Ph-Negative Hematopoiesis Emerging After Successful Treatment of Chronic Myeloid Leukemia Displays Severe and Persistent Telomeric Loss Which Is Particularly Prominent in Patients with Acquired Cytogenetic Abnormalities.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2164-2164
Author(s):  
Chiara Lobetti Bodoni ◽  
Dario Ferrero ◽  
Elisa Genuardi ◽  
Daniela Sia ◽  
Mariella Genuardi ◽  
...  

Abstract Abstract 2164 Poster Board II-141 Background and Aims. Little is known on the functional and genetic integrity of Ph-negative hematopoietic cells (HC) repopulating the bone marrow after successful chronic myeloid leukemia (CML) treatment, although the frequent detection of cytogenetic abnormalities (CA), reminiscent of those seen in myelodysplastic syndromes (MDS), suggests the presence of functional and genetic defects. Telomere attrition represents a useful marker of proliferative and oxidative stress and might provide useful insights to monitor the genetic integrity of the hematopoietic compartment. This approach has been used in combination with a functional study of short and long term progenitors. Patients and methods. We investigated 78 CML patients with persistent (>12 months) complete cytogenetic remission (CCR). Median age was 64 (23-88), M/F ratio was 1.5. Median time from diagnosis and CCR were 64 months (25-915) and 39 months (12-150) respectively. Sokal score was low in 36 patients, intermediate in 28, and high in 14. Six patients were received IFN only, 45 Imatinib only, while 27 were currently on Imatinib, but received previous treatment with INF and/or chemotherapy. Complete and partial molecular responders were 35 and 28 respectively. Fifteen patients had acquired CA (del7: 4 patients, + 8: 5 patients, del5q: 2 patients, del or +Y: 2 patients, and 2 patients had other CA). Short term progenitors (CFU-GM, BFU-E CFU-Mix) and long-term culture-initiating cells (LTC-ICs)(Sutherland HJ et al Blood 1994) have been performed on 30 patients (requiring bone marrow examination for clinical purposes). Telomere length (TRF-L) analysis was performed by Southern Blotting as previously described (Ladetto M et Al, Blood 2004), both on polymorphonucleates (PMN) and on monocyte-depleted PBMC (MD-PBMC) (Rocci et al Exp Hematol 2007) to monitor both the myeloid and lymphoid compartment. Sixty four patients were assessed on repeated samples to monitor the kinetics of telomeric loss (median time 8 months, range 6-20). A control database of 109 healthy subjects has been used for comparison. Results. Ph-negative HC of CML patients were functionally impaired compared to controls, with reduced number of CFU-Mix (median 2,62 vs 4, p=0,010), CFU-GM (median 99,5 vs 181, p<0,001) and particularly of LTC-IC (median 88 vs 198, p<0,001). PMN from CML patients showed a major erosion of their telomeric DNA (median telomeric loss 1536 bp p<0.001, figure 1A). This finding was even more striking in patients with acquired CA, who showed a median TRF-L loss of 1900 bp (p<0.001) compared to healthy subjects, and 500 bp compared to other CML patients (p=0.030, figure 1B). Interestingly telomere attrition was less pronounced in the 4 patients with del or +Y and del5q, compared to those with other CA, such as del7 and +8. Telomeric erosion is more severe in younger CML patients, resulting in loss of the association between TRF-L and age, typically seen in healthy subjects. Telomere shortening was observed regardless of the use of TK inhibitors and chemotherapy. We found no correlation between TRF-L and clinical and demographic parameters. When a multivariate analysis on patients and healthy controls was performed, the presence of CML resulted a stronger predictor of telomeric damage compared to age. Analysis of TRF-L kinetics on the whole population over time showed substantial stability or modest physiological shortening in the majority of patients. In none of the patients a relevant recovery of TRF-L over time was noticed. However in 16 (25%) patients a non-physiological telomeric loss was observed (>400bp obtained by considering maximal physiological loss plus technical variability of the assay) (Figure 2). Interestingly the four patients with the most extreme telomeric loss (>1000 bp/year) showed evidence of either CA or impaired hematopoietic fuction by colony assays. Moreover one of these patients progressed to an overt MDS six month after the second determination. Conclusion. Ph-negative HC repopulating the bone marrow after successful CML treatment: i) have major defects in their functional performances; ii) display severe telomeric loss (roughly comparable to 31 years of physiological aging), which is more pronounced in patients with CA. Moreover the lack of telomeric recovery over time and the presence of a subgroup of patients with ongoing accelerated non-physiological telomeric attrition suggest the need of strict monitoring of the long-term performances and genetic stability of Ph-negative hematopoiesis in CML patients. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 4844-4844
Author(s):  
Hana Klamova ◽  
Jana Brezinova ◽  
Kyra Michalova ◽  
Zuzana Zemanova ◽  
Marek Trneny

Abstract Cytogenetic clonal evolution (CE) - the presence of cytogenetic abnormalities in addition to the Ph chromosome in chronic myeloid leukemia (Ph+ CML) is a known poor prognostic factor associated with disease progression. Occurence of additional cytogenetic abnormalities in both Ph positive and Ph negative mitoses was also described in imatinib treated CML patients and was associated with occuring therapy resistance. The long - term significance is so far poorly understood. Objective. To monitor cytogenetic abnormalities in chronic phase CML patients on imatinib treatment, following long-term interferon alfa (IFN) or hydroxyurea treatment. To compare the haematological disease progression in patients with or without cytogenetic evolution Patients and methods: Cytogenetic evolution was analyzed in 57 patients (median age 56, range 18–73) treated with imatinib in chronic phase, following interferon resistance or intolerance. The duration of IFN application was 22 months (range 3 – 46 months), duration of imatinib treatment was 16 months (range 6 – 55 months). Cytogenetic abnormalities were detected by conventional cytogenetics - caryotype analysis and fluorescence in situ hybridisation (FISH). Results: Complete cytogenetic remission was accomplished in 55 of 57 pts (96%) on imatinib, significant or complete cytogenetic response was observed in 36 of 57 patients (66%). Cytogenetic evolution was observed in 11 patients (19%) treated with imatinib: in the Ph+ clone (9 cases) and in the Ph− clone (2 cases). Median duration of imatinib treatment before the CE identification was 16 months (range 7–36 months). The most common additional abnormality was trisomy 8 (8 pts), second Ph chromosome (4 pts), and del (17) (4 pts). In 5 cases we observed the simultaneous occurence of two different cytogenetic abnormalities. Haematological progression was observed in 7 of 11 patients (63%) following 2 – 22 months imatinib treatment (median 9 months). 5 pts (46%) exited. Six patients live 8–22 months from the detection of cytogenetic evolution. Secondary malignancy was diagnosed in 1 patient. In the group of patients without cytogenetic evolution haematological progression was observed only in 9 of 46 (19.5%) cases, 4 patients died (14.3%). Conclusion: The role of IM concerning the cytogenetic evolution occurence in CML patients is not so far clear, the suppression of the Ph+ clone could enhance the proliferation of resistant ones. In our group of patients CE was documented in 11 patients (19%), in both Ph+ and Ph− cells. Significantly higher was the risk of haematological progression. CML patients treated with imatinib should be regularly monitored with conventional cytogenetic techniques, not only to follow the decrease in the proportion of Ph-positive cells, but also to look for new especially Ph-negative clonal chromosomal abnormalities. A longer follow-up time and systematic monitoring of cytogenetics is needed to establish the prognostic impact of clonal evolution in CML patients treated with imatinib.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1999-1999
Author(s):  
Monica Bocchia ◽  
Elisabetta Abruzzese ◽  
Micaela Ippoliti ◽  
Simona Calabrese ◽  
Alessandro Gozzetti ◽  
...  

Abstract Although the success of imatinib mesylate therapy represents an exciting advance in targeted cancer therapy, it has still to be determined whether responses to this p210 inhibitor in chronic myeloid leukemia (CML) patients will be durable. In fact most of clinical studies agree on the evidence of a persistent molecular disease in the majority of treated patients and altough the absolute level of bcr-abl transcript may vary over the treatment, yet a molecular complete response is of rare observation. In addition, discontinuation of imatinib exerts always in rapid loss of response. In accordance to this the persistence of malignant progenitors in patients in complete cytogenetic response (CCR) after short term imatinib treatment, has been recently demonstrated. In particular, Bathia et al. showed in 12/15 patients studied after a median time of 10 months of imatinib treatment a median of 11% of residual CML CD34+ progenitors in the bone marrow (by FISH Dual Fusion bcr/abl analysis)while only 3/15 patients had no detectable residual CD34+ cells. Less is known about residual Ph+/CD34+ cells surviving after a prolonged therapy with this targeting drug. Thus, we evaluated the amount of bone marrow residual CD34+ cells in 17 CML patients in stable CCR after a long lasting treatment with imatinib. At the time of evaluation, the patients were on conventional dose (400mg) Imatinib for a median time of 48 months (range 36–58 months) having achieved a CCR status (conventionally defined as the complete absence of t(9;22) on caryotypic analysis) within 3 to 6 months of treatment. However all of them still showed molecular disease as detected by nested RT-PCR. Bone marrow CD34+ cell-enriched populations were selected from mononuclear cells using immunomagnetic column separation and were evaluated after cytospin by FISH using a bcr-abl Dual Color Extra Signal Probe(LSI bcr-abl ES, Vysis), that is able to detect bcr-abl fusion in interphase nuclei with a false positive signal rate close to 0. A minimum of 100 CD34+ nuclei per each sample were evaluated. Interestingly, in 8/17 patients no Ph+/CD34+ cells were detected, while in the remaining 9/17 patients a median of 2% (range 0.5–11%) of bcr-abl positive progenitors were still observed. In this small selected serie of patients prolonged treatment with imatinib appears to be correlated with a lower, yet detectable, amount of residual bone marrow Ph+/CD34+ cells when compared to previously published data. This result could be partly explained with the different specificity and sensitivity of the probe used (bcr/abl ES<1% false positive; bcr-abl Dual Fusion 8–10% false positive) The clinical significance of these data as well as the role of this cell target to monitor minimal residual disease in CML needs to be evaluated on a larger serie of patients.


Blood ◽  
2010 ◽  
Vol 115 (16) ◽  
pp. 3185-3195 ◽  
Author(s):  
Mirle Schemionek ◽  
Christian Elling ◽  
Ulrich Steidl ◽  
Nicole Bäumer ◽  
Ashley Hamilton ◽  
...  

Abstract In a previously developed inducible transgenic mouse model of chronic myeloid leukemia, we now demonstrate that the disease is transplantable using BCR-ABL+ Lin−Sca-1+c-kit+ (LSK) cells. Interestingly, the phenotype is more severe when unfractionated bone marrow cells are transplanted, yet neither progenitor cells (Lin−Sca-1−c-kit+), nor mature granulocytes (CD11b+Gr-1+), nor potential stem cell niche cells (CD45−Ter119−) are able to transmit the disease or alter the phenotype. The phenotype is largely independent of BCR-ABL priming before transplantation. However, prolonged BCR-ABL expression abrogates the potential of LSK cells to induce full-blown disease in secondary recipients and increases the fraction of multipotent progenitor cells at the expense of long-term hematopoietic stem cells (LT-HSCs) in the bone marrow. BCR-ABL alters the expression of genes involved in proliferation, survival, and hematopoietic development, probably contributing to the reduced LT-HSC frequency within BCR-ABL+ LSK cells. Reversion of BCR-ABL, or treatment with imatinib, eradicates mature cells, whereas leukemic stem cells persist, giving rise to relapsed chronic myeloid leukemia on reinduction of BCR-ABL, or imatinib withdrawal. Our results suggest that BCR-ABL induces differentiation of LT-HSCs and decreases their self-renewal capacity.


Blood ◽  
1993 ◽  
Vol 82 (10) ◽  
pp. 3211-3219 ◽  
Author(s):  
W Arcese ◽  
JM Goldman ◽  
E D'Arcangelo ◽  
A Schattenberg ◽  
A Nardi ◽  
...  

We studied the clinical course of 130 chronic myeloid leukemia (CML) patients (89 males and 41 females) in the European Bone Marrow Transplantation Group (EBMT) registry who received transplants before January 1, 1988 and who subsequently had evidence of recurrent leukemia. All patients had received a pretransplant conditioning regimen including total body irradiation (TBI). The first evidence of relapse was cytogenetic only in 74 (57%) patients and hematologic in 56 (43%). The overall actuarial survival from relapse was 36% at 6 years, with a significantly higher proportion of survivors among female patients (53% v 30%; P < .002). In univariate analysis, the 6-year probability of survival was 52% for patients with cytogenetic relapse and 30% for patients relapsing in chronic phase (CP), while no patient who relapsed in advanced phase (AP or BC) survived more than 3.5 years from relapse (P < .0001). The actuarial survival of patients relapsing before 6 months, between 6 and 12 months, and later than 12 months after transplant was 27%, 26%, and 45%, respectively (P < .002). Among patients with cytogenetic relapse, partial or complete disappearance of Ph-positive cells occurred in 40% of untreated patients and in 42% of those treated with interferon (IFN). However, IFN therapy significantly delayed progression toward hematologic disease. Cytogenetic responses were observed in 25% of patients who received IFN for relapse into CP, while only one minor cytogenetic response was reported in patients on conventional chemotherapy. For patients presenting with cytogenetic relapse as well as for those in hematologic relapse, IFN therapy significantly improved the 2-year probability of survival. However, long-term survival for IFN-treated patients in either group was not different from long-term survival in comparable patients not receiving IFN therapy. Twenty-nine patients of this series underwent a second bone marrow transplant (BMT) and the projected survival at 4 years after the second transplant is 28%. In multivariate Cox regression analysis, four factors remained significantly associated with survival: disease phase at relapse (P < .0001), duration of time interval from BMT to relapse (P = .0001), interferon therapy at relapse (P = .0024), and patient sex (P = .0032). This retrospective study provides evidence that some patients who relapse after BMT may benefit from treatment with IFN; a second BMT may offer the chance of cure. Data from this analysis may be useful in designing future prospective trials on posttransplant CML relapse.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 52-53
Author(s):  
Elzbieta Szczepanek ◽  
Ositadima Chukwu ◽  
Magdalena Kaminska ◽  
Hubert Wysoglad ◽  
Agnieszka Cenda ◽  
...  

Introduction Imatinib, approved as first-line treatment for patients with newly diagnosed chronic myeloid leukemia (CML) by FDA approximately 20 years ago, revolutionized the treatment of this disease. The life expectancy of newly diagnosed patients with CML has been approaching that of the global population. Second generation TKI (2GTKI) administered as a frontline therapy can induce deeper and faster molecular responses in a higher percentage of patients, however, the overall survival is comparable to that achieved with imatinib. To investigate the outcomes of long-lasting therapy with imatinib administered as initial therapy, we analyzed patients with CML who received imatinib as initial therapy at our institution starting from 2001. Methods We retrospectively analyzed long term outcomes of 267 patients treated with imatinib 400 mg at the Department of Hematology, Jagiellonian University Medical College, Cracow, Poland from 2001. Data from medical records were collected and statistical analysis was performed using R software (R version 4.0.2). Results The median age was 53.5 (16 to 88 years), 129 of patients (pts) (48.31%) were female. At the time of this analysis, 99 pts (37.08 %) remained on imatinib with the median dose at last follow-up (FU) 400 mg. The mean initial dose of imatinib was 410.4 mg/d. Imatinib dose was increased in 53 pts (19.85%), up to 800mg and up to 600 mg in 3 pts (1.13%), and in 49 pts (18.35%) respectively. The mean maximal dose was 465.2 mg. At baseline 124 pts (46.44%) had comorbidities: 79 pts (29.59%) vascular/cardiac, 11 pts (4.12%) renal, and 101 pts (37.33%) other comorbidities. 15 patients (5.63%) had prior malignancies, newly diagnosed malignancies occurred among 11 (4.12%) pts on imatinib. The median follow-up time was 11.37 years (range from 2 months to 19.5 years). 168 pts (62.92%) discontinued imatinib permanently, the median time to imatinib discontinuation was 2.02 years. Among them, 123 pts (71.93 %) switched imatinib to 2GTKI- 79 pts (29.59%) to dasatinib, 68 pts (25.47%) to nilotinib, and 14 pts (5.24%) to bosutinib. During the following treatment 87 pts (32,58%) received one 2GTKI, 33 pts (12.36%) two, and 3 pts (1.12%) more than two 2GTKIs. The main reasons for imatinib therapy discontinuation were intolerance (87 pts, 32.58%) and disease progression (90 pts, 33.71%). The median time to the imatinib discontinuation due to its intolerance was 2 years. Adverse events (AEs) during imatinib therapy were as follows: cardiac/vascular AEs in 22 pts (8.24%), renal in 42 pts (15.73%), hematologic in 43 pts (16.10%), and other in 189 pts (70.79%). Overall, 28 patients died (10.5%), 7 pts (2.2%) transformed to blast phase, 9 pts (3.37%) underwent allo-HSCT. Estimated OS for patients that remained on imatinib for the whole observation period for 15 and 18 years was: 80.2%, and 64.1% respectively (Figure 1). Median follow-up time for patients who continued imatinib was 7.91 years. Intention to treat (ITT) analysis available for 99 pts (37.08%) revealed ITT responses at three months, one, five, ten and fifteen years: 50.52%, 77.4%, 86.25%, 90.28%, 100% for CCyR, 32.99%, 58.07%, 80%, 86.11%, 100% for MMR, 11.34%, 20.44%, 63.75%, 63.89%, 90% for MR4, 2.06%, 12.91%, 35%, 38.89%, 70% for MR4.5, 2.06%, 7.53%, 26.25%, 33.33%, 50% for CMR (undetectable transcripts with ≥100,000 copies ABL) (Table 1). The overall best response rates (at any time) for these 99 pts was 4.04% for MCyr, 5.05% for CCyR, 11.11% for MMR, 14.14% for MR4, 9.09% for MR4.5, 49.49% for CMR. Conclusion The analysis of long-term therapy with imatinib showed that the efficacy of imatinib persisted over time and that long-term administration of imatinib was associated with low rate of late toxic effects. Disclosures Sacha: Novartis: Consultancy, Honoraria, Speakers Bureau; Pfizer: Consultancy, Honoraria, Speakers Bureau; Adamed: Consultancy, Honoraria; Incyte: Consultancy, Honoraria, Speakers Bureau; Bristol-Myers Squibb Company: Consultancy, Honoraria, Speakers Bureau.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3183-3183
Author(s):  
Chiara Lobetti Bodoni ◽  
Dario Ferrero ◽  
Elisa Genuardi ◽  
Daniela Sia ◽  
Mariella Genuardi ◽  
...  

Abstract Background. Most chronic myelogenous leukemia (CML) patients (pts) restore non-neoplastic hematopoiesis following treatment with tyrosine kinase (TK) inhibitors. However little is presently known on the functional and genetic integrity of Ph-negative hematopoietic cells (HC) repopulating the bone marrow after successful treatment. Indeed, the frequent detection of cytogenetic abnormalities (CA) reminiscent of those seen in myelodysplastic syndromes suggests the potential presence of functional and genetic defects. These issues have been addressed using short and long term HC cultures and telomere restriction fragment length (TRF-L) analysis, which is considered a reliable marker of proliferative and oxidative damage. Patients and methods. We investigated 71 CML pts in stable complete cytogenetic remission (CR) (CR had to be documented at least one year before the analysis). 62 pts were treated with Imatinib and 10 with α-interferon associated or not to ara-C. Median age was 64 (23–88), M/F ratio was 1.5, median time from diagnosis and from complete CR were 58 (7–915), and 40 months (12–150). 31 pts had low Sokal score, 27 intermediate, and 13 high. Complete and partial molecular responders were 35 and 21, respectively. 6 pts showed evidence of acquired CA in Ph-negative HC. TRF-L analysis was performed by Southern Blotting as previously described (Ladetto M et Al, Blood 2004), both on polymorphonucleates (PMN) and on monocyte-depleted PBMC (MD-PBMC) (as described by Rocci et al Exp Hematol 2007) to monitor both the myeloid and lymphoid compartment. Colony-forming unit granulocyte-macrophage (CFU-GM), burst-forming unit erythroid (BFU-E) and colony forming unit-mix (CFU-Mix) along with long-term culture-initiating cells (LTC-ICs) have been so far performed on 30 patients, using bone marrow mononuclear cells as previously described (Sutherland HJ et al Blood 1994). For both TRF-L and cell culture studies a control database of 86 healthy subjects has been used for comparison. Results. PMN from CML patients showed a striking erosion of their telomeric DNA (figure 1A). Also MD-PBMC showed a degree of telomere shortening although the finding was much less pronounced (mean telomeric loss in PMN 1683 pb p&lt;0.001; in MD-PBMC: 323 pb, p=0.04) We found no correlation between TRF-L and previously mentioned clinical parameters. Telomeric erosion is more severe in younger CML pts, resulting in loss of the association between TRF-L and age, typically seen in healthy subjects (figure 1B) Telomere shortening was observed regardless of the use of TK inhibitors. When a multivariate analysis on pts and healthy controls was performed, the presence of CML resulted a stronger predictor of telomeric damage compared to age. We found no correlation between TRF-L and previously mentioned clinical and demographic parameters. Telomeric erosion show no evidence of recovery on 40 follow-up samples taken after a median time of 10 months (range 6–13). Moreover, Ph-negative HC of CML pts were functionally impaired compared to controls with reduced numbers of CFU-Mix (median 2,62 vs 4, p=0,01), CFU-GM (median 99,5 vs 181, p&lt;0,0001) and particularly of LTC-IC (median 88 vs 198, p&lt;0,0001) (figure 1C). Discussion. Ph-negative HC repopulating the bone marrow after successful CML treatment display severe telomeric DNA erosion, roughly comparable to 34 years of physiological aging. Moreover they display major defects in their functional performances. These findings, underline the need of additional investigation and careful clinical monitoring of the Ph-negative haemopoietic compartment in these subjects. Figure Figure


2020 ◽  
Vol 58 (8) ◽  
pp. 1214-1222
Author(s):  
Georg Greiner ◽  
Franz Ratzinger ◽  
Michael Gurbisz ◽  
Nadine Witzeneder ◽  
Hossein Taghizadeh ◽  
...  

AbstractBackgroundMonitoring of molecular response (MR) using quantitative polymerase chain reaction (PCR) for BCR-ABL1 is a pivotal tool for guiding tyrosine kinase inhibitor therapy and the long-term follow-up of patients with chronic myeloid leukemia (CML). Results of MR monitoring are standardized according to the International Scale (IS), and specific time-dependent molecular milestones for definition of optimal response and treatment failure have been included in treatment recommendations. The common practice to use peripheral blood (PB) instead of bone marrow (BM) aspirate to monitor the MR monitoring in CML has been questioned. Some studies described differences between BCR-ABL1 levels in paired PB and BM specimens.MethodsWe examined 631 paired PB and BM samples from 283 CML patients in a retrospective single-center study using an IS normalized quantitative reverse transcription (qRT)-PCR assay for quantification of BCR-ABL1IS.ResultsA good overall concordance of BCR-ABL1IS results was found, a systematic tendency towards higher BCR-ABL1IS levels in PB was observed in samples of CML patients in a major MR. This difference was most pronounced in patients treated with imatinib for at least 1 year. Importantly, the difference resulted in a significantly lower rate of deep MR when BCR-ABL1IS was assessed in the PB compared to BM aspirates.ConclusionsIn summary, our data suggest that the classification of deep MR in patients with CML is more stringent in PB than in BM. Our study supports the current practice to primarily use PB for long-term molecular follow-up monitoring in CML.


2019 ◽  
Vol 3 (3) ◽  
pp. 370-374 ◽  
Author(s):  
Naranie Shanmuganathan ◽  
Susan Branford ◽  
Timothy P. Hughes ◽  
Devendra Hiwase

Key Points Bone marrow fibrosis may be a late reversible toxicity of high-dose imatinib therapy in chronic myeloid leukemia.


2009 ◽  
Vol 50 (1) ◽  
pp. 114-118 ◽  
Author(s):  
Francesca Palandri ◽  
Nicoletta Testoni ◽  
Simona Luatti ◽  
Giulia Marzocchi ◽  
Carmen Baldazzi ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document