Incidence and Management of Relapsed Acute Promyelocytic Leukemia

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5179-5179
Author(s):  
Michael R Hessenauer ◽  
Nosha Farhadfar ◽  
Naseema Gangat

Abstract Background: Acute promyelocytic leukemia (APL) has a favorable prognosis with high complete remission rates of over 90 percent, with relapse occurring in only 10 to 15 percent of patients. Our objective was to describe the characteristics of relapsed patients and review management of relapsed disease. Methods: After institutional review board approval, we searched our institutional electronic database for those 18 years and older with a new diagnosis of acute promyelocytic leukemia between January 1, 2000 and April 30, 2016. We gathered all patient information through electronic chart review. Results: Out of 89 patients reviewed, 59 had complete treatment data through induction therapy. Of the 59 patients, with median follow-up of 51 months (range 1-180 months), 7 (11.9%) had relapsed. The median age at APL diagnosis of relapsed patients was 52 years (range: 30-77 years). 6 relapsed patients were intermediate or low risk, and 1 was in the high risk category. 6 relapsed patients received an ATRA and chemotherapy-based induction regimen, and 1 received an ATRA and arsenic-based regimen. Median time to morphological remission was 32 days (range: 22-83 days). 6 of 7 relapsed patients achieved molecular remission. Consolidation regimens included conventional chemotherapy alone (1 patient), ATRA and conventional chemotherapy (4 patients), and arsenic alone (2 patients). Of the 7 relapsed patients, 4 completed maintenance therapy. The median time to relapse from first morphologic remission was 40 months (range: 3-47 months). 6 patients were treated at our institution, and 1 was treated at an outside institution. 2 patients had a molecular relapse, 2 had systemic and central nervous system (CNS) relapse, and 1 patient had an isolated CNS relapse. Treatment regimens at relapse included ATRA and chemotherapy (1 patient), arsenic alone (2 patients), ATRA and arsenic (2 patients), and intrathecal methotrexate with conventional chemotherapy (1 patient). Complications of treatment included infection (2 patients: 1 treated with ATRA and chemotherapy and 1 with intrathecal methotrexate and chemotherapy), thrombotic events (1 patient treated with ATRA and arsenic), rash (1 patient treated with arsenic), and QTc prolongation (1 patient treated with ATRA and arsenic). 5 of 6 patients achieved a molecular remission. Consolidation regimens after reinduction included ATRA and chemotherapy (2 patients), arsenic alone (1 patient), intrathecal methotrexate (1 patient), and ATRA and arsenic (1 patient). Of the 2 patients who had a second relapse, 1 had an isolated CNS relapse, and 1 had a systemic and CNS relapse. Both patients were treated with intrathecal methotrexate and conventional chemotherapy. Due to persistent CNS disease, 1 patient underwent therapy with systemic methotrexate and craniospinal radiation. 1 patient had a third relapse, which was treated with ATRA and arsenic. 4 of 6 patients underwent hematopoietic stem cell transplant, 2 autologous and 2 allogeneic. Of the 7 relapsed patients, 5 are alive, including all 4 patients who underwent transplant. Table 1 provides a detailed summary of the management of relapse in each of these patients. Conclusion: Our data are consistent with the reported relapse rate of 10 to 15 percent in APL. Patients who relapse are successfully treated with both ATRA and arsenic-containing regimens. In our cohort, relapsed patients had favorable survival. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 666-666
Author(s):  
Fouzia N. ◽  
Vibhor Sharma ◽  
Anu Korula ◽  
Anup Joseph Devasia ◽  
Uday Prakash Kulkarni ◽  
...  

Abstract The standard of care for patients with acute promyelocytic leukemia (APL) relapsing after frontline treatment with arsenic trioxide (ATO) based regimens remains to be defined. Available data on response and survival outcomes in patients who had received ATO as part of upfront therapy suggests that there is a high incidence of resistance to ATO and ATRA resulting in inferior response and survival (Zhu HH et al. N Engl J Med. 2014). We present our experience on management of relapse in APL patients treated with frontline ATO based therapy. Data on all consecutive patients with relapsed APL diagnosed and treated in the Depatment of Haematology, Christian Medical College, Vellore, from January, 1998-December, 2015 were included in this retrospective analysis. One hundred and four (29%) out of the total 358 APL patients diagnosed during the study period had relapsed. Out of these 73 (70%) patients received upfront ATO based therapy. Six out of 73 refused treatment and got discharged at request following the diagnosis of relapse, the remaining 67 (91.8%) were included in the analysis. Median age of patients was 28 years (range: 4-54), 44 (65.7%) were males. Median time to relapse from initial diagnosis was 19.6 months (range: 6 - 128). Relapses were isolated molecular in one, isolated CNS in 6 [9%], bone marrow + CNS in 13 [19.4%] and medullary only in 47 [70.1%] patients. All 67 patients received ATO based therapy (ATO ± ATRA +/- Anthracycline/Bortezomib) as re-induction. 63/67 (94%) achieved molecular remission post re-induction, while 3 (4.5%) died during re-induction and one discontinued treatment. An autologous SCT was offered to all patients who achieved molecular remission, however only a proportion of cases opted for SCT due to financial constraints. Those that did not opt for an autologous SCT were given ATO+ATRA maintenance therapy. 35/63 (55.6%) opted for autologous SCT while 28 (44.4%) continued on maintenance therapy alone. Median time to autologous SCT from relapse was 5.8 months (range: 2 - 32). There were no treatment related deaths following autologous SCT. At a median follow up of 46 months (range: 1-224), the 5 year estimate of OS and EFS (Fig 1) of the whole cohort (n=67) was 73.6% ± 5.7% and 67% ± 6.1%. The 5 year estimate of OS and EFS (Fig 2) of those who received autologous SCT vs those who were on chemotherapy were 90.3% ±5.3 vs 58.6±10.4 %, (P=0.004) and 87.1% ±6.0% vs. 47.7% ±10.3%, (P=0.001) respectively. In unadjusted analysis, those who received chemotherapy as post relapse consolidation had 5.73 (95%CI: 1.86 - 17.65) times risk of relapse as compared to those who received autologous SCT, which was statically significant (P=0.002). In the adjusted analysis (Table 1) those who received chemotherapy the risk of relapse was 7.6 (95%CI: 1.98 - 28.87) times compared to those who received autologous SCT, which was statically significant (P=0.003) after adjusting for age, total WBC at relapse and duration of CR1. Remission induction with ATO based regimens followed by an autologous SCT in patients with relapsed APL who were treated with frontline ATO based regimens is associated with excellent long term survival and should be considered the standard of care even in this setting. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 2085-2085
Author(s):  
Graham Tooker ◽  
Ashraf Z. Badros ◽  
Jennifer Nishioka ◽  
David Riedel

Abstract Background: BKV HC is a well-known complication following allo-SCT. Supportive care with bladder irrigation and blood transfusions were the only available treatment. Since our initial report (Bridges B et al. Am J Hematol 2006;81:535), several studies confirmed that intravesicular cidofovir is a potential effective treatment for BKV HC. In this study, we report a large series of consecutive patients who developed BKV HC following allo SCT and received intravesicular cidofovir. Methods: We conducted a retrospective review of allo SCT patients who developed BKV HC and were prescribed intravesicular cidofovir from 2012 to 2017. Results: 33 patients were diagnosed with BKV HC. The median age was 50 years (range=23-73), and 18 (55%) were male. Acute myeloid leukemia (n=12, 35%) was the most common diagnosis followed by non-Hodgkin lymphoma (n=7, 21%) and B cell acute lymphoblastic leukemia (n=4, 12%). Conditioning regimens were myeloablative (n=19, 58%) or reduced-intensity (n=14, 42%); 15 (45%) patients received cyclophosphamide, and 22 (67%) received total body irradiation. The median time to onset of HC symptoms following SCT was 37 days (range: 8-178); 17 (52%) patients had acute graft vs. host disease. HC symptom severity ranged from grade 0-4 (median=2). The median BK urine viral load pre-treatment was 100,000,000 IU/ml. Patients received a median of 2 intravesicular treatments (range=1-7) at a dose of 5 mg/kg. Four patients (12%) were also treated concurrently with intravenous cidofovir. 19 (59%) patients demonstrated complete clinical resolution of symptoms, 9 (28%) demonstrated partial response to treatment, and 4 (13%) had no change in symptoms following treatment. These improvements in clinical status were independent of viral load, though most had reductions in the viral load. The median time to symptom resolution was 17 days (range=7-53; n=28). 82% of patients had no recurrent symptoms of HC. The main side effect of intra-vesicular instillation was increased discomfort and bladder spasms; severe in 3 patients (9%). No patient had impaired renal function directly attributable to intra-vesicular cidofovir. At 12 months after BKV HC diagnosis, 26 (79%) patients were alive. Conclusions: To our knowledge this is the largest study of intravesicular cidofovir for BKV HC reported to date; 77% of patients with BVK HC achieved clinical improvement of symptoms with minimal side effects. Clinical trials of intravesicular cidofovir could provide further evidence for adding intravesicular cidofovir as a standard tool for the treatment of BKV HC. Disclosures Badros: Celgene: Consultancy, Research Funding; Karyopharm: Research Funding; GSK: Research Funding.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1175-1175
Author(s):  
Xiaomin Zheng ◽  
Anita Seshire ◽  
Elena Puccetti ◽  
Hilal Gul ◽  
Tim Beissert ◽  
...  

Abstract Acute promyelocytic leukemia (APL) is distinguished from other AMLs by cytogenetic, clinical, as well as biological characteristics. The hallmark of APL is the t(15;17) which leads to the expression of the PML/RAR fusion protein. PML/RAR is the central leukemia-inducing lesion in APL and is directly targeted by all trans retinoic acid (t-RA). Patients suffering from APL undergo complete hematologic but not molecular remission upon treatment with t-RA. Virtually all patients treated with t-RA-monotherapy had a rapid relapse within few months. But in the combination with an anthracycline, such as doxorubicin or idarubicin, t-RA improved the long term outcome of APL-patients dramatically. Nothing is known about why t-RA-monotherapy is unable to eradicate completely the leukemic population and how it increases the response to chemotherapy. In vitro, the exposure of early hemopoietic stem cells (HSCs) to t-RA does not induce differentiation but selects immature progenitors. Moreover, mice lacking the t-RA-specific receptor RARalpha do not exhibit an impairment of granulopoiesis or hemopoiesis. The indication, that t-RA may be involved in the hemopoietic differentiation, is given by the HL-60 cell line which undergoes granulocytic differentiation at the pharmacological dosages (10−6M) of t-RA. Furthermore vitamin A-deficient mice or mice treated with a antagonist of t-RA accumulate more immature granulocytes in the bone marrow. PML/RAR mediates the response of APL blasts to t-RA, but it is completely unclear, which effect t-RA exerts on the PML/RAR-positive leukemic stem cells which maintains the blast population and represents the source of relapse. Therefore we investigated the effect of t-RA on a cell population with stem cell capacity expressing PML/RAR isolated from the APL cell line NB4 as well as from CD34+/CD38- KG-1 cells transfected with PML/RAR. Here we report that i) the NB4 cells engrafted in NOD/SCID mice indicating the presence of a subpopulation with stem cell capacity in NB4 cells; ii) NB4 had a Hoechst 3342 excluding side population (SP) representing about 1% of the whole cell population; iii) t-RA reduced but did not deplete the side population in NB4 cells; iv) the expression of PML/RAR increased CD34+/CD38- population in KG-1 cells from 75% to over 95%; v) t-RA reduced the CD34+/CD38- population from 75% to 3,5% in mock transfected KG-1 confirming its capacity to induce differentiation, whereas in PML/RAR-positive KG-1 cells it led only to a reduction from 98% to a 25%, which still maintain the capacity to engraft in NOD-SCID mice; vi) also the expression of other fusion proteins, such as AML-1/ETO or PLZF/RAR, associated with t-RA-resistant AML-subtypes, increased the percentage of CD34+/CD38- KG-1 cells over 90%, which was reduced by t-RA only to 35% and 19%, respectively. Taken together these data suggest that a subset of early HSC expressing PML/RAR exhibit the same t-RA-resistant phenotype as HSC expressing fusion proteins associated with AML-subtypes which, in contrast to APL, do not respond to t-RA. These data may give an explanation, why APL-patients do not achieve complete molecular remission upon t-RA monotherapy and undergo early relapse.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 395-395 ◽  
Author(s):  
Wing Y. Au ◽  
James C. Chim ◽  
Albert K. Lie ◽  
Cyrus R. Kumana ◽  
Anskar Y. Leung ◽  
...  

Abstract Background: The optimal therapy for relapsed acute promyelocytic leukemia (APL) after arsenic trioxide (As2O3)-induced remission is unclear. Hematopoietic stem cell transplantation (HSCT) is associated with high morbidity and mortality. Moreover, lasting remission is observed in many patients who are not candidates for HSCT, owing to advanced age or lack of donors, implying that HSCT is not mandatory for durable remission. We evaluated our results of an As2O3-based, non-HSCT regimen for patients with relapsed APL. Materials and methods: Forty-two consecutive patients (18 men, 24 women, median age: 35 years, 12–72) with relapsed (relapse 1, R1=39, R2=3) APL were treated with an-As2O3 based, non-HSCT regimen. The time from last complete remission (CR) was 22 (6–243) months (mo). Initial treatment was As2O3 (10 mg/day) either intravenously (n=16) or orally (n=28) until CR, followed by idarubicin consolidation (6 mg/m2/day x 9). Twenty-five patients received oral-As2O3 maintenance. Post-As2O3 relapses were treated with oral As2O3 + all-trans retinoic acid (ATRA, 45 mg/m2/day) until CR, followed by maintenance (two weeks of ATRA+As2O3 every 2 mo. for 2 years). Post-As2O3/ATRA relapses were treated with oral As2O3+ATRA+ascorbic acid (1g/day) until CR, followed by consolidation/maintenance with the same regimen (2 weeks every 2 mo. for 2 years). Part of the induction and all of the maintenance therapies were given in the outpatient clinic. Results: Forty-one patients (98%) achieved CR after initial As2O3 treatment. One 72-year old man with XYY syndrome, diabetes and mental retardation died of pneumonia. Thirteen relapses occurred at a median of 15 (6–22) mo. As2O3-maintenance significantly decreased further relapses (3/24 with versus 10/17 without As2O3-maintenance, p=0.003). Two relapses died of cerebral APL before further treatment could be administered. Of eleven patients treated with As2O3+ATRA, 10 achieved CR, 8 of whom have remained in remission (median follow-up: 33 mo.). Two post-As2O3/ATRA relapses achieved CR again with As2O3+ATRA+ascorbic acid, and have remained in remission after maintenance treatment with As2O3+ATRA+ascorbic acid. All patients in continuous remission (n=38) were PML/RARa negative on polymerase chain reaction (sensitivity 10−3 to 10−4). Conclusion: Our regimen resulted in a leukemia-free-survival of 89.3%. The results suggest that an oral and mainly outpatient As2O3-based, non-HSCT strategy is efficacious for relapsed APL. In terms of survival, costs, treatment side effects and patient tolerance, the results appear to be comparable to, if not more favorable than, other treatment options based on high dose chemotherapy, graft-versus-leukemia effect, or anti-myeloid antibody therapy.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2958-2958
Author(s):  
Wing-Yan Au ◽  
Sidney Tam ◽  
Anskar Y.H. Leung ◽  
Eric Tse ◽  
Cyrus Kumana ◽  
...  

Abstract Background. For patients with relapsed acute promyelocytic leukemia (APL), the optimal therapy after arsenic trioxide (As2O3)-induced complete remission (CR) is unclear. Autologous or allogeneic hematopoietic stem cell transplantation (HSCT) has been advocated. These strategies are associated with morbidity and considerable mortality, and not all patients are eligible. The role of maintenance therapy with As2O3 remains undefined. Materials and methods. From 1997–2008, 50 consecutive APL patients (25 men, 25 women, median age: 35 (12–72) years) in relapse (R1, n=47; R2, n=3) were treated with As2O3 until CR. This was followed by idarubicin consolidation (6 mg/m2/day × 9) in 43 patients. Seventeen patients did not receive maintenance treatment. Thirty-two patients received oral-As2O3 maintenance (10 mg/day × 14 every 2 months). All-trans retinoic acid (ATRA, 45 mg/m2/day) maintenance was used together with oral-As2O3 in 27 patients. As2O3 protocol was approved by the institutional review board at Queen Mary Hospital. All maintenance treatment was given in the outpatient clinic as oral medication. Results. Of 50 relapsed cases, As2O3-induced CR was achieved in 49 patients, 1 patient dying of pneumonia. At a median follow-up of 61 (6–122) months, 27 patients (19 with and 8 without As2O3 maintenance) had remained in remission. Further relapses (R2, n=20; R3, n=2.) occurred in 22 patients (13 with and 9 without As2O3 maintenance), at a median of 16 (6–28) months. Concomitant central nervous system (CNS) relapse occurred in 8 cases. Treatment of post-As2O3 relapses included oral-As2O3 (10 mg/day) + ATRA (45 mg/m2/day) with (n=10) or without (n=10) ascorbic acid (1 gm/day) until CR, and then maintenance with As2O3 + ATRA +/− ascorbic acid. Leucocytosis during treatment was controlled with mitoxantrone. In these 22 cases of post-As2O3 relapses, further remission was still achieved in 19 patients (CR3, n=18; CR4, n=1), with 3 patients dying from cerebral bleeding. At a median follow-up of 88 (8–98) months, 8 patients had remained in remission. Further relapses occurred in 11 patients. Salvage therapy consisted of As2O3+ATRA+ascorbic acid, together with chemotherapy (mitoxantrone, n=8; amascrine, n=3), gemtuzumab ozogamicin (n=4), and autologous HSCT (n=1). Eight patients finally died of refractory leukemia after a median of 6 (2–33) months. Three patients had remained in remission (CR4, n=2, CR5 n=1) at a median of 41 (26–102) months. On an intention-totreat basis, our oral-As2O3 treatment/maintenance regimen for patients with relapsed APL resulted in a 4-year overall survival of 71%, and event-free-survival of 53%. Conclusion. Our findings showed that an oral-As2O3-based treatment/maintenance strategy resulted in durable remission in a significant proportion of patients with relapsed APL. As most of the treatment is administered at home, this regimen involves much less financial, personnel and emotional costs as compared with HSCT strategies. Further improvement should be directed towards prevention of CNS relapses with appropriate prophylaxis in high-risk patients. For remissions at or beyond CR3, consideration of HSC harvest and storage at molecular remission should be considered. The use of demethylation agents and histone deacetylase inhibitors in combination with As2O3 remains to be investigated.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4827-4827
Author(s):  
Fleur M. Aung ◽  
Jordan Myint ◽  
Erin T Roughneen ◽  
Benjamin Lichtiger

Introduction Acute Promyelocytic Leukemia (APL), a distinct subtype of acute myeloid leukemia is a relatively rare disease, characterized by a severe coagulopathy which is often present at the time of diagnosis. Mortality due to bleeding complications during induction is more common in this subtype than in other FAB classifications. The number of newly diagnosed cases in the US is estimated to be 600 to 800 cases a year. The introduction of all-trans retinoic acid (ATRA) into the therapy of APL has completely revolutionized the management and outcome of this disease. The treatment and cure of patients with APL depend not only on the effective use of combination therapy but also involves critical supportive care measures. Aim The aim of this study was to analyze the number of red cells, platelets, plasma and cryoprecipitate transfused during the induction phase of treatment until time to response. Method Patient and transfusion data was retrospectively collected from the Leukemia Department files and Blood bank records at the UT MD Anderson Cancer Center from 2010 to 2011. Results There were 28 newly diagnosed APL patients ([16F: 12 M]; 2 AA/6 Hispanic/20 White), median age 49 (21-84) and included patients who did not go on to clinical trials due to early complications. Karyotyping was obtained on 26 (93%) patients. Confirmation of the PML-RARα short or long transcripts was obtained in 25 (89%) patients by quantitative RT-PCR all of whom showed the PML-RARα fusion transcript. Induction therapy was started on day -1 to day 0 from the date of diagnosis in 5 (18%) patients, Day 1 in 13 (46%), Day 2 in 1 (3%), Day 3 in 3 (11%), Day 4 in 2 (7%), Day 6 in 3 (11%) and Day 7 in 1 (3%) patient. 24 (86%) patientsreceived Arsenic + ATRA, 3 (11%) received Arsenic + ATRA + Idarubicinand 1 (3%)received Arsenic + ATRA + Gemtuzumab Ozogamicin. 4 (14%) patients died early from complications of severe coagulopathy. Response to therapy was noted in 24 (86%) patients, median 25 (range 19-63) days from start of treatment. Red cells were transfused to 25 (89%) patients, median 6 (range 1-29) units, platelets to 23 (82%) patients, median 5 (1-47) units, plasma to 11 (39%) patients, median 8 (2-38) units and cryoprecipitate to 14 (50%) patients, median 10 (2-20) units. There was 1 (3%) patient who did not require blood or blood products, 3 (11%) did not require red cell transfusions, 5 (18%) platelet transfusions, 17 (61%) plasma transfusions and 14 (50%) did not require cryoprecipitate. Of the 24 patients who responded to therapy, 22 (79%) patients are alive. One patient has been lost to follow up. The remaining 21 (75%) patients are in molecular remission with a median follow-up of 714 (256-1110) days from the date of response. Two (7%) patients died in molecular remission from unrelated non-hematologic causes (204, 283 days from their date of response). Table 1 The results of the laboratory studies at the time of diagnosis/ time of response are as follows; WBC median 1.2 K (0.5-17.9)/median 3.3 K/UL (1.0-5.5), Hgb median 8.39 G/Dl (5.9-12.1/median 10.3 G/Dl (8.1-12.1), platelet count median 31 K/UL (3-87)/median 180 K/UL (49-1335), BM blast median 1% (0-64), median 1% (0-4), BM progranulocytes median 59% (0-93)/median 1% (0-7), BM normoblast median 9% (1-35)/median 28% (0-72%), PT median 16.2 secs (14.7-21.0)/ median 14.3 sec (13.1-15.5), INR median 1.29 (1.12-1.76)/median 1.10 (0.97-1.20), aPTT median 29.9 secs (26.0-41.0)/ median 32.2 secs (24.1-47.4), D -Dimer median 19.83 mcg/ml (3.71->20.00)/median 0.96 mcg/ml (0.39-6.09), Fibrinogen median 172 MG/DL (77-461)/ median 399 MG/DL (164-856) and LDH median 883 IU/L (374-2561)/median 591 IU/L (444-1084). Conclusion In conclusion, our review found that the majority of cases required red cells and platelet transfusion but only 50% of the patients required plasma or cryoprecipitate transfusion support for their coagulopathy. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document