unselected anemic patients with myelodysplastic syndrome ...

4 downloads 0 Views 141KB Size Report
Antithymocyte globulin (ATG) has re- cently been popularized as an effective treatment in myelodysplastic syndrome. (MDS). We treated 8 anemic MDS patients.
From bloodjournal.hematologylibrary.org by guest on May 24, 2011. For personal use only.

2003 101: 2156-2158 Prepublished online October 31, 2002; doi:10.1182/blood-2002-09-2867

Antithymocyte globulin has limited efficacy and substantial toxicity in unselected anemic patients with myelodysplastic syndrome David P. Steensma, Angela Dispenzieri, S. Breanndan Moore, Georgene Schroeder and Ayalew Tefferi

Updated information and services can be found at: http://bloodjournal.hematologylibrary.org/content/101/6/2156.full.html Articles on similar topics can be found in the following Blood collections Hematopoiesis and Stem Cells (2837 articles) Neoplasia (4217 articles) Immunotherapy (577 articles) Brief Reports (1405 articles) Clinical Trials and Observations (3153 articles) Information about reproducing this article in parts or in its entirety may be found online at: http://bloodjournal.hematologylibrary.org/site/misc/rights.xhtml#repub_requests Information about ordering reprints may be found online at: http://bloodjournal.hematologylibrary.org/site/misc/rights.xhtml#reprints Information about subscriptions and ASH membership may be found online at: http://bloodjournal.hematologylibrary.org/site/subscriptions/index.xhtml

Blood (print ISSN 0006-4971, online ISSN 1528-0020), is published weekly by the American Society of Hematology, 2021 L St, NW, Suite 900, Washington DC 20036. Copyright 2011 by The American Society of Hematology; all rights reserved.

From bloodjournal.hematologylibrary.org by guest on May 24, 2011. For personal use only. CLINICAL OBSERVATIONS, INTERVENTIONS, AND THERAPEUTIC TRIALS

Brief report

Antithymocyte globulin has limited efficacy and substantial toxicity in unselected anemic patients with myelodysplastic syndrome David P. Steensma, Angela Dispenzieri, S. Breanndan Moore, Georgene Schroeder, and Ayalew Tefferi

Antithymocyte globulin (ATG) has recently been popularized as an effective treatment in myelodysplastic syndrome (MDS). We treated 8 anemic MDS patients (refractory anemia [RA] and refractory anemia with excess blasts [RAEB-1]) with ATG (40 mg/kg/d for 4 days) and prednisone in a phase 2 trial. The study was stopped early according to a preset termi-

nation rule because of lack of efficacy. There were no salutary responses. Toxicities included serum sickness (in all patients), transient neutropenia and thrombocytopenia, diarrhea, vomiting, and syncope with a generalized seizure. At least 3 patients had the HLA-DR15 (DR2) allele. We conclude that the risk-benefit ratio of ATG in an unselected population

of MDS patients may be unfavorable, and more work is needed to define the subset of patients who will respond to ATG before its widespread use can be recommended. (Blood. 2003;101:2156-2158)

© 2003 by The American Society of Hematology

Introduction The myelodysplastic syndrome (MDS) includes a heterogeneous group of marrow disorders characterized by ineffective hematopoiesis and a variable risk of leukemic transformation. Because most patients with MDS are diagnosed after the age of 60 years, therapy is often limited to supportive care with transfusions and hematopoietic growth factors. A subset of younger patients may benefit from hematopoietic stem cell transplantation, but new, nontoxic treatments are sorely needed. Suppression of hematopoiesis by cytotoxic T cells may contribute to anemia in some patients with MDS.1 MDS can be associated with vasculitides, T-cell large granular lymphocytic disease, and autoimmune conditions, suggesting the presence of immune dysregulation in at least a subset of MDS sufferers and implying that immunomodulation might provide palliative benefit.2,3 Antithymocyte globulin (ATG), derived from immunization of horses or other animals with human thoracic duct lymphocytes, has complex activity within the human hematopoietic milieu. ATG suppresses cytotoxic and potentially inhibitory T lymphocytes, may indirectly stimulate hematopoiesis via augmentation of hematopoietic growth factor release by T cells and stromal cells, and promotes cellular differentiation.4-6 ATG has long enjoyed a well-established clinical role in the transplantation setting and in the treatment of aplastic anemia.7 More recently, 34% of patients with MDS have been reported to achieve red cell transfusion independence after 4 days of ATG treatment; platelet responses were observed in 48% of patients with severe thrombocytopenia, and neutrophil responses were observed in 55% of patients.8 Based on an earlier encouraging report from that cohort, we conducted a nonrandomized, single-institution, single-arm phase 2 trial of ATG in MDS.9

From the Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN; Tissue Typing Laboratory, Division of Transfusion Medicine, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN; and Division of Biostatistics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN. Submitted September 19, 2002; accepted October 20, 2002. Prepublished online as Blood First Edition Paper, October 31, 2002; DOI 10.1182/blood-2002-09-2867.

2156

Study design Eight anemic (hemoglobin less than 9 g/dL [90 g/L]) MDS patients (5 women; ages 62 to 74 years) with refractory anemia (RA) (2 patients) and refractory anemia with excess blasts (RAEB-1) (6 patients) were recruited from the Mayo Clinic hematology practice between 1998 and 2002. Patient characteristics are given in Table 1. Patients with refractory anemia with ringed sideroblasts (RARS) were excluded because of lack of response to ATG in the study that prompted this trial.9 Patients with secondary (therapy-related) MDS, poor liver or renal function (bilirubin more than 2 mg/dL [34.2 ␮M] or creatinine more than 2 mg/dL [176.8 ␮M]), HIV infection, active nonmyeloid malignancy, or poor performance status (Eastern Cooperative Oncology Group [ECOG] 3 or 4) were also excluded. All patients gave informed consent prior to enrollment, and the Mayo Clinic Institutional Review Board approved the study. ATG (ATGAM; Pharmacia, Peapack, NJ) was administered intravenously at a dose of 40 mg/kg/d for 4 days. Oral prednisone was given at a dose of 1 mg/kg/d and then rapidly tapered unless persistent rash and/or serum sickness prohibited this. All patients had 10% or fewer marrow blasts at trial enrollment. According to the International Prognostic Scoring System (IPSS), 5 patients fit into the intermediate-1 (INT-1) risk group and 3 were INT-2.10 The bone marrow was hypocellular in 2 patients (both had normal flow cytometric testing for a paroxysmal nocturnal hemoglobinopathy clone [CD59, CD14, and fluorescently labeled inactive variant of aerolysin (FLAER)]), normocellular in 1 patient, and hypercellular in 5 patients. Bone marrow karyotypes are listed in Table 1. Peripheral blood T-cell gene rearrangement studies were done in 7 of 8 patients and failed to demonstrate a clonal T-cell population. After this study was initiated, reports appeared of an increased frequency of HLA-DR15 (DR2) in MDS patients and positive predictive value of this allele with regard to response to immunosuppressive therapies such as ATG.11 To study the class II HLA profile of patients in this trial, DNA was extracted from methanol-glacial acetic acid–fixed cells that had been preserved at the time of

Reprints: David P. Steensma, Mayo Clinic, 200 First St, SW, Rochester, MN 55905; e-mail: [email protected]. The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked ‘‘advertisement’’ in accordance with 18 U.S.C. section 1734. © 2003 by The American Society of Hematology

BLOOD, 15 MARCH 2003 䡠 VOLUME 101, NUMBER 6

From bloodjournal.hematologylibrary.org by guest on May 24, 2011. For personal use only. BLOOD, 15 MARCH 2003 䡠 VOLUME 101, NUMBER 6

ATG EFFICACY AND TOXICITY IN MDS

2157

Table 1. Characteristics of patients enrolled in the Mayo Clinic ATG trial

Patient no.

Sex

Age, y

1

F

2

Treatments prior to ATG trial enrollment†

Date of initial MDS diagnosis

Alive or dead (specific cause of death)‡

8/94

Died 2/00

WHO diagnosis

IPSS

Date of trial enrollment*

62

RAEB-1

INT-1

4/98

EPO,

F

74

RA

INT-1

6/98

Amifostine

9/97

Died 9/00

46,XX,del(5)(q13q33),idic(X)(p10)[13]/46,

3

F

70

RAEB-1

INT-1

10/99

None

9/99

Died 1/00

46,XX[30]

4

M

63

RAEB-1

INT-1

10/01

Thalidomide,

10/99

Alive 9/02

46,XY,del(5)(q15q33),add(21)(q11.2)[8]/

6/01

Alive 9/02

47,XY,⫹8[2]/46,XY[18]

deferoxamine

Karyotype 46,XX[30]

(AML) XX,del(5)(q13q33)[2]/46,XX[5]

EPO 5

M

6 7

INT-2

11/01

EPO

46,XY[12]

74

RAEB-1

M

62

RA

INT-1

1/02

EPO

2/01

Alive 9/02

45,X,⫺Y[13]/46,XY[7]

F

73

RAEB-1

INT-2

3/02

None

11/01

Died 6/02

44,XX,⫺2,del(5)(q13q33),add(16)(q24),

(sepsis) 8

F

72

RAEB-1

INT-2

5/02

None

9/01

Alive 9/02

psu dic(19;20)(p13.3;q13.3),⫺21,⫹mar[17] 46,XX,⫹8,der(1;7)(q10;p10)[20]

WHO indicates World Health Organization classification of hematopoietic and lymphoid neoplasms; RA, refractory anemia; RAEB-1, refractory anemia with excess blasts; IPSS, International Prognostic Scoring System for MDS (of the International MDS Risk Analysis Workshop); INT-1 and INT-2, IPSS intermediate-1 and intermediate-2 risk categories; EPO, recombinant human erythropoietin. *The first 3 patients enrolled in this trial paid for ATG themselves. A large number of other MDS patients seen at the Mayo Clinic during the years 1998, 1999, and 2000 were also approached about participation in this trial but declined enrollment (exact records of the number of patients approached were not kept). The most common reasons patients declined enrollment were concern about the cost of the drug and administration fees and/or inability to stay in Rochester, MN, for the expected duration of treatment. In 2001, Pharmacia agreed to provide the study drug free of cost to patients, which allowed more rapid accrual of 5 remaining patients. †Not including transfusions or antimicrobial support for infections. ‡Patients 1, 2, 3, and 7 were followed until their deaths 22, 27, 15, and 3 months after ATG treatment, respectively. None of these patients enjoyed a hematologic improvement. Patients 4, 5, 6, and 8 are still alive and have been followed for 11, 10, 8, and 4 months, respectively; patients 6 and 8 were started on other therapies 3 months after ATG therapy (thalidomide and EPO) when no signs of improvement had been seen.

initial marrow cytogenetic studies using the High Pure PCR Template Preparation Kit (Roche Molecular Biochemicals, Indianapolis, IN). Class II HLA DNA was amplified via polymerase chain reaction (PCR) and typed according to established PCR with sequence-specific primers (SSP) technique.

Results and discussion Unfortunately, there were no hematologic responses to ATG and prednisone in the dose and schedule used in this study, and transfusion needs were unaffected. Enrollment of 20 patients was planned, but the study was terminated early due to a predetermined stopping rule. Systemic toxicity potentially attributable to the drug included classic serum sickness and/or rash in all 8 patients (7 fever, 4 rash, 4 myalgias/arthralgias), diarrhea (2 patients), nausea/vomiting (1 patient), syncope with a generalized tonic-clonic seizure in a patient with no seizure history (1 patient, during the first day of therapy), and reactivation of oral herpes simplex virus (HSV) (1 patient). All toxicities eventually resolved. Transient hematopoietic toxicity was observed but did not lead to any complications. These toxicities included worsening of neutropenia in 1 of 4 already neutropenic patients, new neutropenia in an additional patient, worsening of pre-existing thrombocytopenia in 6 patients, and new, mild thrombocytopenia in the 2 patients who had a normal platelet count at trial enrollment (platelet count of 253 ⫻ 109/L fell to 60 ⫻ 109/L, and 412 ⫻ 109/L fell to 145 ⫻ 109/L). The nadir platelet count occurred on day 5 of therapy in 6 of 8 patients, and recovery to pretreatment values was rapid. A sufficient quantity of DNA for HLA typing (range, 47.2 to 70.3 ␮g) was obtained from 7 of 8 patient samples. In 5 patients, this DNA was of high enough quality to allow successful amplification and HLA class II typing; the other 2 patients are deceased, so additional genetic material is unavailable. In 3 of 5 patients (60%), HLA-DR15 (DR2) was detected; the expected frequency of HLA-DR15 (DR2) in North Americans is 15% to 21%.11

With 0 hematopoietic responses in 8 patients, the 95% confidence interval for the true response rate to ATG and prednisone in a similar population of MDS patients is 0% to 37%. Although this does not statistically exclude the 34% red cell response rate seen in the study by Molldrem et al, the response rate in this cohort was disappointing.8 It is possible that the lack of response occurred by chance alone, but the group enrolled in this study might actually have been expected to have a higher probability of including ATG-responding patients than a more typical consecutive series of MDS patients seen in clinical practice. There were 2 patients in this cohort with a hypocellular marrow (hypocellular MDS may have pathophysiologic overlap with aplastic anemia, which often responds to ATG,12 and 38% of patients in the series reported by Molldrem et al were hypocellular9—more than in most general MDS series), no patients had more than 10% blasts, patients with secondary MDS (which is typically more refractory to treatment than is de novo disease) were excluded, only 2 enrolled patients had an IPSS high-risk karyotype, at least 3 were HLA-DR15 (DR2)–positive, and 6 of 8 patients were thrombocytopenic at trial enrollment (a low platelet count predicted ATG response in the study by Molldrem et al8). Although there may be a subset of MDS patients who will respond to therapy with ATG, we believe this population is not yet clearly defined. HLA-DR15 (DR2) typing alone is not of sufficient predictive value. In conclusion, in the dose and schedule used in the trial, ATG had no efficacy in a population of patients with intermediate-risk RA and RAEB-1. Given the toxicity observed and the substantial cost of therapy (the direct drug acquisition cost of the 4-day ATG regimen for a typical 70-kg patient is approximately $13,60013), further research is needed before this drug might be widely recommended in MDS.

Acknowledgments We thank Terra Reeder, Cynthia Kroning, and Jack Spurbeck for their valuable technical assistance.

From bloodjournal.hematologylibrary.org by guest on May 24, 2011. For personal use only. 2158

BLOOD, 15 MARCH 2003 䡠 VOLUME 101, NUMBER 6

STEENSMA et al

References 1. Sugawara T, Endo K, Shishido T, et al. T cellmediated inhibition of erythropoiesis in myelodysplastic syndromes. Am J Hematol. 1992;41:304305.

5. Hunter RF, Mold NG, Mitchell RB, Huang AT. Differentiation of normal marrow and HL60 cells induced by antithymocyte globulin. Proc Natl Acad Sci U S A. 1985;82:4823-4827.

10. Greenberg P, Cox C, LeBeau MM, et al. International scoring system for evaluating prognosis in myelodysplastic syndromes. Blood. 1997;89: 2079-2088.

2. Hamblin T. Immunologic abnormalities in myelodysplastic syndromes. Hematol Oncol Clin North Am. 1992;6:571-586.

6. Raefsky EL, Gascon P, Gratwohl A, Speck B, Young NS. Biological and immunological characterization of ATG and ALG. Blood. 1986;68:712719.

11. Saunthararajah Y, Nakamura R, Nam JM, et al. HLA-DR15 (DR2) is overrepresented in myelodysplastic syndrome and aplastic anemia and predicts a response to immunosuppression in myelodysplastic syndrome. Blood. 2002;100: 1570-1574.

3. Saunthararajah Y, Molldrem JL, Rivera M, et al. Coincident myelodysplastic syndrome and T-cell large granular lymphocytic disease: clinical and pathophysiological features. Br J Haematol. 2001;112:195-200. 4. Mangan KF, D’Alessandro L, Mullaney MT. Action of antithymocyte globulin on normal human erythroid progenitor cell proliferation in vitro: erythropoietic growth-enhancing factors are released from marrow accessory cells. J Lab Clin Med. 1986;107:353-364.

7. Young NS. Acquired aplastic anemia. Ann Intern Med. 2002;136:534-546. 8. Molldrem JJ, Leifer E, Bahceci E, et al. Antithymocyte globulin for treatment of the bone marrow failure associated with myelodysplastic syndromes. Ann Intern Med. 2002;137:156-163. 9. Molldrem JJ, Caples M, Mavroudis D, Plante M, Young NS, Barrett AJ. Antithymocyte globulin for patients with myelodysplastic syndrome. Br J Haematol. 1997;99:699-705.

12. Barrett J, Saunthararajah Y, Molldrem J. Myelodysplastic syndrome and aplastic anemia: distinct entities or diseases linked by a common pathophysiology? Semin Hematol. 2000;37:1529. 13. Drug Topics Red Book 2002. Montvale, NJ: Thomson Medical Economics; 2002.