Materials and Methods

Similar documents
Acute myeloid leukemia. M. Kaźmierczak 2016

Concomitant WT1 mutations predicted poor prognosis in CEBPA double-mutated acute myeloid leukemia

SWOG ONCOLOGY RESEARCH PROFESSIONAL (ORP) MANUAL LEUKEMIA FORMS CHAPTER 16A REVISED: DECEMBER 2017

Acute myeloid leukemia: prognosis and treatment. Dimitri A. Breems, MD, PhD Internist-Hematoloog Ziekenhuis Netwerk Antwerpen Campus Stuivenberg

Mixed Phenotype Acute Leukemias

Group of malignant disorders of the hematopoietic tissues characteristically associated with increased numbers of white cells in the bone marrow and

HEMATOLOGIC MALIGNANCIES BIOLOGY

MS.4/ Acute Leukemia: AML. Abdallah Al Abbadi.MD.FRCP.FRCPath Feras Fararjeh MD

JPMA ( Journal Of Pakistan Medical Association) Vol 53, No.9,Sep Original Articles

New treatment strategies in myelodysplastic syndromes and acute myeloid leukemia van der Helm, Lidia Henrieke

Done By : WESSEN ADNAN BUTHAINAH AL-MASAEED

WBCs Disorders 1. Dr. Nabila Hamdi MD, PhD

Myeloperoxidase Expression in Acute Myeloid Leukemia Helps Identifying Patients to Benefit from Transplant

Case Presentation No. 075

Case Workshop of Society for Hematopathology and European Association for Haematopathology

HAEMATOLOGICAL MALIGNANCY

Therapy-related MDS/AML with KMT2A (MLL) Rearrangement Following Therapy for APL Case 0328

Case Report Blasts-more than meets the eye: evaluation of post-induction day 21 bone marrow in CBFB rearranged acute leukemia

Acute Lymphoblastic Leukemia in Elderly Patients The Philadelphia Chromosome May Not Be a Significant Adverse Prognostic Factor

CLASSIFICATION OF NINETY-EIGHT ADULT CASES OF ACUTE LEUKEMIAS ACCORDING TO MORPHOLOGY, IMMUNOLOGY AND CYTOGENETICS

MS.4/ 1.Nov/2015. Acute Leukemia: AML. Abdallah Abbadi

ABERRANT EXPRESSION OF CD19 AND CD43

Myelodysplasia/Myeloproliferative Neoplasms (MDS/MPN) Post-HCT Data

Hematology MUHAMMAD M. KHURRAM* SAGHIR A. JAFRI* ABDUL MANNAN** AFTAB NADEEM*** ASIF JAMAL*

CYTOGENETIC STUDY OF 50 DE NOVO CASES OF ANLL FROM ARGENTINA. Susana Acevedo, Irma Slavutsky, Gabriela Andreoli, Irene Larripa

If unqualified, Complete remission is considered to be Haematological complete remission

If unqualified, Complete remission is considered to be Haematological complete remission

Keywords: Acute Myeloid Leukemia, FLT3-ITD Mutation, FAB Subgroups, Cytogenetic Risk Groups

Corporate Medical Policy

Classification of Hematologic Malignancies. Patricia Aoun MD MPH

Laboratory Correlates and Prognostic Significance of Granular Acute Lymphoblastic Leukemia in Children A Pediatric Oncology Group Study

CHAPTER:4 LEUKEMIA. BY Mrs. K.SHAILAJA., M. PHARM., LECTURER DEPT OF PHARMACY PRACTICE, SRM COLLEGE OF PHARMACY 8/12/2009

Original Article. Clinical features and outcome of acute myeloid leukemia, a single institution experience in Saudi Arabia INTRODUCTION

THE LEUKEMIAS. Etiology:

Corporate Medical Policy. Policy Effective February 23, 2018

CME/SAM. Mixed Phenotype Acute Leukemia

Research Article Myeloid Sarcoma: Clinicopathologic, Cytogenetic, and Outcome Analysis of 21 Adult Patients

Characteristics and Outcome of Therapy-Related Acute Promyelocytic Leukemia After Different Front-line Therapies

Outcome of acute leukemia patients with central nervous system (CNS) involvement treated with total body or CNS irradiation before transplantation

Johann Hitzler, MD, FRCPC, FAAP Jacqueline Halton, MD, FRCPC Jason D. Pole, PhD

CLINICAL STUDY REPORT SYNOPSIS

MUD SCT for Paediatric AML?

Daniel A. Arber, MD, 1 Anthony S. Stein, MD, 2 Nora H. Carter, MS, 3 David Ikle, PhD, 3 Stephen J. Forman, MD, 2 and Marilyn L.

Risk factors and clinical outcomes of acute myeloid leukaemia with central nervous system involvement in adults

Myeloproliferative Disorders - D Savage - 9 Jan 2002

Mast Cell Disease Case 054 Session 7

LEUKAEMIA and LYMPHOMA. Dr Mubarak Abdelrahman Assistant Professor Jazan University

Acute Promyelocytic Leukemia with i(17)(q10)

Adult Acute leukemia. Matthew Seftel. August

Jeanne Palmer February 26, 2017 Mayo Clinic, Phoenix, AZ

OUTCOME OF ACUTE MYELOID LEUKEMIA IN PATIENTS UP TO 65 YEARS OF AGE AT HEMATOLOGY AND BONE MARROW TRANSPLANTATION UNIT IN ALGIERS DURING 14 YEARS

A.M.W. van Marion. H.M. Lokhorst. N.W.C.J. van de Donk. J.G. van den Tweel. Histopathology 2002, 41 (suppl 2):77-92 (modified)

The patient had a mild splenomegaly but no obvious lymph node enlargement. The consensus phenotype obtained from part one of the exercise was:

Acute myeloid leukemia (AML) is a

AML: WHO classification, biology and prognosis. Dimitri Breems, MD, PhD Internist-Hematoloog Ziekenhuis Netwerk Antwerpen

Journal of the Egyptian Nat. Cancer Inst., Vol. 13, No. 3, September: , 2001

Stem cell transplantation for patients with AML in Republic of Macedonia: - 15 years of experience -

Fluorescence in-situ Hybridization (FISH) ETO(RUNX1T1)/AML1(RUNX1) or t(8;21)(q21.3;q22)

Form 2011 R4.0: Acute Lymphoblastic Leukemia (ALL) Pre-HCT Data

BCR-ABL1 positive Myeloid Sarcoma Nicola Austin

The Adult Leukemias - Part 1: Acute Myeloid Leukemia

Extramedullary precursor T-lymphoblastic transformation of CML at presentation

STUDY OF PROGNOSIS IN ACUTE MYELOID LEUKEMIAS (AML) BY CLUSTER ANALYSIS

Introduction. of some recurrent aberrations, for example, 8, del(9q), or CLINICAL OBSERVATIONS, INTERVENTIONS, AND THERAPEUTIC TRIALS

Remission induction in acute myeloid leukemia

Diagnostic challenge: Acute leukemia with biphenotypic blasts and BCR-ABL1 translocation

Original article. Influence of cigarette smoking on the presentation and course of acute myeloid leukemia

HLA-DR-matched Parental Donors for Allogeneic Hematopoietic Stem Cell Transplantation in Patients with High-risk Acute Leukemia

Aberrant Expression of CD7 in Myeloblasts Is Highly Associated With De Novo Acute Myeloid Leukemias With FLT3/ITD Mutation

Early Clearance of Peripheral Blood Blasts Predicts Response to Induction Chemotherapy in Acute Myeloid Leukemia

Extramedullary Relapse of the AML Transformed from MDS Following Auto-HSCT: A Case Report

HTA. Efficacies of HTA regimen for acute myeloid leukemia AML. J of Wannan Medical College HHT THP AML 1 DA AML

DOWNLOAD OR READ : TREATMENT OF ACUTE LEUKEMIAS NEW DIRECTIONS FOR CLINICAL RESEARCH REPRINT PDF EBOOK EPUB MOBI

Appendix 6: Indications for adult allogeneic bone marrow transplant in New Zealand

Leukemia and Myelodysplastic Syndromes

First relapsed childhood ALL Role of chemotherapy

Myeloid neoplasms. Early arrest in the blast cell or immature cell "we call it acute leukemia" Myoid neoplasm divided in to 3 major categories:

Introduction CLINICAL OBSERVATIONS, INTERVENTIONS, AND THERAPEUTIC TRIALS

Juvenile Myelomonocytic Leukemia Pre-HCT Data

Long-term outcomes of Acute Myeloid Leukemia in Adults in Pakistan

Acute Leukemia in Adults: Recent Developments in Diagnosis and Treatment

Flow Cytometry. Leukemia and Myelodysplastic Syndromes. Bone Marrow Aspirate and Biopsy

Prospective Randomization Trial of G-CSF-Primed Induction Regimen versus Standard Regimen in Patients with AML

Form 2012 R3.0: Chronic Myelogenous Leukemia (CML) Pre-Infusion Data

Peking University People's Hospital, Peking University Institute of Hematology

Imatinib Mesylate in the Treatment of Chronic Myeloid Leukemia: A Local Experience

Acute Lymphoblastic and Myeloid Leukemia

Leukemia and Myelodysplastic Syndromes

Management of Extramedullary Leukemia as a Presentation of Acute Myeloid Leukemia

N Engl J Med Volume 373(12): September 17, 2015

Background CPX-351. Lancet J, et al. J Clin Oncol. 2017;35(suppl): Abstract 7035.

Beyond the CBC Report: Extended Laboratory Testing in the Evaluation for Hematologic Neoplasia Disclosure

Mantle Cell Lymphoma

ADx Bone Marrow Report. Patient Information Referring Physician Specimen Information

Charles Mxxx DCEM2 Toulouse Purpan Medical School 01/26/2012 ECN Item 162

Supplemental Material. The new provisional WHO entity RUNX1 mutated AML shows specific genetics without prognostic influence of dysplasia

SUPPLEMENTARY FIG. S3. Kaplan Meier survival analysis followed with log-rank test of de novo acute myeloid leukemia patients selected by age <60, IA

Better Prognosis for Patients With Del(7q) Than for Patients With Monosomy 7 in Myelodysplastic Syndrome

Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA and 2

SH/EAHP WORKSHOP 2017 CASE 210 PRESENTATION

Transcription:

Acute Myeloid Leukemia with CD56 Expression HIGH INCIDENCE OF CD56 EXPRESSION AND RELAPSE RATE IN ACUTE MYELOID LEUKEMIA PATIENTS WITH t(8;21) IN TAIWAN Chi-Huang Hsiao, 1,3 Jih-Luh Tang, 1 Ming Yao, 1 Woei Tsay, 1 Chiu-Hwa Wang, 2 Yao-Chang Chen, 2 Ming-Ching Shen, 2 and Hwei-Fang Tien 1 Background and Purpose: CD56 is a marker of natural killer cells, but can also be found on blast cells in acute myeloid leukemia (AML). The prognostic implications of CD56 expression in AML are not clear. In this study, we evaluated the correlation among CD56 expression, cytogenetic abnormality, and clinical outcome in AML. Methods: CD56 expression was analyzed in leukemic cells from 94 adults with primary AML in Taiwan and was correlated with clinical and hematologic features, cytogenetics, and immunophenotypes of the leukemia. Results: Thirty patients (32%) showed CD56 expression. CD56 + AML patients had a higher lactate dehydrogenase level than CD56 patients (1,136 vs 730 U/L, p = 0.048). Patients with t(8;21) had a significantly higher incidence (89%, 8/9) of CD56 positivity in leukemic cells than those with normal karyotype or other cytogenetic abnormalities (26%, 22/85, p < 0.001). In general, there was no difference in overall survival time in CD56 + and CD56 AML patients. However, three patients had central nervous system involvement at initial presentation; two of these had concomitant CD56 expression and t(8;21). In addition, five of the seven patients with t(8;21) and CD56 expression who achieved complete remission later relapsed. Conclusions: The incidence of CD56 expression in AML patients with t(8;21) is very high in Taiwan, and it may imply a poor prognosis in this subgroup of patients. (J Formos Med Assoc 2002;101:393 8) Key words: CD56 acute myeloid leukemia CD56 is a 200 to 220 kd glycoprotein expressed predominantly on human natural killer cells, a minor subset of T cells and neural/neuroendocrine tissues [1]. It has been recognized as a neural cell adhesion molecule (N-CAM) [2]. In acute myeloid leukemia (AML), CD56 expression has been demonstrated in leukemic cells from 20% to 40% of patients [3 5]; however, the exact role of CD56 in AML is largely unknown. Some investigators suggest that it may affect and modulate both cell cell and cell substrate interactions [6]. The prognostic implications of CD56 expression in AML are not clear. Controversial results have been reported [3, 4, 7, 8]. Recently, Baer et al reported that 55% of AML patients with t(8;21) showed CD56 expression, which was associated with shorter complete remission (CR) duration and survival [9]. It was also demonstrated that t(15;17)-positive AML patients with CD56 expression had a lower CR rate and shorter survival than those without CD56 expression [10]. Most reports concerning CD56 expression in AML are from Western countries, and only few studies have involved Oriental people [11, 12]. In this study, we evaluated the correlation among CD56 expression, cytogenetic abnormality, and clinical outcome in AML patients. Materials and Methods Patients From October 1996 to November 1999, immunophe- Departments of 1 Internal Medicine and 2 Laboratory Medicine, National Taiwan University Hospital, Taipei, and 3 Department of Internal Medicine, Far Eastern Memorial Hospital, Pan-Chiao, Taipei. Received: 15 November 2001. Revised: 28 January 2002. Accepted: 2 April 2002. Reprint requests and correspondence to: Dr. Hwei-Fang Tien, Department of Internal Medicine, National Taiwan University Hospital, 7 Chung-Shan South Road, Taipei, Taiwan. J Formos Med Assoc 2002 Vol 101 No 6 393

C.H. Hsiao, J.L. Tang, M. Yao, et al notype including CD56 expression was determined in leukemic cells from 94 newly diagnosed patients with primary AML at National Taiwan University Hospital. None of the patients had a history of prior chemotherapy or radiotherapy, and none had a history of other hematologic disorders. The median age was 50 years, ranging from 16 to 84 years. There were 49 males and 45 females. The diagnosis and classification of AML were based on the criteria established by the French-American-British (FAB) Cooperative Study Group [13]. Seventy-five patients received standard 3 + 7 induction chemotherapy with doxorubicin 30 mg/ m 2 /day or idarubicin 12 mg/m 2 per day for 3 days plus cytosine arabinoside (ara-c) 100 mg/m 2 per day for 7 days (in non M3 patients) or all-trans retinoic acid with or without chemotherapy (in M3 patients). The remaining 19 patients did not receive any chemotherapy, or were only treated with low-dose ara-c. Fifty-eight (77%) of the patients who received standard chemotherapy achieved CR. Among them, eight patients received hematopoietic stem cell transplantation (6 allogeneic and 2 autologous), 26 patients received consolidation chemotherapy with regimens similar to those in induction treatment, 16 received high-dose ara-c 2 to 3 g/m 2 twice per day for eight to 12 doses plus one of anthracyclines, two received low-dose ara-c 10 mg/m 2 per day for 14 to 21 days, and the remaining six patients did not receive further consolidation treatment (Table 1). Cytogenetic study Chromosome analysis was performed as described previously [14]. Briefly, bone marrow (BM) cells were harvested either directly or after 1 to 3 days of unstimulated culture. Metaphase chromosomes were banded by the conventional trypsin-giemsa banding Table 1. Correlation of CD56 expression with clinical and hematologic features in patients with acute myeloid leukemia CD56 + CD56 Total p Number of patients 30 64 94 Median age, yr (range) 49 (16 79) 50 (18 84) 50 (16 84) 0.370 Lymphadenopathy (%)* 6 (20) 12 (19) 18 (19) 0.886 Hepatomegaly (%) * 8 (27) 10 (16) 18 (19) 0.205 Splenomegaly (%)* 3 (10) 4 (6) 7 (7) 0.676 CNS involvement (%)* 2 (7) 1 (2) 3 (3) 0.238 Cytogenetics** < 0.001 t(8;21) 8 (89%) 1 (11%) 9 Others 22 (26%) 63 (74%) 85 Hemoglobin (median, g/dl) 8.5 7.9 8.0 0.901 WBC (median, /µl) 18,845 13,920 15,550 0.408 Blast (median, %) 60 44 51 0.541 Platelet (median, x10 3 /µl) 36 41 40 0.244 Lactate dehydrogenase (median, U/L) 1136 730 900 0.048 FAB subtype, n M0 1 (20%) 4 (80%) 5 1.000 M1 8 (32%) 17 (68%) 25 0.991 M2 12 (40%) 18 (60%) 30 0.250 M3 3 (25%) 9 (75%) 12 0.746 M4 3 (19%) 14 (81%) 17 0.163 M5 3 (60%) 2 (40%) 5 0.323 CR rate (%) 90.5% (19/21) 72.2 %(39/54) 77.3 (58/75) 0.127 Consolidation C/T 0.366 Standard 10 (52%) 16 (41%) 26 High dose 2 (11%) 14 (36%) 16 Low dose 1 (5%) 1 (3%) 2 No treatment 3 (16%) 3 (8%) 6 Transplantation 3 (16%) 5 (13%) 8 CR duration (median, mo) 13 12 12 0.9106 Survival (median, mo) 23 18 23 0.3362 CNS = central nervous system; WBC = white blood cell count; FAB = French-American-British; C/T = chemotherapy. *% patients with characters among those with or without CD56 expression; numbers in parenthesis represent % patients with or without CD56 expression among subgroups; consolidation chemotherapy in patients obtaining complete response (CR): standard = conventional dose of cytosine arabinoside (ara-c) plus anthracycline; high dose = high dose of ara-c plus anthracycline; low dose = low dose of ara-c. 394 J Formos Med Assoc 2002 Vol 101 No 6

Acute Myeloid Leukemia with CD56 Expression technique and karyotyped according to the International System for Human Cytogenetic Nomenclature (ISCN) [15]. Cytochemical staining and immunophenotyping Air-dried smears of BM aspirates, stained by Riu s method (Romanowsky system), were observed for morphologic abnormalities [16, 17]. The smears were also stained routinely for myeloperoxidase, chloroacetate esterase, and α-naphthyl butyrate esterase, as described previously [16]. A panel of monoclonal antibodies to myeloidassociated antigens including CD13, CD33, CD14, CD11b, CD15, CD41, and glycophorin A, as well as to lymphoidassociated antigens including CD2, CD5, CD7, CD10, CD19, and CD20, lineage-nonspecific antigens HLA-DR and CD34, and CD56, were selected to characterize immunologic phenotypes of leukemic cells. Expression of surface antigens on leukemic cells was shown by an indirect immunoalkaline phosphatase method [18]. Samples in which more than 20% of leukemic cells were positively stained with antibody were considered positive for that marker. Statistical methods Continuous variables were compared by Wilcoxon ranksum test and discrete variables were compared by chisquare or Fisher s exact test. Curves of survival and CR duration were plotted using the Kaplan-Meier method; differences between curves were analyzed using the log-rank test. Results Correlation of CD56 expression with clinical and hematologic features Thirty (32%) patients were found to have CD56 expression in leukemic cells. Comparison of clinical and hematologic features between the patients with and without CD56 expression are summarized in Table 1. There was no statistical difference in age distribution, incidence of lymphadenopathy, hepatomegaly or splenomegaly, white blood cell (WBC) count, percentage of blasts, hemoglobin, or platelet count between the two groups of patients. However, lactate dehydrogenase (LDH) was significantly higher in the patients with CD56 expression than in those without (1,136 vs 730 U/L, p = 0.048). Three patients were found to have central nervous system (CNS) involvement at presentation; two had t(8;21) and CD56 expression and the other was CD56-negative. Correlation of CD56 expression with chromosomal change, FAB classification, and expression of other immunologic markers There were adequate metaphase cells for analysis for all patients but one. Fifty-five patients (58%) showed clonal chromosomal abnormalities, including 11 with t(15;17), nine with t(8;21), four with t(7;11) [19], three with 11q23 abnormalities, one with t(3;3), and one with inv(16). Four patients had trisomy 8 (+8), and three had monosomy 7 (-7) or deletion of 7q. Interestingly, eight (89%) of the nine AML cases with t(8;21) showed CD56 positivity, compared with 22 (26%) patients with normal karyotype or other chromosomal abnormalities (p < 0.001). Patients with t(15; 17) showed a similar incidence of CD56 expression (3/ 11, 27%) to others. The percentage of CD56 + leukemic cells in BM from the eight patients with t(8;21) and CD56 expression ranged from 41% to 100% with a mean of 82% (Table 2 and Figure). Among FAB subtypes, M5 had the highest incidence of CD56 expression (60%), followed by M2 and M1 (Table 1). The correlation of CD56 positivity with expression of myeloid- and lymphoid-associated surface markers was also analyzed. There was no difference between these two groups. However, when patients with specific chromosomal abnormalities t(8;21) and t(15;17) were excluded, those with CD56 expression showed a significantly lower incidence of CD34 expression than others (37% vs 69%, p = 0.028). The former group of patients also had a trend of lower frequency of HLA-DR expression than the latter (72% vs 89%, p = 0.125). Correlation of CD56 expression with outcome Among the 75 patients who received standard induction chemotherapy, CR rate, CR duration, and overall survival were not significantly different between patients with and without CD56 expression (Table 1). However, although all seven patients who had t(8;21) and CD56 expression and were treated achieved CR, five (71%) relapsed (Table 2). This incidence was higher than that in other CD56 + patients without t(8; 21) (42%) or in CD56 patients (46%), but the difference was not statistically significant (p = 0.350 and p = 0.414, respectively), probably because of the small number of patients. The same was also true if only patients receiving standard or more intensive postinduction treatment were included in the analysis (60% vs 40% vs 43%). Because the number of patients was limited, no statistical difference could be found in the outcome between t(15;17) patients with or without CD56 expression. The two CD56 + APL patients who were treated in this study remained in the first CR for 2 and 24 months, respectively. J Formos Med Assoc 2002 Vol 101 No 6 395

C.H. Hsiao, J.L. Tang, M. Yao, et al Table 2. Immunophenotype of leukemic cells and outcome in acute myeloid leukemia patients with t(8;21) and CD56 expression UPN Markers (% cells with positive staining)* CR HLA- CD13 CD33 CD11b CD14 CD19 CD15 CD34 CD56 DR Post-induction Relapse CR Survival treatment duration (mo) (mo) 436 428 460 471 478 508 514 474 97 48 10 0 0 29 19 85 95 Yes No Yes 4 +14 100 57 75 0 0 16 53 58 41 NT 95 42 66 0 0 20 40 89 95 Yes high dose No +36 +37 ND 60 54 18 10 15 80 42 85 Yes auto-bmt Yes 12 13 100 42 88 1 0 81 75 90 48 Yes No Yes 4 10 100 56 85 15 0 50 92 89 100 Yes allo-bmt No +8 +9 100 99 90 ND 2 0 48 58 100 Yes high dose Yes 2 4 81 42 7 18 ND 52 ND 84 94 Yes allo-bmt Yes 21 25 *Positivity 20% leukemic cells react with antibody. CD2, CD5, CD7 and CD10 negative. UPN = unique patient number; allo-bmt = allogeneic bone marrow transplantation; auto-bmt = autologous bone marrow transplantation; CR = complete remission; high dose = high-dose cytosine arabinoside (ara-c) plus anthracycline; ND = not done; NT = no induction chemotherapy. Discussion As demonstrated in this study, AML patients with t(8;21) had a significantly higher frequency of CD56 expression in leukemic cells than others. Patients with normal karyotype, t(15;17), and other chromosomal abnormalities had similar incidences of CD56 expression. Furthermore, with a similar incidence of CD56 expression in total AML to that reported by other centers [4, 5, 7, 12], the frequency of CD56 expression in patients with t(8;21) in this area was truly very high, compared with 55% reported by Baer et al [9] and 54% reported by Seymour et al [4]. The percentage of CD56 + leukemic cells in these patients was also high (mean, 82%; range, 41 100%) (Table 2 and Figure). Both of the two additional cases with t(8;21) referred from other hospitals, who were not included in this study, also showed CD56 expression in leukemic blasts (data not shown). Uneven geographic distribution of nonrandom chromosome aberrations in malignant disorders has been reported [20]. The incidence of t(8; 21) among AML M2 patients with abnormal karyotype is higher in Japan and Taiwan than in Western countries [14, 20]. Whether the distribution of CD56 ex- A B C Figure. A) Smear of bone marrow aspirate from one acute myeloid leukemia (AML) patient with t(8;21) (Liu stain). B) Immunoalkaline phosphatase (IAP) staining using normal mouse ascites as negative control. C) IAP staining using anti-cd56 antibody showing positive staining on leukemic cells (red). 396 J Formos Med Assoc 2002 Vol 101 No 6

Acute Myeloid Leukemia with CD56 Expression pression in AML with t(8;21) is also different in various geographic areas merits further study. Other antigen expression, including positivity for CD19 and CD34 (Table 2), in CD56 + AML with t(8;21) was similar to that of total t(8;21) AML [5, 21]. An association of CD56 expression in AML and extramedullary disease has been demonstrated in some reports [12, 22] but not others [4, 9]. In this study, there were no differences in the incidence of lym-phadenopathy, hepatomegaly, or splenomegaly between patients with CD56 expression and those without. Two of the 30 (6.7%) CD56 + patients showed CNS leukemia at presentation, compared with one of the 64 (1.6%) CD56, but the difference was not statistically significant. Baer et al demonstrated that granulocytic sarcomas were present exclusively in t(8;21) cases with CD56 expression [9], and Seymour et al also found that CNS leukemia was detected only in CD56 +, but not in CD56, AML patients [4], though the differences were not statistically significant in both reports. The incidence of leukemic infiltration of lymph nodes, liver, or spleen was similar between CD56 + and CD56 patients in the latter study [4], as in this series. It is possible that the occurrence of extramedullary leukemia involving lymph nodes, liver, or spleen is similar between AML patients with and without CD56 expression, but that involving CNS or soft tissue is more frequent in the CD56 + AML patients than CD56 patients. Studies on larger groups of patients are needed to clarify this point. Conflicting results regarding the prognostic implications of CD56 expression in AML have been reported [3, 4, 7 10]. In this study, CR rate, CR duration, and survival were not significantly different between AML patients with CD56 expression and those without. These results were similar to those reported by Thomas et al [8] and Seymour et al [4]. However, Lauria et al reported a shorter survival in CD56 + patients than in others [7], while Vidriales et al found a trend toward better outcome in these patients [3]. It may be more meaningful to perform analysis in subgroups of this heterogeneous disease separately than to do it in all patients. According to a recent study [9], CD56 expression is associated with a short remission duration and survival in AML patients with t(8;21)(q22;q22). Because all but one patient with t(8;21) in this study showed CD56 expression, a comparison of outcome between CD56 + and CD56 cases with t(8;21) was impossible. However, patients with t(8;21) and CD56 expression had a high relapse rate (71%), compared with the good prognosis in total t(8;21) cases reported in the literature [23, 24]. In a previous study, we found that 16 of 17 patients with t(8;21) who were treated obtained a CR; excluding the two patients who did not receive post-induction chemotherapy, seven of the nine patients treated with standard-dose consolidation chemotherapy relapsed, while only one of the five patients who received allogeneic BM transplantation did so [14]. It seems obvious that patients with t(8;21) in Taiwan have a high relapse rate if they are only treated with conventional consolidation chemotherapy. We have noticed this condition for some time, but could not find any cause to explain it in the past. The high incidence of CD56 expression in leukemic cells from patients with t(8;21) shown in this study may explain the high relapse rate in these patients in Taiwan. Whether high-dose chemotherapy or hematopoietic stem cell transplantation may prolong CR duration and prevent relapse in CD56 + t(8;21) patients needs to be determined. ACKNOWLEDGMENTS: This study was supported in part by grants from the National Science Council of the Republic of China, NSC 89-2314-B002-341. References 1. Cunningham BA, Hemperly JJ, Murray BA, et al: Neural cell adhesion molecule: structure, immunoglobulin-like domains, cell surface modulation, and alternative RNA splicing. Science 1987;236:799 806. 2. Lanier LL, Testi R, Bindl J, et al: Identity of Leu-19 (CD56) leukocyte differentiation antigen and neural cell adhesion molecule. J Exp Med 1989;169:2233 8. 3. Vidriales MB, Orfao A, Gonzalez M, et al: Expression of NK and lymphoid-associated antigens in blast cells of acute myeloblastic leukemia. Leukemia 1993;7:2026 9. 4. Seymour JF, Pierce SA, Kantarjian HM, et al: Investigation of karyotypic, morphologic and clinical features in patients with acute myeloid leukemia blast cells expressing the neural cell adhesion molecule (CD56). Leukemia 1994;8:823 6. 5. Dunphy CH: Comprehensive review of adult acute myelogenous leukemia: cytomorphological, enzyme cytochemical, flow cytometric immunophenotypic, and cytogenetic findings. J Clin Lab Anal 1999;13:19 26. 6. Acheson A, Sunshine JL, Rutishauser U: NCAM polysialic acid can regulate both cell cell and cell substrate interactions. J Cell Biol 1991;114:143 53. 7. Lauria F, Raspadori D, Birtolo S, et al: CD56 antigenic expression in acute myeloid leukemia identifies patients with poor clinical outcome. Abstracts from ISH 2000, Toronto, Canada, August 26 to 30, 2000. Abstract 15153. 8. Thomas X, Vila L, Campos L, et al: Expression of N-CAM (CD56) on acute leukemia cells: relationship with disease characteristics and outcome. Leuk Lymphoma 1995; 19:295 300. 9. Baer MR, Stewart CC, Lawrence D, et al: Expression of the neural cell adhesion molecule CD56 is associated with short remission duration and survival in acute myeloid leukemia with t(8;21)(q22;q22). Blood 1997;90:1643 8. J Formos Med Assoc 2002 Vol 101 No 6 397

C.H. Hsiao, J.L. Tang, M. Yao, et al 10. Murray CK, Estey E, Paietta E, et al: CD56 expression in acute promyelocytic leukemia: a possible indicator of poor treatment outcome? J Clin Oncol 1999; 17:293 7. 11. Suzuki R, Yamamoto K, Seto M, et al: CD7+ and CD56+ myeloid/natural killer cell precursor acute leukemia: a distinct hematolymphoid disease entity. Blood 1997; 90:2417 28. 12. Kuwabara H, Nagai M, Yamaoka G, et al: Specific skin manifestations in CD56 positive acute myeloid leukemia. J Cutan Pathol 1999;26:1 5. 13. Bennett JM, Catovsky D, Daniel MT, et al: Proposed revised criteria for the classification of acute myeloid leukemias. A report of the French-American-British Cooperative Group. Ann Intern Med 1985;103:626 9. 14. Tien HF, Wang CH, Lin MT, et al: Correlation of cytogenetic results with immunophenotype, genotype, clinical features, and ras mutation in acute myeloid leukemia. Cancer Genet Cytogenet 1995;84:60 8. 15. Mitelman F, ed: ISCN 1995: An International System for Human Cytogenetic Nomenclature (1995). Basel, Farmington, CT: Karger, 1995. 16. Tien HF, Wang CH, Su IJ, et al: A subset of acute nonlymphocytic leukemia with expression of surface antigen CD7 morphologic, cytochemical, immunocytochemical and T cell receptor gene analysis on 13 patients. Leuk Res 1990;14:515 23. 17. Riu CH: A study on method of staining blood films (Romanowsky system). J Niigata Med Assoc 1957; 70:635 43. 18. Tien HF, Wang CH, Chen YC, et al: Characterization of acute myeloid leukemia (AML) coexpressing lymphoid markers: different biologic features between T-cell antigen positive and B-cell antigen positive AML. Leukemia 1993;7:688 95. 19. Huang SY, Tang JL, Liang YJ, et al: Clinical, haematological and molecular studies in patients with chromosome translocation t(7;11): a study of four Chinese patients in Taiwan. Br J Haematol 1997:96:682 7. 20. Johansson B, Mertens F, Mitelman F: Geographic heterogeneity of neoplasia-associated chromosome aberrations. Genes Chromosomes Cancer 1991;3:1 7. 21. Ferrara F, Di Noto R, Annunziata M, et al: Immunophenotypic analysis enables the correct prediction of t(8; 21) in acute myeloid leukaemia. Br J Haematol 1998; 102:444 8. 22. Byrd JC, Edenfield WJ, Shields DJ, et al: Extramedullary myeloid cell tumors in acute nonlymphocytic leukemia: a clinical review. J Clin Oncol 1995;13:1800 16. 23. Grimwade D, Walker H, Oliver F, et al: The importance of diagnostic cytogenetics on outcome in AML: analysis of 1,612 patients entered into the MRC AML 10 trial. Blood 1998;92:2322 33. 24. Schiffer CA, Lee EJ, Tomiyasu T, et al: Prognostic impact of cytogenetic abnormalities in patients with de novo acute nonlymphocytic leukemia. Blood 1989;73:263 70. 398 J Formos Med Assoc 2002 Vol 101 No 6