Chronic Myeloid Leukemia Stem Cells

2008 ◽  
Vol 26 (17) ◽  
pp. 2911-2915 ◽  
Author(s):  
Edward Kavalerchik ◽  
Daniel Goff ◽  
Catriona H.M. Jamieson

Although rare, chronic myeloid leukemia (CML) represents an important paradigm for understanding the molecular events leading to malignant transformation of primitive hematopoietic progenitors. CML was the first cancer to be associated with a defined genetic abnormality, BCR-ABL, that is necessary and sufficient for initiating chronic phase disease as well as the first cancer to be treated with molecular targeted therapy. Malignant progenitors or leukemia stem cells (LSCs) evolve as a result of both epigenetic and genetic events that alter hematopoietic progenitor differentiation, proliferation, survival, and self-renewal. LSCs are rare and divide less frequently, and thus, represent a reservoir for relapse and resistance to a molecularly targeted single agent. On subverting developmental processes normally responsible for maintaining robust life-long hematopoiesis, the LSCs are able to evade the majority of current cancer treatments that target rapidly dividing cells. Enthusiasm for the enormous success of tyrosine kinase inhibitors at controlling the chronic phase disease is tempered somewhat by the persistence of the LSC pool in the majority of the patients. Combined therapies targeting aberrant properties of LSC may obviate therapeutic resistance and relapse in advanced phase and therapeutically recalcitrant CML.

2020 ◽  
Vol 52 (10) ◽  
pp. 1663-1672
Author(s):  
Chun Shik Park ◽  
H. Daniel Lacorazza

Abstract Chronic myeloid leukemia is a hematological cancer driven by the oncoprotein BCR-ABL1, and lifelong treatment with tyrosine kinase inhibitors extends patient survival to nearly the life expectancy of the general population. Despite advances in the development of more potent tyrosine kinase inhibitors to induce a durable deep molecular response, more than half of patients relapse upon treatment discontinuation. This clinical finding supports the paradigm that leukemia stem cells feed the neoplasm, resist tyrosine kinase inhibition, and reactivate upon drug withdrawal depending on the fitness of the patient’s immune surveillance. This concept lends support to the idea that treatment-free remission is not achieved solely with tyrosine kinase inhibitors and that new molecular targets independent of BCR-ABL1 signaling are needed in order to develop adjuvant therapy to more efficiently eradicate the leukemia stem cell population responsible for chemoresistance and relapse. Future efforts must focus on the identification of new targets to support the discovery of potent and safe small molecules able to specifically eradicate the leukemic stem cell population. In this review, we briefly discuss molecular maintenance in leukemia stem cells in chronic myeloid leukemia and provide a more in-depth discussion of the dual-specificity kinase DYRK2, which has been identified as a novel actionable checkpoint in a critical leukemic network. DYRK2 controls the activation of p53 and proteasomal degradation of c-MYC, leading to impaired survival and self-renewal of leukemia stem cells; thus, pharmacological activation of DYRK2 as an adjuvant to standard therapy has the potential to induce treatment-free remission.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 5173-5173
Author(s):  
Elza Lomaia ◽  
Ekaterina Romanova ◽  
Larisa Girshova ◽  
Yulia Alexeeva ◽  
Eugenia Sbityakova ◽  
...  

Abstract Dramatic changes in overall survival of patients (pts) with chronic myeloid leukemia (CML) in chronic phase (CP) have occurred since tyrosine kinase inhibitors (TKIs) were implemented in the treatment strategy. But there are still many issues in therapy of advanced phase disease, especially in blastic phase (BP). Allogeneic stem cell transplantation (alloSCT) is still the only curative option for CML BP, so all efforts should be focused on bringing pts to alloSCT. Thus optimal approach to obtain at least stable hematologic response before alloSCT is needed. Since 2008, 14 pts (4 more pts with isolated extramedullary BP were not included) with CML BP were admitted to our clinic. These were 8 males and 6 females with a median age of 44 years (range; 21-63) at the time of BP. The types of BP were: biphenotypic (n=1), undifferentiated (n=1), myeloid (n=8) and lymphoid (n=4). All pts except 2 (1 with BP and 1 with accelerated phase) were initially diagnosed as CP. Median time from diagnosis to BP was 37 months (range; 0-83). Before BP all pts except 2 were pretreated with imatinib and 6 of them, after failing imatinib, received one or more new TKIs. First line therapy in BP was monotherapy with new TKI (n=5) or chemotherapy (“7+3”, “RACOP”, low doses of Ara-C, “Hyper-CVAD”, “Dexa+VCR”) with or w/o TKI (n=9). Responses are specified in table 1. FLAG regimen was subsequently given to 5 pts as second or more line therapy after failure of previous monoTKI (n=1) or Rx + TKI (n=4). Median time from BP to FLAG was 3,5 months (range; 1,5-21). The best response to FLAG therapy was complete hematologic (n=1), complete cytogenetic with (n=1) or w/o (n=1) major molecular response. There were no responses in 2 cases. All responders maintain their response after median follow up (FU) of 2 months (range; 1,5-5). All patients treated with FLAG are alive (2 after alloSCT, 3 pending alloSCT). Only 1 ptn reached alloSCT w/o any Rx after monoTKI. AlloSCT was successful in 4/5. Median FU time for patients alive after alloSCT is 12 months (range; 3,5-25). For whole group after a median FU of 14 months, 7/14 (50%) pts are alive, including 4 pts after alloSCT. Estimated 3-year overall survival for all pts is 54% (fig. 1). Conclusion All CML BP patients treated with TKIs alone lost their response in a short time. Responses were much more durable in pts treated with Rx +/- TKIs. FLAG regimen was effective even in pts with failure to previous Rx+TKIs. The majority of pts after alloSCT are alive. Chemotherapy, including FLAG with concomitant or subsequent TKIs, had advantage over monoTKI both in overall and progression free survival in CML BP. Disclosures: Lomaia: Novartis: Honoraria, Travel grants Other; Bristol-Myers Squibb: Honoraria, Travel grants, Travel grants Other. Zaritskey:University of Heidelberg: Research Funding.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3766-3766
Author(s):  
Paolo Strati ◽  
Hagop M. Kantarjian ◽  
Deborah A. Thomas ◽  
Susan M. O'Brien ◽  
Elias J. Jabbour ◽  
...  

Abstract Abstract 3766 Background: Chronic Myeloid Leukemia (CML) may progress at advanced phase at the rate of 1–1.5% per year. Blastic phase (BP) CML (defined by a bone marrow blast count >30%) can show lymphoid features in up to 20–30% of cases. With the use of single agent imatinib or dasatinib, median overall survival (OS) ranges between 7 and 11 months. Combination therapy may offer an improved outcome. We analyzed the outcome of patients (pts) with lymphoid BP-CML treated with hyperfractionated cyclophosphamide, vincristine, adriamycin, dexamethasone (HCVAD) plus imatinib or dasatinib. Methods: 32 pts with lymphoid BP-CML were treated at MD Anderson with HCVAD plus imatinib or dasatinib between 2000 and 2011. The starting dose of imatinib was 400 mg (2 pts), 600 mg (20 pts) and 800 mg (1 pt). The starting dose of dasatinib was 50 mg (1 pt), 100 mg (7 pts) and 140 mg (1 pt). Survival curves were calculated using Kaplan-Meier estimates and were compared using the log-rank test. Results: the median age was 48 (22–74) and 72% were male. Four (12%) pts had a de novo diagnosis, 21 (66%) were previously treated with a tyrosine kinase inhibitor (TKI) for chronic phase (CP) and 3 (9%) for BP. At diagnosis, median WBC was 23.4 (1.1–165.4) x109/L, hemoglobin 10.6 (6.3–16.4) g/dL, platelets 51 (6–526) x109/L, blasts 33 (0–91)%, basophils 0 (0–2)%, creatinine 1 (0.6–1.5) mg/dL, albumin 3.8 (2–4.7) g/dL, bilirubin 0.5 (0.2–3.4) mg/dL, alanine aminotransferase 34 (12–446) IU/L; on bone marrow, median blasts were 78 (26–97)%, basophils 0 (0–4)% and additional chromosomal aberrations (ACA) were found in 15/24 (62%) pts, affecting mostly chromosome (chr) 7 (60%), chr9 (40%), chr8 (33%) and chr1 (27%). Before BP diagnosis, median Philadelphia (Ph) positivity by FISH was 67% (0–96); 6/14 (43%) pts showed a Ph mutation (Y253H, T315I, Q252H, F317L, E255K, M244V) at time of progression to BP. Median time from CML diagnosis to BP was 18 (2–33) months, with no significant differences according to previous Ph FISH positivity or CML therapies. Imatinib was added to HCVAD in 23 pts and Dasatinib in 9. Complete Remission (CR) was obtained in 27 (84%) of them (78% with imatinib, 100% with dasatinib). Twenty-three of 27 (87%) CR were achieved after 1stcycle of induction. Early mortality (i.e., within 60 days) occurred in 3 pts. Patients received a median of 4 (1–8) cycles of HCVAD. At the time of CR, median BCR-ABL transcript levels were 1.7 (0–100). The levels decreased to a median of 0.01 (0–100) after 3–4 cycles of therapy; 7/27 (26%) pts achieved negative values of BCR-ABL transcripts after a median of 2 (1–4) months. Three (43%) of 7 pts who achieved complete molecular remission relapsed. MRD by flow cytometry became negative in 15/17 (88%) pts: 14 after induction, 1 after 2 months. Six (40%) of the pts with negative flow cytometry for MRD relapsed. Thirteen pts received SCT in remission: 4 relapsed and died after SCT. Median Progression Free Survival (PFS) was not reached and was longer among SCT recipients (p=0.03) and patients who had a negative flow cytometry at the time of CR (p<0.001). OS was 17 (7–27) months and was longer in patients with no more than 1 line of treatment for CP of CML, with ACA (p=0.01) and among SCT recipients (p<0.001). Among patients who had a CR, OS was longer if flow cytometry was negative at the time of CR (p=0.02) and if BCR-ABL transcript levels were < 1.7% (p=0.01) at the time of CR or <0.025% as best result (p=0.03). Conclusions: HCVAD plus imatinib or dasatinib is an effective regimen for pts with lymphoid BP CML, particularly when followed by SCT. ACA and less than 1 treatment for CML are positive prognostic factors. Better results are observed if negative flow cytometry and low levels of BCR-ABL transcripts are achieved with therapy. Disclosures: Ravandi: BMS: Honoraria, Research Funding.


2021 ◽  
Vol 10 (24) ◽  
pp. 5805
Author(s):  
Mohammad Houshmand ◽  
Alireza Kazemi ◽  
Ali Anjam Najmedini ◽  
Muhammad Shahzad Ali ◽  
Valentina Gaidano ◽  
...  

Chronic myeloid leukemia stem cells (CML LSCs) are a rare and quiescent population that are resistant to tyrosine kinase inhibitors (TKI). When TKI therapy is discontinued in CML patients in deep, sustained and apparently stable molecular remission, these cells in approximately half of the cases restart to grow, resuming the leukemic process. The elimination of these TKI resistant leukemic stem cells is therefore an essential step in increasing the percentage of those patients who can reach a successful long-term treatment free remission (TFR). The understanding of the biology of the LSCs and the identification of the differences, phenotypic and/or metabolic, that could eventually allow them to be distinguished from the normal hematopoietic stem cells (HSCs) are therefore important steps in designing strategies to target LSCs in a rather selective way, sparing the normal counterparts.


2022 ◽  
Vol 13 (1) ◽  
Author(s):  
Yosuke Tanaka ◽  
Reina Takeda ◽  
Tsuyoshi Fukushima ◽  
Keiko Mikami ◽  
Shun Tsuchiya ◽  
...  

AbstractLeukemia stem cells (LSCs) in chronic myeloid leukemia (CML) are quiescent, insensitive to BCR-ABL1 tyrosine kinase inhibitors (TKIs) and responsible for CML relapse. Therefore, eradicating quiescent CML LSCs is a major goal in CML therapy. Here, using a G0 marker (G0M), we narrow down CML LSCs as G0M- and CD27- double positive cells among the conventional CML LSCs. Whole transcriptome analysis reveals NF-κB activation via inflammatory signals in imatinib-insensitive quiescent CML LSCs. Blocking NF-κB signals by inhibitors of interleukin-1 receptor-associated kinase 1/4 (IRAK1/4 inhibitors) together with imatinib eliminates mouse and human CML LSCs. Intriguingly, IRAK1/4 inhibitors attenuate PD-L1 expression on CML LSCs, and blocking PD-L1 together with imatinib also effectively eliminates CML LSCs in the presence of T cell immunity. Thus, IRAK1/4 inhibitors can eliminate CML LSCs through inhibiting NF-κB activity and reducing PD-L1 expression. Collectively, the combination of TKIs and IRAK1/4 inhibitors is an attractive strategy to achieve a radical cure of CML.


2020 ◽  
Vol 90 ◽  
pp. 46-51.e2
Author(s):  
Yosuke Tanaka ◽  
Tsuyoshi Fukushima ◽  
Keiko Mikami ◽  
Keito Adachi ◽  
Tomofusa Fukuyama ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document