clones, we also performed more sensitive analysis for the T315I mutation by pyrosequencing (5% sensitivity) and allele-specific oligonucleotide probe (ASO) PCR (1:500 sensitivity). KD mutations were not seen by direct sequencing in ALL cases at diagnosis. The T315I mutation was also not detected by pyrosequencing (n =25) or ASO-PCR (n = 10) in newly diagnosed ALL. In contrast, Bcr-Abl KD mutations (Y253H in 3, Q252H, T315I, F317L, E355Q, H396R in 1 each) were seen in 8 of 25 (32%) relapsed/persistent ALL, occurring in patients who had been receiving imatinib for a median of 14 months (range 2–26). An additional 3 patients treated with dasatinib or nilotinib for relapse subsequently developed KD mutations (T315I and Y253H, and F317L) after 1, 4 and 9 months of second therapy. KD mutations were seen in 16 of 22 (73%) patients with lymphoid blast crisis, including T315I in 7, E255K and M244V in 2 each, and Y253H, V299L, F311I, E355G, F359V in 1 each. All KD mutations in LyBC developed following imatinib or nilotinib therapy. As with CML, kinase inhibitor therapy particularly in the relapse/salvage setting is the primary risk factor for emergence of Bcr-Abl KD mutations in Ph+ ALL. There is a high frequency of Bcr-Abl KD mutations associated the lymphoid transformation of CML. However, Bcr-Abl KD mutations develop more rapidly in persistent or relapsed Ph+ ALL than in CML and there is a higher frequency of Y253H mutations noted. These findings will likely have consequences for the timing and dosages of imatinib and other kinase inhibitors in maintenance and relapsed ALL regimens.


2021 ◽  
Author(s):  
Rongrong Chen ◽  
Lulu Wang ◽  
Ting Shi ◽  
Lixia Zhu ◽  
Xiujin Ye

Abstract Tyrosine kinase inhibitors (TKI) resistance is a predominant cause of therapy failure of Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL). The mutation in the BCR-ABL kinase domain, as the indicator of drug resistance, suggested recurrence and poor prognosis. Our data aimed to evaluate the factors having significant roles in the prognostic value of the acquired BCR-ABL mutations. Three hundred and thirteen Ph+ ALL patients (64 patients with BCR-ABL mutations and 171 patients without BCR-ABL mutations) were enrolled in this study. Univariate and multivariable logistic regression analysis was used to evaluate the relationship between clinical features and BCR-ABL KD mutation. Univariate analysis showed that the patients with higher WBC (P=0.005), MMR after first induced chemotherapy (P=0.053), and Age≥45 years (P=0.072) were more likely to occur BCR-ABL mutations during the treatments. Finally, multivariable logistic regression indicated that higher WBC (≥22×109/L) was an independent risk of resistance to TKI and raise the possibility of KD mutations in Ph+ ALL patients. The happen of BCR-ABL mutations mean poor prognosis with shorter OS (P=0.000) and allogeneic hematopoietic stem-cell transplantation (allo-HSCT) would improve the long-term survival (OS: P=0.000). Conversely, for the defined low-risk populations, no significant difference was found between the transplant group and the non-transplant group in subgroup analysis, providing a rationale to potentially avoid allo-HSCT in this subgroup of patients.


Blood ◽  
2020 ◽  
Vol 136 (15) ◽  
pp. 1786-1789 ◽  
Author(s):  
Neeraj Saini ◽  
David Marin ◽  
Celina Ledesma ◽  
Ruby Delgado ◽  
Gabriela Rondon ◽  
...  

How to best use tyrosine kinase inhibitors (TKIs) of BCR-ABL after allogeneic stem cell transplantation for Philadelphia chromosome–positive acute lymphoblastic leukemia (ALL) is unknown but will almost certainly not be addressed by a definitive randomized trial. Saini and colleagues provide a large body of observational data to reinforce earlier circumstantial evidence favoring prophylactic use of TKIs for at least 2 years posttransplant.


Chemotherapy ◽  
2019 ◽  
Vol 64 (2) ◽  
pp. 81-93 ◽  
Author(s):  
Yingying Ma ◽  
Quanchao Zhang ◽  
Peiyan Kong ◽  
Jingkang Xiong ◽  
Xi Zhang ◽  
...  

With the advent of tyrosine kinase inhibitors (TKIs), the treatment of Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL) has entered a new era. The efficacy of TKIs compared with other ALL treatment options is emphasized by a rapid increase in the number of TKI clinical trials. Subsequently, the use of traditional approaches, such as combined chemotherapy and even allogeneic hematopoietic stem cell transplantation (allo-HSCT), for the treatment of ALL is being challenged in the clinic. In light of the increased use of TKIs in the clinic, several questions have been raised. First, is it necessary to use intensive chemotherapy during the induction course of therapy to achieve a minimal residual disease (MRD)-negative status? Must a patient reach a complete molecular response/major molecular response before receiving allo-HSCT? Does MRD status affect long-term survival after allo-HSCT? Is auto-HSCT an appropriate alternative for allo-HSCT in those Ph+ ALL patients who lack suitable donors? Here, we review the recent literature in an attempt to summarize the current status of TKI usage in the clinic, including several new therapeutic approaches, provide answers for the above questions, and speculate on the future direction of TKI utilization for the treatment of Ph+ ALL patients.


Sign in / Sign up

Export Citation Format

Share Document