Comprehensive Characterization of Cytogenetic and Mutational Analysis of Acute Promyelocytic Leukemia: Is PML-Rara Everything?

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1404-1404
Author(s):  
Hassan Awada ◽  
Jibran Durrani ◽  
Tariq Zuheir Kewan ◽  
Ashwin Kishtagari ◽  
Valeria Visconte ◽  
...  

Acute promyelocytic leukemia (APL) is characterized by PML-RARA fusion caused by the t(15;17)(q24;q21) translocation. Although PML-RARA fusion explains the dedifferentiation in most of APL patients, it still does not entirely represent the unique cause of all the clinical manifestations of the disease failing to determine the full leukemic phenotype. Up to 40 % of APL patients have an additional chromosomal abnormality other than PML-RARA. Murine studies have reported that additional cytogenetic abnormalities and secondary somatic mutations (for instance FLT3-ITD) might contribute to leukemia progression. Indeed, mice expressing mutant PML-RARA develop definitive leukemia after one year, suggesting that additional hits are required for transformation. Moreover, no distinct genetic signature has been characterized by next generation sequencing (NGS). This confirms that no gene has been reproducibly identified. APL respond to all-trans retinoic acid (ATRA) in the great majority of patients. However, one quarter of APL develop ATRA resistance suggesting that additional secondary chromosomal abnormalities might be evolving resistance. Combination of low dose arsenic, modify certain epigenetics, with ATRA decreased resistance potential and improved response. In the line with other possible factors involved in ATRA resistance, is the broad nature of the targets of ATRA. A molecular core network of ATRA's targets has been clustered in differentiation, growth factors and nuclear receptors possibly cooperating with PML-RARA and additional chromosomal abnormalities. Herein, we aimed to characterize the gene mutations and chromosomal abnormalities playing key roles in cellular differentiation and epigenetic regulation and to correlate the occurrence of these alterations with treatment response and survival outcomes in APL. We took advantage of a large cohort of APL patients (n=145). Median age of the cohort was 50 yrs (19-85); equal gender distribution; median blood counts were: [WBC 6.2 x 109/L (0.4-155); 37% had leukopenia], hemoglobin [9.8 g/dL (2.7-16.2); 32% had anemia] and platelets [29 x 109/L (range of 0-228); 93% had thrombocytopenia]. In terms of karyotype, 15% of the patients carried +8, 7% had complex karyotyping (≥3 cytogenetic abnormalities), 2% had -7/del (7q) or del (12p), 1% had -17/del(17p), and 1 patient had -5. Mutational analysis of 30 genes panel, identified 141 mutations carried by 65% (94/145) of APL patients. The most frequent mutations were observed in FLT3-ITD (61/143; 43%), WT1 (26/139; 23%), and ASXL1 (7/136; 5%) genes. Less frequent mutations were found in 3.7% of CEBPA, KRAS, and NRAS genes as well as in CBL, EZH2, TET2 (3% each) genes. Additionally, we noted that all mutations were recurrent in specific functional pathways and patients carried mutations in more than 1 gene of the same pathway. Of note, cell signaling and proliferation genes (CBL, NRAS, KRAS, KIT, FLT3) were the most frequently mutated (77/141, 55%) and impacted OS (HR: 1.7, P=0.02). Moreover, transcriptional factors which are often mutated in AML (e.g. CEBPA, TP53, NPM1, RUNX1, WT1) as well as major determinants of cell's fate were markedly mutated (38/141, 27%) suggesting that genetic impairment of signaling and transcription might contribute to the lack of differentiation observed in APL phenotypes. Mutations in epigenetic genes and histone methyltransferases (ASXL1, BCORs, DNMT3A, EZH2, IDH1/2, TET2) were also found in 18/141 (13%) while genes regulating cell proliferation and RAS family (CBL, NRAS, KRAS, NPM1) were enriched in 16/132 (12%) of APL cohort. We then analyzed the genetic picture of remission (APLRm, n=131, 90%) and relapsed (APLR, 1sr relapsed to ATRA, n=14, 10%) patients. Acknowledging the low number of APLR, we observed that molecular mutations did not make a key difference in APLRvs. APLRm [except for a complete lack of mutations in epigenetic pathways (0% vs. 13%)]. Contrarily, specific cytogenetic abnormalities were more common in APLR compared to APLRm as the case of +8 (36% vs. 11%; P= .02) and -17/del(17p) (2/14 vs. 0/131; P= .008). In sum, our study demonstrates that PML-RARA might be accompanied by additional acquired chromosomal change with a variety of genetic mutations in key pathways driving cellular differentiation. These molecular/ cytogenetic associations could determine resistance to ATRA and overall APL patients' survival. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 3-3
Author(s):  
Gustavo Milone ◽  
Samuel Sarmiento Doncel ◽  
Carol Agudelo Rico ◽  
Fabiola Vizcarra Reyes ◽  
Gina Alejandra Diaz Mosquera ◽  
...  

Acute promyelocytic leukemia (APL) is a subtype of Acute Myeloid Leukemia (AML) in which a chromosomal translocation t (15; 17) (q22; q12) is generated by fusing produces a hybrid PML / RARα gene, generating an altered signal . The combination of transretinoic acid (ATRA) plus arsenic trioxide (ATO) has been shown to be superior to ATRA plus chemotherapy in the treatment of newly diagnosed standard risk patients with acute promyelocytic leukemia (APL) in several countries. The objective of the present study is to describe the frequency of remission in patients with acute promyelocytic leukemia who were administered as a first line Arsenic Trioxide (varitrinox) during the period from November 2017 to June 2020 in Colombian patients. Methods: Retrospective observational and descriptive study of 12 patients diagnosed with acute promyelocytic leukemia treated with ATO Arsenic trioxide (Varitrinox) as first line, the source of information was provided by the treating hematologists (medical records) by filling out the technical concept format. Active pharmacovigilance scientist in Colombia, this format keeps the identification information of the patient anonymized and the confidentiality of the data is guaranteed as well as compliance with the rules of good clinical practice. Results: Twelve patients with age range between 22 and 69 years with a median age of 34.0 were analyzed. It was found in the analysis that 100% had induction hematologic remission with a median of 45 days. 75% of patients received ATO + ATRA and were at low and intermediate risk, the remaining 25% received ATRA + ATO + Chemotherapy and were at high risk, and intermediate risk. 91.7% of molecular remission in consolidation was obtained and it was measured in cycle 3 by means of PCR (undetectable), 8.3% (n = 1) was positive 3% and is finishing consolidation. Regarding the most frequent adverse events, intravascular coagulation (n = 9), neutropenia (n = 6) and thrombocytopenia (n = 6) were observed. 75% of patients are disease-free, 16.7% are on maintenance (they received ATO + ATRA + Induction chemotherapy) and 8.3% are on consolidation. So far, none of the patients under study have died. Conclusions: Our results support the use of ATO (Varitrinox) in newly diagnosed APL patients (as first line), as a care strategy for low, intermediate and high risk patients. The role of ATRA-ATO is guaranteed in other studies where they manage patients of different risks. Key words: Arsenic trioxide, leukemia promyelocytic acute, leukemia myeloid acute, remission induction, tretinoin. Disclosures No relevant conflicts of interest to declare.


1999 ◽  
Vol 112 (1) ◽  
pp. 113-118 ◽  
Author(s):  
Zahid Kaleem ◽  
Michael S. Watson ◽  
Mary M. Zutter ◽  
Morey A. Blinder ◽  
Jay L. Hess

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 4284-4284
Author(s):  
Zhang Jie ◽  
Xiaojian Meng ◽  
Zhen Cai ◽  
Xiujin Ye ◽  
He Huang

Abstract Abstract 4284 Background: Acute promyelocytic leukemia (APL) is a subtype of acute myeloid leukemia (AML) with distinctive biologic and clinical features that is now highly curable. However, there are complicated issues in treatment strategies for induction, consolidation and maintenance that remain to be studied. The optimal regimen and the duration of consolidation is one of the above controversies. Patients and methods: Six patients (4 males, 2 females) were enrolled in this observation with a median age of 28 years (18–36). All were diagnosed de novo APL with demonstration of the abnormal increased promyelocytes of 53–90% accompanied with detection of t(15;17) or PML/RARα rearrangements. At diagnosis, WBC counts were 0.9–4.6×109/L and platelet counts were 13–84×109/L. Induction therapy was composed of all-trans retinoic acid (ATRA) with or without anthracycline or homoharritonine. After achieving complete remission (CR) following the above regimens, consolidation therapy was given monthly consisting single anthracycline agent of idarubincin (8–10 mg/m2/d, day 1–3) or aclarubicin (8–12 mg/m2/d, day 1–7) and alternative ATRA (25 mg/m2/d, day 1–15). After 12–18 months of consolidation, patients received maintenance therapy including methotrexate (12 mg/m2/d, per week) plus 6 mercaptopurine (30 mg/m2/d, qod ×12 days) and alternative ATRA (25 mg/m2/d, day 1–15) for one year. Results and conclusions: At present, all of the six patients are in continuous CR status, four of whom ceased treatment and are undergoing regular monitoring. Preliminary results from our experience demonstrates that single anthracycline and alternative ATRA can act as a valid option with limited toxicity for APL and might be used as a consolidation strategy, particularly for low and intermediate-risk patients. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3956-3956 ◽  
Author(s):  
Nithya Balasundaram ◽  
Saravanan Ganesan ◽  
Hamenth Kumar Palani ◽  
Ansu Abu Alex ◽  
Sachin David ◽  
...  

Abstract The focus of ATO resistance in acute promyelocytic leukemia (APL) has centered on mutations in PML-RARA gene (Blood 2011, NEJM 2014). However such mutations are rare and cannot explain the majority of relapses seen in the clinic. To evaluate the mechanisms of ATO resistance, we generated ATO resistant NB4 sub clone NB4-EVAsR1 (A216V - VAF-91.7%) in our laboratory. We also had another ATO resistant cell line (UF1) which does not have the A216V mutation. In an expression array we noted that redox signaling, AMPK signaling and energy metabolism pathways were significantly dysregulated in the ATO resistant cell lines compared to naïve NB4 cells. Towards validating the microarray data and to characterize the ATO resistant cell lines we measured the basal levels of reactive oxygen species (ROS), glutathione(GSH), mitochondrial membrane potential (MMP), glucose uptake and their sensitivity to glycolytic inhibitor 2-Deoxy glucose (2-DG) in comparison to naïve NB4 cells. We observed that resistant cell lines have significantly lower ROS, MMP, glucose uptake (Fig 1a) and increased GSH. We also observed that the resistant cell lines were significantly less susceptible to treatment with 2-DG in comparison to naïve NB4 cells (Fig 1b) suggesting that resistant cell lines were less dependent on glycolysis. ATO has been reported to directly inhibit the glycolytic pathway, this effect is believed to contribute to its cytotoxic effect (PNAS 2015). However, we did not observe any cytotoxic synergy between ATO and 2-DG on naïve NB4 cells and neither did this combination restore sensitivity to ATO in the resistant cell lines (Fig 1b). Next we assessed the sensitivity of these resistant cell lines to oxidative phosphorylation (OXPHOS) inhibitors. We used an uncoupler (FCCP at 10uM) of OXPHOS which promotes uncoupled respiration by deregulating the proton gradient which drives ATP synthesis via ATP synthase. We observed that the FCCP treatment alone did not reduced the viability of naïve NB4 cells. Similarly, viability of ATO resistant cell lines also did not reduce significantly suggesting the ability of these cells to uncouple their metabolic pathway from OXPHOS to glycolysis when inhibited. However, when FCCP was combined with ATO it significantly restored the sensitivity of the resistant cell lines to ATO (Fig 1c). The same combination did not have any additive effect on naïve NB4 cells. The combination not only restored the sensitivity of the ATO resistant cell lines but also sensitized the conventionally ATO resistant cell lines such U937 (Fig 1c) and THP1. In spite of the profound effect on leukemic cells we also observed a significant bystander effect on the normal peripheral blood mononuclear cells (Fig 1c). The data suggests that the sensitivity of these resistant cell lines could be potentially restored by combining ATO with an OXPHOS uncoupler. A number of molecules that are FDA approved and used in the clinic also have OXPHOS uncoupling activity and could potentially be evaluated for their synergistic activity with ATO in leukemia. This data also draws attention to possible severe systemic off-target toxicity of such combinations which may be inadvertently used in the clinic. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2329-2329
Author(s):  
Miwa Adachi ◽  
Akihiro Takeshita ◽  
Tomohiko Taki ◽  
Shigeki Ohtake ◽  
Katsuji Shinagawa ◽  
...  

Abstract Background: A combination of all-trans retinoic acid (ATRA) and chemotherapy (CT) has dramatically improved the prognosis of acute promyelocytic leukemia (APL). Nevertheless, considerable number of patients are either refractory to the treatment or relapse after an initial complete remission (CR). Although prognostic factors for APL have been studied, the influence of chromosomal variations in addition to t(15;17) remains controversial. One of the reasons is due to the numbers of cases studied were relatively small (47 to 513 cases). Here, we analyzed clinical features and outcomes of 775 APL caseswith or without additional chromosome abnormalities (ACAs) who were treated with ATRA and CT in the JALSG-APL studies including a large number of cases analyzed for karyotype. Methods: 1,024 cases aged between 15 and 70 yrs with newly diagnosed APL were enrolled in the JALSG APL92, 95, 97 and 204 studies and 775 patients were assessable for karyopypes. All protocols included induction therapy with ATRA and CT, following several courses of post-remission chemotherapy including anthracyclines. Arsenic trioxide (ATO) was not included. Clinical and biological characteristics such as age, gender, initial leukocyte count, platelet count, number of APL cells, DIC score, lack of Auer-rod and incidence of variant type were analyzed in relation to chromosomal abnormalities in 766 cases. CR rate, relapse rate (RR), overall survival (OS), event-free survival (EFS), and disease-free survival (DFS) were assessed and compared between patients with or without ACAs. Each variation of ACAs was also analyzed with clinical and biological features. This work was supported in part by the National Cancer Center Research and Development Fund (26-A-24), Grants-in-Aid from the Cancer Research from the Japanese Ministry of Health, Labor and Welfare (#23-004 and #25100501). These studies were approved by our IRB. Results: ACAswere noted in 235 patients (30%). Sanz score and the initial leukocyte count were significantly lower in patients with ACAs (p=0.027 and p=0.027, respectively). No other clinical or biological differences were found between patients with and without ACAs. The subgroups of ACAs were shown in Figure 1. Trisomy 8 was found in 76 cases (32%). Other ACAs were found involving chromosome 15 in 37 cases (16%), both chromosomes 15 and 17 in 31 cases (13%), chromosome 7 in 19 cases (8%), chromosome 9 in 12 cases (5%), chromosome 6 in 8 cases (3%), chromosome 21 in 7 cases (3%) and alternative ACAs in 43 cases (18%). A low initial leukocyte count (<3,000/µl) was significantly associated with an abnormality of chromosome 15 (p=0.039) and a high initial leukocyte count (≥10,000/µl) was associated with other unspecified chromosomal abnormalities (p=0.010). In all cases, CR rate, OS, EFS and DFS were not different between patients with and without ACA (p=0.341, p=0.694, p=0.414, p=0.852, respectively). However, in elderly patients (≥50 yrs) with ACAs, OS, EFS and DFS were significantly lower compared to younger patients (<50 yrs) (p=0.019, p=0,023 and p=0.030, respectively) (Figure 2). No such age related difference was observed for patients without ACAs (OS, p=0.068; EFS p=0.485; DFS, p=0.672). In each risk group divided by initial leukocyte count, clinical outcomes were not different between patients with and without ACAs. In patients without ACAs, OS, EFS and DFS of patients assigned to no maintenance or retinoid maintenance were significantly better than in those allocated to the maintenance CT (p<0.001, for all). (Figure 3) The significance was not observed in patients with ACAs except DFS (OS, p=0.161; EFS p=0.293; DFS p=0.043). Conclusions: The present study is the largest to date to focus on the influence of ACAs on clinical outcomes of patients with APL treated with ATRA and CT. The analysis revealed exact variation and frequency of ACAs. We found that patients with ACAs were associated with the lower initial leukocyte count and the lower survival outcomes in elderly patents, suggesting a possible link to age and post-remission chemotherapy. Some promising agents, such as ATO, tamibarotene and gemtuzumab ozogamicin might change the prognostic factors, including ACAs. Careful chromosomal analyses, especially ACAs related to chromosome 15 and/or 17, need to be analyzed by molecular methods and performed in future prospective studies with alarge number of cases. Figure 1 Figure 1. Figure 2 Figure 2. Figure 3 Figure 3. Disclosures Kiyoi: Bristol-Myers Squibb: Research Funding; Chugai Pharmaceutical Co. LTD: Research Funding; Kyowa Hakko Kirin Co. LTD.: Research Funding; Dainippon Sumitomo Pharma: Research Funding; Zenyaku Kogyo: Research Funding; FUJIFILM Corporation: Research Funding. Kobayashi:Ohtsuka: Research Funding; Behringer: Research Funding; Simic: Research Funding. Asou:Chugai Pharmaceutical Co., Ltd.: Research Funding. Miyazaki:Nippon-Shinyaku: Honoraria.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2845-2845
Author(s):  
Zeng Chengwu ◽  
Yikai Zhang ◽  
Tianyu Qin ◽  
Lijian Yang ◽  
Shaohua Chen ◽  
...  

Abstract Acute promyelocytic leukemia (APL) is characterized by the presence of PML-RARA fusion protein. The PML-RARA oncoprotein is known to be the initiating factor for APL development. We have previously found that miR-125b is highly expressed in APL and upregulated by PML-RARA. However, the molecular mechanisms by which PML-RARA controls miR-125b expression had been uncharacterized. In the present study, we further confirmed that expression of PML-RARA resulted in a significant increase in primary miR-125b expression, and we found that the nuclear transcription factor E2-related factor 2 (Nrf2) targeting genes (HO1) was also upregulated (Figure 1A). Consistently, HO1 and miR-125b are transcriptionally upregulated by proteasome inhibitor (MG132) which has been showed to activate Nrf2 (Figure 1B). Knockdown of Nrf2 can inhibit the effect of PML-RARA expression and MG132 (Figure 1C). Thus, these results indicated that Nrf2 may mediate PML-RARA-induced miR-125b expression. Furthermore, we demonstrated the importance of miR-125b in APL cell proliferation (Figure 1D), we showed that promoting leukemic cell grow is associated with AKT pathway and MAPK signaling (Figure 1E). Subsequent bioinformatic and RNA-seq data analysis (Figure 1F and G) identified CEBPA as a putative target of miR-125b, and we further experimentally verified using luciferase reporter constructs (Figure H). In conclusion, we identified the mechanisms responsible for the high expression and the function of miR-125b in APL, suggesting that the expression of miR-125b could be a primary oncogenic event in APL leukemogenesis. This study was supported by grants from the National Natural Science Foundation of China (No.81400102), the China Postdoctoral Science Foundation (No.2015M570751), and the Medical Scientific Research Foundation of Guangdong Province, China (A2015420). Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 4857-4857
Author(s):  
Maria L. Chauffaille ◽  
Rodrigo Proto Siqueira ◽  
Daniela Borri ◽  
Eloisa S. Moreira ◽  
Fernando L. Alberto

Abstract Acute promyelocytic leukemia (APL) is a distinct subtype of acute myeloid leukemia (AML) characterized by t(15;17)(q22;q21), a balanced reciprocal translocation involving PML and RARA genes. Studies conducted in animal models with PML/RARA suggest that this gene rearrangement is not sufficient for the development of APL, emphasizing the importance of additional mutations. Another gene that could be involved in APL leukemogenesis is FLT3, a Fms-like tyrosine kinase receptor which mutations have been observed in different AML subtypes as the internal tandem duplications (ITD), in around 18–37% of APL cases or less frequently, the D835Y mutation. The association of FLT3/ITD in APL gave rise to the proposition that it could provide a common class of cooperative mutation in the development of the disease. Indeed, APL represents 21% of all cases of AML with karyotype confirmation in our institution, located in São Paulo, Southeast region of Brazil. Bone marrow samples from 92 consecutive APL patients at diagnosis were enrolled in the study. Mean age was 29.5 years old, varying from 4 months to 67 years old, with the following age distribution: 35.7% of the patients were younger than 20 years old; 55.2% were between 20 and 49 years old and only 17.2% were older than 50 years. There were 50 males and 42 females. The WBC count demonstrated a wide variation, from severe leukopenia (&lt;500 leukocytes/μL) until marked leukocytosis (174.600/μL), most of them presenting circulating blasts. Hemoglobin level varied from 3.7g/dL to 14g/dL and platelets counts were frequently low. Cases included five (5.4%) microgranular variant patients. We observed that 23 (25%) out of 92 cases presented chromosomal abnormalities in addition to t(15;17), among which trisomy 8 was the most frequent (21.7%, 5/23). Other abnormalities included tetraploidy, trisomy 6, trisomy 21 (non constitutional), nulissomy Y, ider(17q), trisomy 11, del(9)(q22q32), add(3)(p26), del(6)(p21), abnormalities of chromosome 7 and 1, t(1;3)(q21;q29); t(7;9)(q36;q31); monosomy of chromosomes 6, 7, 8 and 9; dup(1)(q23q42), del(10)(q24); add(7)(q36) and marker chromosomes. Seventy nine cases had enough DNA for complimentary molecular studies; in the remaining 13 cases we could not retrieve any DNA amount from fixed cells. Among those, FLT3/ITD was detected in 29 (36.7%), 5 of them showing additional karyotype abnormalities and D835Y mutation was present in only 2 (2.8%) of the cases. None of the two patients with D835Y showed additional chromosomal abnormality, but one of them presented ITD and D835Y mutation simultaneously at diagnosis. No correlation was found between age, sex, WBC count, hemoglobin level, platelet count or clinical symptoms and the presence or absence of additional karyotype abnormalities, FLT3-ITD and D835Y. The frequencies of additional karyotype abnormalities as well as FLT3/ITD in Brazilian APL patients were similar to those reported in several worldwide published papers. On the other hand, the incidence of D835Y was lower than that described elsewhere (2.8% versus 7% in AML or 5.7% in APL). So although Latin American population has an increased incidence of APL, it seems not to bear a higher frequency of clonal additional chromosome abnormalities or mutations in FLT3 gene. Moreover, FLT3/ITD was not over-represented in cases with additional chromosomal abnormalities (17.2% versus 36.7%, p=0,2), thus suggesting an independent role for each FLT3/ITD and extra clonal chromosomal abnormalities in t(15;17) APL. In the present study, a significant proportion of APL patients (56%) showed additional karyotype alterations and/or FLT3/ITD or D835Y mutation. In fact, one fourth of APL cases presented additional chromosomal abnormalities at diagnosis while 1/3 of the cases showed the FLT3/ITD and only two patients presented D835Y mutation. So, roughly more than one half of the cases presented at least one of the alterations investigated, raising the possibility that these abnormalities may be complimentary events to PML/RARA in the process of leukemogenesis. The influence of additional chromosomal abnormalities predisposing to genomic instability and cooperating with genetic mutations in acute leukemia still deserves investigation, since about 45% of APL cases have not been shown to bear any detectable molecular abnormality to date.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4383-4383
Author(s):  
Clayton B Hess ◽  
W. Christopher Ehmann

Abstract Abstract 4383 Here we describe a case of acute promyelocytic leukemia (APL) (M3 leukemia) which developed in a patient treated with single-agent fludarabine for lymphoplasmacytic lymphoma (Waldenstrom's macroglobulinemia [WM]). This 69-year-old male was diagnosed with WM in 2003. He received single-agent therapy with 4 cycles of fludarabine, after having an adverse reaction to his first and only dose of rituximab. Therapy was completed in 2007, with the patient in a complete remission. Two years later, he presented with leukocytosis and a consumptive coagulopathy. A bone marrow biopsy showed 90% abnormal promyelocytes with prominent Auer rods. PML/RARA fusion was positive by FISH. PCR was positive for t(15;17) (PML/RAR long (bcr1 and bcr2) and short (bcr3) form translocations. Cytogenetic studies showed abnormal 3q, monosomy 15 and 17, 2 unidentified marker chromosomes and a complex insertional translocation of chromosomes 15 and 17, with a PML/RARA fusion. The patient's APL was treated with ATRA and 7+ 3 chemotherapy because of a rising number of blasts in his peripheral blood. His subsequent clinical course was complicated by refractory atrial fibrillation, renal failure, a catheter-related upper-extremity deep venous thrombosis, and severe deconditioning. The patient died four months later from probable sepsis. A marrow performed two weeks prior to his death showed a complete hematologic remission, and no evidence of either Waldenstrom's or acute promyelocytic leukemia. There are forty-eight cases published of hematologic malignancies which occurred after single-agent nucleoside-analog treatment: none were APL. Twenty-four of these cases consisted of a lymphoid transformation to a more aggressive histology and twenty-four cases described development of a myeloid leukemia or myelodysplastic syndrome. In these patients, a variety of chromosomal changes were demonstrated with abnormalities of chromosome 7 being the most frequently observed (exclusively in those with myeloid disorders). There were no cases of a t(15;17) reported. We then reviewed all documented cases of therapy-related APL. No cases implicating nucleoside analogs were found. Topoisomerase inhibitors were the most common agents implicated as were radiation, anthracyclines, and alkylating agents. Because fludarabine was the only chemotherapy agent to which the patient was exposed, and the patient developed APL within the time frame expected for therapy-related leukemogenesis, we conclude that this is the first reported case of fludarabine-associated APL. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document