12
1995 85: 3367-3377 NS Young and AJ Barrett The treatment of severe acquired aplastic anemia http://bloodjournal.hematologylibrary.org/site/misc/rights.xhtml#repub_requests Information about reproducing this article in parts or in its entirety may be found online at: http://bloodjournal.hematologylibrary.org/site/misc/rights.xhtml#reprints Information about ordering reprints may be found online at: http://bloodjournal.hematologylibrary.org/site/subscriptions/index.xhtml Information about subscriptions and ASH membership may be found online at: reserved. Copyright 2011 by The American Society of Hematology; all rights 900, Washington DC 20036. weekly by the American Society of Hematology, 2021 L St, NW, Suite Blood (print ISSN 0006-4971, online ISSN 1528-0020), is published use only. For personal at PENN STATE UNIVERSITY on February 23, 2013. bloodjournal.hematologylibrary.org From

The treatment of severe acquired aplastic anemia · 2015-07-29 · tic anemia, most particularly the decision between allogeneic transplantation and immunosuppression and the consider-

  • Upload
    others

  • View
    10

  • Download
    0

Embed Size (px)

Citation preview

Page 1: The treatment of severe acquired aplastic anemia · 2015-07-29 · tic anemia, most particularly the decision between allogeneic transplantation and immunosuppression and the consider-

1995 85: 3367-3377   

 NS Young and AJ Barrett The treatment of severe acquired aplastic anemia

http://bloodjournal.hematologylibrary.org/site/misc/rights.xhtml#repub_requestsInformation about reproducing this article in parts or in its entirety may be found online at:

http://bloodjournal.hematologylibrary.org/site/misc/rights.xhtml#reprintsInformation about ordering reprints may be found online at:

http://bloodjournal.hematologylibrary.org/site/subscriptions/index.xhtmlInformation about subscriptions and ASH membership may be found online at:

reserved.Copyright 2011 by The American Society of Hematology; all rights900, Washington DC 20036.weekly by the American Society of Hematology, 2021 L St, NW, Suite Blood (print ISSN 0006-4971, online ISSN 1528-0020), is published    

use only.For personal at PENN STATE UNIVERSITY on February 23, 2013. bloodjournal.hematologylibrary.orgFrom

Page 2: The treatment of severe acquired aplastic anemia · 2015-07-29 · tic anemia, most particularly the decision between allogeneic transplantation and immunosuppression and the consider-

BLOOD VOL 85, NO 12

The Journal of The Amem'can Society of Hematology

JUNE 15, 1995

PERSPECTIVE

The Treatment of Severe Acquired Aplastic Anemia By Neal S. Young and A. John Barren

F OR MOST OF ITS HISTORY, aplastic anemia has had a dire reputation. In the decades after Ehrlich's original

description of the disease a century ago, many patients died simply of anemia. The introduction of blood product support and antibiotics improved the care of patients, but in Win- trobe's 1956 textbook, aplastic anemia could still be de- scribed as "progressively, inexorably, and more or less rap- idly fatal. . . . Death ensues in the course of a few weeks or life may linger as long as six months." Today, the disease can be cured or ameliorated in most patients. By the end of the 1960s, replacement of the characteristically empty bone marrow by a marrow transplant from a sibling donor was shown to be curative in studies performed by Thomas et all in Seattle and in several European centers.* In the 1970s, the observation of autologous marrow recovery after treat- ment with antilymphocyte sera suggested that nonreplace- ment therapy could restore marrow function. Increasingly refined transplant regimens and intensified immunosuppres- sive therapy, as well as improved transfusion support and modem antimicrobials, have produced spectacular hemato- logic improvement and long-term survival rates, which will be described below.

Unfortunately, these advances are unavailing to patients with severe pancytopenia observed for weeks for the rare spontaneous remission, prescribed useless long courses of corticosteroids, or subjected as an individual to a test course of the newest growth factor. Applications of remedies of doubtful or unproven value are dilatory strategies with often disastrous consequences in a disease infamous for its cascad- ing complications. Even the experienced physician-and well-informed patient-face therapeutic dilemmas in aplas- tic anemia, most particularly the decision between allogeneic transplantation and immunosuppression and the consider- ation of transplant from an unrelated marrow donor.

We deal here with current treatment issues in severe aplas- tic anemia (severity defined by blood count criteria3). Read- ers are referred to recent monographs for fuller discussions and extensive bibli~graphies.~.~

PATHOPHYSIOLOGY OF APLASTIC ANEMIA

Hematopoietic activity is reduced in all patients with aplastic anemia, as reflected in marrow histology, low num- bers of CD34+ cells, and poor colony formation in functional progenitor a ~ s a y s . ~ * ' ~ Bone marrow failure may directly re- sult from dose-dependent destruction of stem cells, as caused

Blood, Vol 85, No 12 (June 15). 1995: pp 3367-3377

by radiation, cytotoxic chemotherapy, and benzene. In some rare individuals, abnormal drug metabolism might produce toxic intermediate compounds that harm marrow cells, but evidence for such a mechanism in patients has been elusive. In a large epidemiologic study, only about one quarter of cases were attributed to drug use." A significant history of radiation, benzene, or chloramphenicol use is now very unusual among patients with community acquired aplastic anemia.

In theory, marrow failure in aplastic anemia could be the consequence of damage either to hematopoietic cells or to the stromal cells required for their production. However, most of the evidence points strongly to effects on hematopoi- etic cells, and stromal cell function and growth factor pro- duction are normal in almost all patients with aplastic ane-

Much, perhaps most, acquired aplastic anemia appears to be secondary to immune system mediated destruction of marrow cells. The immune hypothesis, first inferred from the response of patients to antilymphocyte sera by Math6 et al and from coculture experiments performed at Sloan- Kettering, has been supported by a large variety of subse- quent laboratory experiments and clinical observations. He- matopoietic failure in aplasia likely is mediated by cytotoxic T lymphocytes that are detectable in blood and These cells produce the cytokines y-interferon and tumor necrosis factor+, both of which broadly inhibit in tissue culture both progenitor cell growth and the generation of long-term culture-initiating cells, an in vitro surrogate for the stem cell.I5 The action of inhibitory cytokines is not merely suppressive but destructive, inducing cell death in the CD34+ compartment, probably through Fas-mediated apoptosis.16 y-Interferon, which is not a normal marrow fac- tor, is produced in the marrow of most patients with acquired aplastic anemia.'7'ls The potent activity of a locally secreted inhibitor molecule was shown in recent experiments in our laboratory. Stromal cells, engineered by transduction of the

mia.12.13

From the Hematology Branch, National Heart, Lung, and Blood

Submitted November 28, 1994; accepted February 9, 1995. Address reprint requests to Neal S. Young, MD, Bldg IO, Room

This is a US government work There are no resm'ctiotw on its use.

Institute, Bethesda, MD.

7C103, NIH, Bethesda MD 20892-1652.

ooW-4971/95/8512-00$0.00/0

3367

use only.For personal at PENN STATE UNIVERSITY on February 23, 2013. bloodjournal.hematologylibrary.orgFrom

Page 3: The treatment of severe acquired aplastic anemia · 2015-07-29 · tic anemia, most particularly the decision between allogeneic transplantation and immunosuppression and the consider-

3368 YOUNG AND BARRETT

y-interferon gene to constitutively express low levels of the cytokine, completely suppressed hematopoiesis at measur- able levels about 100-fold lower than equivalently inhibitory concentrations of exogenous y-interferon (Selleri, Macie- jewski, and Young, unpublished data). The large number of clinical associations with aplastic anemia-drugs, viruses, pregnancy, and graft-versus-host disease (GVHD)-suggest that a variety of events can activate the immune system to cause marrow aplasia. Increased representation of certain histocompatibility antigens among aplastic anemia patients implicates host genetic factors in the peculiar immune re- sponse to these

The pathophysiology of the disease suggests two ap- proaches to therapy: replacement of deficient stem cells (and coincidentally of the immune system) by bone marrow trans- plantation or suppression of a destructive immunologic pro- cess. Unfortunately, neither measures of stem cell number nor immune system dysfunction are clinically useful guides to treatment selection in individual patients.

ALLOGENEIC BONE MARROW TRANSPLANTATION

In almost all cases the donor is a full sibling, identical at HLA A, B, and DR histocompatibility loci with the recipient. The 25% probability of finding a fully compatible match within the recipient’s family makes allogeneic transplanta- tion available to only a minority of patients.

Bone marrow transplant data come from two sources. First, a few hospitals accrue sufficient aplastic patients to generate case series. Single-center studies can evaluate spe- cific research protocols, and both protocol design and patient care gain from the high level of expertise in such specialized units. Selection bias before admission, based on the presence of adverse prognostic factors or financial status, is possible, but analysis of current patient data suggest that most trans- plant centers include poor-risk patients in their program (older, heavily transfused, or more chronic cases). Transplant data also have been derived from the International Bone Marrow Transplant Registry and the European Group for Bone Marrow Transplant. Registries are observational data bases, and they usually include diverse patient populations and protocols, which vary with respect to conditioning regi- men and GVHD prophylaxis. Analysis of the data base can be complicated by the heterogeneity of treatment ap- proaches, but registries also allow comparison of different therapeutic strategies. Selection bias may be introduced be- cause reporting is voluntary. However, their substantial sam- ple size and high level of participation make registries likely to accurately represent treatment outcomes in practice. Large patient numbers and long follow-up facilitate statistical anal- yses even if early mortality is high and late events infrequent.

Despite the differences between single-center and registry data, the results of bone marrow transplantation in aplastic anemia from these two sources are in broad agreement (Table 1). Consideration of the confidence intervals in actuarial sur- vival estimates suggest that the figures from the best single- center trials may not differ significantly from registry num- bers. Allogeneic bone marrow transplantation is effective in restoring normal hematopoiesis in the majority of patients to whom it is applied. The main issues are whether mortality

for an unselected group is greater than 30% or less than 10% and how these differences relate to variations in treatment management. Recent results, whether from single centers or registries, show improvement over earlier published reports. In Seattle, survival increased from about 45% between 1970 and 1976 to 60% from 1977-1988 to about 90% in 1988- 1993.” Similarly, for the European Group for Bone Marrow Transplantation, survival increased from 43% in 1970-1979 to 63% in 1980-1989 and 72% in 1990-1994.22 These im- provements have been attributed to multiple factors: more intensified conditioning regimens to prevent graft rejection, the use of cyclosporine both to improve engraftment and prevent GVHD, better blood product preparation that re- duces allosensitization, and improved supportive care with new antimicrobial and antiviral agents. Nevertheless, graft rejection, GVHD, and infection remain limiting factors to the success of transplantation.

Grafr rejection. Hematopoietic failure because of either failure to engraft or engraftment followed by rejection is a major complications in marrow transplant for aplastic ane- mia, related in part to insufficient immunosuppression of the recipient and perhaps also to the underlying immune pathophysiology of the disease. Graft failure has a high mor- tality. Second transplants are only successful in one third of patients, and only rarely do patients recover autologous hematop~ies i s .~’~~~

Engraftment requires an adequate stem cell dose, usually provided by a single marrow harvest that includes donor T lymphocytes in the inoculum.25 Graft rejection is related to presensitization of the recipient to minor histocompatability antigens of the donor by blood product transfusion or preg- nancy. Untransfused or minimally transfused patients have engraftment rates similar to those of identical twin recipi- enhz6 Although transplantation in the untransfused patient may be optimal, in reality, transfusion in the severely pancy- topenic patient is rarely avoidable. The impact of transfusion allosensitization may have been exaggerated. In one registry survey, transfusion of limited numbers of blood products (40 U or less) had only a modest impact on the rate of graft fail~re.’~ Probably the introduction of filtered, leucocyte- depleted red blood cell and platelet products has contributed to reducing pretransplantation alloimmunization.28

Graft rejection rates have decreased over time. Histori- cally, combinations of cyclophosphamide and irradiation were used in the conditioning regimens to ensure en- graftment. Irradiation (and inevitably radiomimetic agents), although reducing deaths from graft failure, increase post- transplant mortality and cause infertility and late malignant disease.29-” More acceptable and currently successful proto- cols that reduce graft failure with less toxicity incorporate prolonged used of cyclosporine p~sttransplant~~.~’ and anti- thymocyte globulin (ATG) in the conditioning regimen.”,”43s

GVHD. GVHD is a major cause of late mortality and morbidity, both as an immediate and a delayed consequence of allogeneic bone marrow transplantation. Up to 80% of matched sibling bone marrow recipients develop some de- gree of acute GVHD in the first 3 months after transplant, and acute GVHD contributes to mortality in about 10% of recipients. Chronic GVHD develops in up to 50% of patients

use only.For personal at PENN STATE UNIVERSITY on February 23, 2013. bloodjournal.hematologylibrary.orgFrom

Page 4: The treatment of severe acquired aplastic anemia · 2015-07-29 · tic anemia, most particularly the decision between allogeneic transplantation and immunosuppression and the consider-

TREATMENT OF SEVERE APLASTIC ANEMIA 3369

Table 1. Recent Results of Allogeneic Bone Marrow TransDlantation in Aolastic Anemia

Rejection1 Graft

Institution Years N Age (yr) Conditioning Rx GVHD Px (%) (%l (%l (%) FN (yr)

H8pital St. Louis"' 1980-89 107 5-46 (19) CY + TA1 MTX, CSA, . 3 36 35 68 5 10 (at 5 yr) 1-10 (3.75)

Johns Hopkins" 1984-91 24 4-53 (21) CY + CSA 12 78 % 15 (at 5 yr) 0.5-5 (2.3)

CSA 29 0 7 9 + 8 2-5 (3) 1988-1993 39 2-52 (24.5) CY + ATG

5 HutchinsonJS MTX + CSA IBMTR"

5 15 1980-87 595 1->40 (-) CY 5 TLI. TAI, MTX, CSA. 10 40 45 63 (at 5 yr)

34 92 (at 3 yr) 0.8-5 12.5)

IBMTR"' 1985-91 737 64 f 4 (at 5 yr) EGBMT" 1980-89 540

1990-94 165

Failure AGVHD CGVHD Actuarial Survival

or MTX + CSA UCLA"2 1984-88 290 0.7-41 (19) CY + T U M T X + CSA 17 21

or TB1 or MTX + CSA

63 (at 5 yr) 72 (at 3.5 yr)

Abbreviations: Px, prophylaxis; Rx, therapy; AGVHD, acute GVHD; CGVHD, chronic GVHD; CY, cyclophosphamide; MTX, methotrexate; CSA. cyclosporine; TBI. total body irradiation; TAI, total abdominal irradiation; TLI, total lymphoid irradiation; ATG, antithymocyte globulin; IBMTR. International Bone Marrow Transplant Registry; EGBMT, European Group for Bone Marrow Tranplantation.

GVHD. prevalence in the susceptible population is the relevant figure, to correct for censoring due to death"'; for the first four studies quoted, the percentages represent Especially when results are reported early, Kaplan-Meier estimates may be higher than ultimate actual survival figures."' For late complications such as chronic

the actual prevalence in the population at risk (either explicit in the publication or extracted from the results), and for the Seattle study, the stated actuarial risk is quoted.

surviving more than 3 months after transplant, in 20% as a serious complication, and in 5% as a fatal event. Even in recent aplastic anemia transplant series, chronic GVHD was frequent among long-term survivors (Table 2).

GVHD is mediated by donor lymphocytes but much of its biology remains unclear.36 Perhaps because of thymic involution, GVHD increases in frequency and severity in recipients more than 20 years of age. The incidence of acute GVHD increases with age: in a Seattle series, from 6% to 26% in children S I O years old to 50% to 80% in adults 241 years old; in International Bone Marrow Transplant Registry patients from 34% 2 4% in children less than I O years old to 50% t 7% in adults 2 4 0 years old." Acute GVHD is itself a powerful risk factor for chronic GVHD, but independently age more than 20 years is also a predictor of chronic GVHD..'* In a recent University of Minnesota analysis, chronic GVHD was three times more frequent in transplant recipients more than 18 years old than in younger patients3' Among Seattle aplastic anemia patients, the rate of chronic GVHD showed a progressive increase with age (19% for patients 0 to 10 years old, 46% at 11 to 30 years of age, and 90% in patients more than 31 years old).40 The risk of chronic GVHD in children (< 18 years old) with aplastic anemia is now probably about

The increased risks of GVHD with age contribute to the poorer survival posttransplant in older patients (Table 2). For transplants reported to the International Bone Marrow

Transplant Registry between 1986 and 1992, the total 5-year survival was 66%, but for patients more than 25 years of age, it was less than 60%, whereas for children less than 16 years old survival was about 75%."* Bone marrow trans- plantation for aplastic anemia in patients more than 45 years of age has been only occasionally successfu1.4' Cyclosporine prophylaxis appears to have reduced the incidence of both acute and chronic GVHD in single-center studie~'~.".~~ as well as in registry ~ o m p a r i s o n s . ' ~ ~ ~ ' ~ ~ ~ Reduction in the mor- tality from chronic GVHD by aggressive immunosuppres- sion" may ultimately blunt the influence of age on GVHD.

In summary, excellent survival and low morbidity in younger patients make allogeneic bone marrow transplanta- tion the treatment of choice for children and adolescents. Patients older than the age of about 40 years have a high risk of transplant-related morbidity and mortality. Young adults in the intermediate age group have a reasonable oppor- tunity for cure with a bone marrow transplant but also face more complications than children. In addition to age, a pro- longed interval between diagnosis and transplant, multiple transfusions, and serious infections before transplant are risk factors.

IMMUNOSUPPRESSION

Antilymphocyte globulins (ALG). The mechanism of ac- tion of antilymphocyte sera and cyclosporine is almost cer- tainly immunosuppressive. ALG is cytolytic of T cells and

Table 2. Age and Survival After Bone Marrow Transplantation

Survival (%)

iutchinson, 1984'15 130 Minnesota, 1987"' 58 JCLA, 19901'2 29 EMTR, 1993"' 737 tGEMT, 1994" 540

K-M, 2 yr K-M, 6-8 yr K-M, 5 yr K-M, 5 yr K-M

Age in Years

InstitutionlGroup, Year N Method 0 5 10 15 20 25 30 35 40 45 50

l I I

I 1 1 1 E 1

Abbreviation: K-M, Kaplan-Meier estimate.

85

Qr, I

I 68 I I I CO

use only.For personal at PENN STATE UNIVERSITY on February 23, 2013. bloodjournal.hematologylibrary.orgFrom

Page 5: The treatment of severe acquired aplastic anemia · 2015-07-29 · tic anemia, most particularly the decision between allogeneic transplantation and immunosuppression and the consider-

3370 YOUNG AND BARRETT

both antilymphocyte globulins and cyclosporine inhibit T- cell function, especially the production of suppressive lym- phokines. ALG administration rapidly reduces circulating lymphocytes, usually to less than 10% of starting values, and when total lymphocytes return to pretreatment values months later, activated lymphocyte numbers in recovered patients remain de~reased .~~.~’ Antilymphocyte preparations in vitro stimulate T-cell proliferation and promote secretion of some growth f a ~ t o r s ~ ~ - ” However, these actions are prob- ably peripheral to the therapeutic effect, because growth fac- tors are abundant in untreated aplastic anemia. Improve- ments in response rates have largely been caused by intensification of immunosuppression with cyclosporine rather than the addition of growth factors.

In contrast to bone marrow transplantation, in which en- graftment results in the establishment of normal (donor) he- matopoiesis, successful immunosuppressive therapy only oc- casionally leads to rapid normalization of blood counts. More commonly, levels of hemoglobin, neutrophils, or platelets improve gradually over months or even years to levels suffi- cient to prevent spontaneous bleeding, bacterial and fungal infection, and the need for red blood cell transfusion^.^^ Pos- sible reasons for slow or partial recoveries include continued inhibition of marrow by lymphocytes or irreversible stem cell loss. The assessment of clinical recovery is not simple and complicates the interpretation of clinical trials. Transfu- sion independence may be a subjective measure of response, because hematologists vary in their criteria for administra- tion of blood products and individual patients tolerate anemia and thrombocytopenia differently. Blood counts are affected by continued transfusion. Even modest improvement in one or two lineages can be clinically significant, eg, even margin- ally higher neutrophil numbers can profoundly alter the prob- ability of survival, whether or not transfusion independence is also achieved. For these reasons, hematologic recovery or response rates have been derived from absolute increases in blood counts, transfusion-independent status, or failure to satisfy the criteria for severe disease.

Despite lack of agreement on these criteria, a review of results with immunosuppression in the early 1980s suggested that about 45% of patients with severe disease responded to ALG,53 including randomized controlleds4 and large multicenter trials” of ATG in the United States. European preparations against thoracic duct lymphocytes (ALG; from Merieux [Lyon, France] and the Swiss Serum Institute [Berne, Switzerland]) appeared to have equivalent efficacy to ATG (Upjohn, Kalamazoo, MI) available in the United States. In vitro, there are no major differences between ATG and ALG or among individual lots in cytolytic activity or in T-cell antigens re~ognized.~~”’ Differences in treatment regimens also do not appear to appreciably affect response rates. Some early protocols used 10 to 28 daily intravenous infusions, but a short course (40 mgkglday for 4 days) is equally effective and less toxic, especially for induction of serum ~ickness.5~ Although clearly active in aplastic anemia, ATG and ALG have generally not been very useful in chil- dren or in patients who are severely neutropenic60-62 (very severe cases, with absolute neutrophil counts <200/pL, have the poorest prognosis63).

Cyclosporine. Cyclosporine had been reported anecdot- ally to produce responses in some patients with aplastic ane- mia.@ In more systematic studies, cyclosporine produced about a 50% response rate in patients who were refractory to ATG or ALG Most hematologic improvement occurred within a few months of starting therapy and was usually sustained after cyclosporine was discontinued; how- ever, recovery was sometimes dependent on continued cyclosporine administration. A French multicenter trial at- tempted a direct comparison of cyclosporine to ALG as ini- tial treatment for aplastic anemia and found no difference in outcome, but the response rates to either agent were poor, probably because of the low doses of cyclosporine and ALG

Although in some studies lower doses have been effective:’ best results have been seen with high doses of cyclosporine (12 mgkg/d for adults and 15 mg/kg/d for children). These regimens require frequent monitoring of blood cyclosporine levels and dose adjustment for creatinine levels. Serious complications from cyclosporine include hy- pertension, seizures (perhaps related to hypomagnesemia), and Pneumocystis carinii infection. Responses have been sufficiently consistent and impressive that cyclosporine has been enthusiastically advocated over antilymphocyte sera by some European in~estigators.6~

Intensive immunosuppression. Clinical trials that com- bined cyclosporine and ALG followed from the observation of clinical efficacy in aplastic anemia of each agent alone. Joining their different mechanisms of action, lymphocytoxi- city provided by ATG or ALG and functional block of lym- phocyte activation and activity by cyclosporine, would pro- duce more intensive immunosuppression. Recent trials have indicated that intensive immunosuppression is superior to ALG or ATG alone in severe aplastic anemia (Table 3). In a large German multicenter protocol, patients were random- ized between ALG and ALG plus cyclosporine in full doses.70 Added cyclosporine greatly increased the hemato- logic response rates (65% v 39% at 3 months and 70% v 46% at 6 months in all patients). The combined regimen more than doubled the response rate for very severe disease (65% v 3 1 % for ALG alone), and actuarial survival at 41 months also was higher (80% v 44%). Responses occurred sooner and were more complete and relapses were less fre- quent with combined therapy. Survival for all patients in the two groups was not different, perhaps because of the routine addition of cyclosporine to nonresponders in the control (ALG only) arm at 3 months. Similar results were found when ALG was compared with ALG plus cyclosporine in 42 severe and moderate Korean aplastic anemia patients (70% v 43% response rates at 6 month^).^' In a study at the National Institutes of Health Clinical Center of untreated severe aplas- tic anemia, cyclosporine plus ATG produced hematologic remissions in almost 80% of patients, more than twice the remission rate for matched historical control^.'^ Severely neutropenic patients had hematologic response and survival rates indistinguishable from patients with granulocyte num- bers greater than 2001pL. Children, who have been poor responders to ATG or ALG alone, actually had a higher response rate to combined therapy than did adults (-30 years of age, 86% at 1 year; 220 years of age, 72%). Children

use only.For personal at PENN STATE UNIVERSITY on February 23, 2013. bloodjournal.hematologylibrary.orgFrom

Page 6: The treatment of severe acquired aplastic anemia · 2015-07-29 · tic anemia, most particularly the decision between allogeneic transplantation and immunosuppression and the consider-

TREATMENT OF SEVERE APLASTIC ANEMIA 337 1

Table 3. intensive Immunosu~~rerrion in Severe blastic Anemia

Median Median

(range; Cases Age ANCIpU

Study, Year Regimen N vrl <O.Z/!.kL Response Survivel Relapse Median F/U (dl

German multicenter, 1992’’ ALG + CSA 43 32 (7-80) 0.48/19% 70% at 6 mo 64% at 41 mo 11% 516 (183-1.295)

EGBMT, 1995’‘ ALG + CSA + 40 16 (2-72) 0.19/50% 82% at 1 yr 92% at 34 mo 3% 428 (122-1,005) G-CSF

NIH, 199572 ATG + CSA 51 28 (3-79) 0.34/42% 78% at 1 yr 86% at 1 yr. 18% at 1 yr. 912 (218-1,714) 72% at 2 yr 36% at 2 yr

Abbreviations: ALG, antilymphocyte globulin; ATG, antithymocyte globulin; CSA, cyclosporine; G-CSF, granulocyte colony-stimulating factor; EGMBT, European Group for Bone Marrow Transplantation; NIH. National Institutes of Health.

have done well in other, small pilot studies of intensive immunos~ppression.~~ In a recent prospective European co- operative trial, intensive immunosuppression was combined with granulocyte colony-stimulating factor (G-CSF) ther- a ~ y . ~ ~ Almost all patients showed hematologic responses, and the actuarial probability of transfusion independence at 18 months was 97%. Actuarial survival at almost 3 years was 92% (there were 3 early deaths and 4 treatment failures among the 40 patients treated), and even among very severe cases the rate was 86%.74 Interpretation of the contribution of G-CSF is confounded by the inclusion of 20 patients who were retreated with ALG.

Corticosteroids. Methylprednisolone in modest doses, usually 1 mgkgld, is administered during ALG and ATG therapy to ameliorate serum sickness. Low-dose corticoste- roids are not effective treatment in aplastic anemia, and their role in stabilizing vascular integrity and preventing bleeding is quite unproven. Extremely high doses of methylpredniso- lone (20 to 50 mgkgld) can induce hematologic responses in aplastic anemia7’ but are not preferable to either ALG or cyclosporine. The purported benefit of combining ALG and extremely high doses of corticosteroid^^^ was not confirmed in a controlled trial.77 Corticosteroids at even modest doses have significant toxicities; the development of aseptic necro- sis of femur or humerus are particularly unpleasant compli- cations in the thrombocytopenic aplastic anemia ~atient.~’

Relapse and evolution of clonal disease. For all immu- nosuppressive therapy in 358 European patients, the risk of relapse at 14 years was 35%.7q The same proportion of NIH patients treated with ATG and cyclosporine also relap~ed.~’ Relapse may be less frequent when patients are treated promptly after diagnosis.79 The majority of relapsed patients respond again to immunosuppression, and their prognosis appears similar to those cases who do not relapse.

Evolution of aplastic anemia to another hematologic dis- ease can occur years after a successful response to immuno- suppression.80 Laboratory evidence of paroxysmal nocturnal hemoglobinuria (PNH) is most common, ie, 13% in a large retrospective European survey but higher (57% in Basel’l) or lower (<g% in Seattle77) in smaller series. The signifi- cance of these figures is now uncertain, because a high pro- portion of aplastic anemia patients at presentation have flow cytometric evidence of defective expression of glycophos- phoinositol-linked pr~teins’’-~~ (the Ham test is insensitive in aplastic anemia, not only because patients are so commonly

transfused with red blood cells but because granulocytes may manifest the defect more f req~ent ly~~) . Posttreatment PNH is often subclinical and rarely associated with severe hemoly- sis or thrombotic events. More serious complications are myelodysplasia and leukemia. In a European study, the 10- year cumulative incidence rate was 9.6% for myelodysplasia (without leukemia) and 6.6% for acute le~kemia.’~ Myelo- dysplasia may possibly be milder in long-term survivors of aplastic anemia than as a primary diagnosis, but some pa- tients progress rapidly to le~kemia.~’*’~ In addition to clonal bone marrow disorders, aplastic anemia patients postimmu- nosuppression and after bone marrow transplantation (espe- cially with radiation in the conditioning regimen) have an increased rate of occurrence of malignant tumors, ie, 2% to 3% after a decade.”

ROLE OF HEMATOPOIETIC GROWTH FACTORS

As described above, neither deficient stromal cell function nor growth factor production is central to the etiology of aplastic anemia. Administered growth factors probably in- crease blood counts by stimulating the existing committed progenitor cell pool. G-CSF and granulocyte-macrophage colony-stimulating factor (GM-CSF) usually affect neutro- phil numbers only, and improvement is generally propor- tional to the patient’s initial neutrophil c o ~ n t . ~ ~ . ’ ~ Granulo- cyte numbers often decline despite continued treatment, and higher cell counts, if sustained, are dependent on continued factor administration. However, a Japanese multicenter trial of chronic G-CSF therapy showed marked increases in neu- trophil count in almost all patients, and 10 of 27 patients also showed improvement in anemia, with 3 having higher platelet counts as well.g9 Interleukin-3, alone or in combina- tion with GM-CSF, has had even less impact on myelo- poiesis than G-CSF or GM-CSF and little on platelet produc- t i~n . ’~ .~’ Bilineage and trilineage responses to G-CSF and GM-CSF therapy cited anecdotally may represent delayed improvement after immunosuppression or spontaneous re- mission.

Despite the lack of objective support for their use, growth factors are now frequently offered to patients with severe bone marrow failure, before definitive therapy by marrow transplant or immunosuppression, a practice appropriately deplored as wasteful and dangerous by a group of European expert^.^' Nevertheless, G-CSF and GM-CSF can shorten the period of neutropenia postmarrow tran~piant?~ and they

use only.For personal at PENN STATE UNIVERSITY on February 23, 2013. bloodjournal.hematologylibrary.orgFrom

Page 7: The treatment of severe acquired aplastic anemia · 2015-07-29 · tic anemia, most particularly the decision between allogeneic transplantation and immunosuppression and the consider-

3372 YOUNG AND BARRElT

may be beneficial when used as an adjunct in an immunosup- pressive regimen.74

ANDROGENS

In the 1960s androgens seemed a treatment advance for aplastic anemia, but the early studies grouped moderate and severe as well as acquired and congenital aplastic anemia. Androgens alone as first treatment generally have not been shown to improve survival in severe aplastic anemia,3 if still considered beneficial by some investigator^.".^^ An Ameri- can controlled trial of androgens as an adjunct to ATG showed no benefit:6 and a European randomized protocol showed only higher hematologic response rates but without a survival advantage in a subset of severely affected women.97 Improvement and even androgen dependence can be seen in an occasional patient who has failed immunosuppression, and in this setting they may be useful and are unlikely to be harmful.

MARROW TRANSPLANTATION FROM UNRELATED DONORS

Most aplastic anemia patients lack an HLA-matched sib- ling donor. Large donor banks can now provide lists of po- tential donors for patients with more common haplotypes but are far less successful for uncommon HLA types and members of ethnic minorities?' Even donors phenotypically matched on all three paired HLA A, B, and DR antigens are likely to have both major and minor histocompatability antigen disparities with the recipient. Serologic testing misses important heterogeneity at the HLA A and B loci, now apparent with molecular typing methods99; identifica- tion of major and minor antigen disparity by functional test- ing is still under development.

Mismatching for a single antigen has a pronounced effect on survival because of graft rejection.lW An intensified con- ditioning regimen, which includes radiation or an alkylator drug, is required to prevent graft rejection. T-cell depletion of the donor inoculum, effective in preventing GVHD from a matched but unrelated donor, so increases the risk of graft failure as to be precluded in aplastic anemia. A further nega- tive effect is the considerable logistic delay. An average of 4 months is still required to locate a donor, confirm HLA matching, and arrange the harvest, by which time the risks for marrow transplant have increased due to infection and allosensitization.

As a consequence of the higher rates of rejection, GVHD, and infection and the longer interval from diagnosis to proce- dure, transplantation from an unrelated matched donor has not produced the same excellent outcome as sibling trans- plant (or transplant from a phenotypically matched family donor; Table 4). Twenty-nine aplastic anemia patients were among 459 unrelated transplants reported from the National Marrow Donor Program; their survival at 2 years was 29%, and 4 of 10 still alive continued to require transfusions.'" In the International Marrow Unrelated Search and Transplant Study (IMUST), underway in several European hospitals, a cohort of unrelated donor transplants has been compared with matched concurrent identical sibling transplants per-

Table 4. Unrelated Donor Marrow Transplantation in ADhtiC Anemia

Study N

Literature review (to 1979)"' 3 1owa118 4 Hammersmith11g~'20 11 EGBMT12' 24 M i l ~ a u k e e ' ~ ' ~ ~ ~ ~ 16 IMUST'23 15

Alive GVHD (cases) (cases)

Early Death or Graft Failure (cases)

0 1 1 2* 1 1

-

- 10 6 9 4

3 3 5 - 1 4

Abbreviations: GVHD, chronic severe; EGBMT, European Group for Bone Marrow Transplantation; IMUST, International Marrow Unre- lated Search and Transplant Study.

* Autologous reconstitution in 1 case.

formed at the same centers, all treated for severe aplastic anemia."* The mean age of recipients of unrelated marrow was 16 years; 36% of the patients were alive at 1 year, with most deaths occurring from the high rate of graft failure (52%), despite intensive conditioning. By comparison, the standard transplant recipients, who were older, showed a 65% l-year survival rate and less than a 10% rate of graft failure.'03 The best results using unrelated donors have been in young children," probably because they tolerate the highly toxic chemotherapy used for conditioning and also are less prone to serious GVHD.

The generally poor results of unrelated donor transplanta- tion have been blamed on the deleterious effects of the lengthy search process. As earlier performance of unrelated transplants becomes possible, comparison of unrelated and matched transplants as initial therapy becomes feasible; this high-risk procedure will then also compete more directly with immunosuppressive treatments. A search for a donor should be undertaken early and children especially should be considered as candidates. At present, unrelated transplants would best be performed in experienced units testing their own research protocols or contributing data to a registry.

BONE MARROW TRANSPLANT OR IMMUNOSUPPRESSION?

Immunosuppression as first therapy for patients who lack an HLA-identical sibling and marrow transplantation for children with appropriate donors are uncontroversial (Table 5). A dilemma may exist in selecting the best initial therapy for young to middle-aged adult patients who have compatible siblings. Despite the applicability of both immunosuppres- sion and allogeneic transplant to a small group of patients, views on the merits and disadvantages of each treatment tend to be polarized. At one extreme, transplantation has been advocated for all patients with sibling donors to the age of 55; at the other extreme, immunosuppression has been recommended as first treatment because of the equivalent survival rates.

Marrow transplantation cures aplastic anemia but with a risk of morbidity and mortality secondary to treatment. Immunosuppression induces hematologic improvement- partial correction of blood counts is the rule-but with little

use only.For personal at PENN STATE UNIVERSITY on February 23, 2013. bloodjournal.hematologylibrary.orgFrom

Page 8: The treatment of severe acquired aplastic anemia · 2015-07-29 · tic anemia, most particularly the decision between allogeneic transplantation and immunosuppression and the consider-

TREATMENT OF SEVERE APLASTIC ANEMIA 3373

Table 5. Treatment Options in Severe Aplastic Anemia

Recommended first therapies Allogeneic bone marrow transplantation

Sibling donor Phenotypically matched family member

ATG Cyclosporine Combined therapy

Adjunctive/second therapies

Immunosuppression

Hematopoietic growth factors G-CSF

Androgens Unrelated bone marrow transplantation

Hematopoietic growth factors Stem cell factor Combinations of factors Thrombopoietin

Experimentalfiuture therapies

Monoclonal antibodies to T cell

toxicity and almost no treatment-related mortality. However, successfully treated patients after either therapy may suffer late complications: after immunosuppression, relapse and death due to pancytopenia and transformation to clonal and sometimes malignant hematologic disease; after marrow transplantation, late graft failure, lethal GVHD, and second malignancies. At least initially, marrow transplantation is more expensive than immunosuppression.

In retrospective analyses, immunosuppression and bone marrow transplantation show equivalent beneficial effects on survival in severe aplastic anemia. Survival curves at 6 years of the European Group for Bone Marrow Transplantation are superimpo~able.~~ The most recent analysis of these data showed a statistically significant survival advantage for transplantation over immunosuppression only among se- verely neutropenic children (<20 years old), and since 1991 immunosuppression has been equivalent or superior to trans- plantation for all ages, even among very severe cases (A. Bacigalupo, unpublished data). Series from large single cen- ters may favor immunosuppression (Basel"') or transplant (Seattle"), but the differences have not reached statistical significance. The impact of late myelodysplasia and leuke- mia remains uncertain; a plateau in actuarial survival after immunosuppression has been discerned in someM." but not all 0the8~*" studies. Current long-term survival curves may not reflect relatively recent improvements, notably cyclo- sporine to intensify immunosuppression and better prepara- tive regimens and cyclosporine in tran~plantation.'~~

The excellent results of marrow transplantation in children with HLA-identical siblings and the low mortality and mor- bidity make transplantation the treatment of choice in the young patient. Transplantation is sufficiently hazardous in patients older than 40 years of age that immunosuppression is generally favored. For other patients, a clear recommenda- tion for one therapy is difficult, perhaps impossible, in the absence of a consensus among experts-or of a prospective comparison of immunosuppression and transplantation, never performed and unlikely to be undertaken. Sometimes

a decision will be. based on the patient's insurance status or personality. On medical grounds, immediate transplant should be favored for the younger, relatively untransfused and uncomplicated case. For higher risk or less willing pa- tients, transplant could be reserved to rescue immunosup- pression failures.'O'

FUTURE THERAPIES

Aplastic anemia patients will benefit from general ad- vances in the prevention and treatment of GVHD and the general trend to improved survival after allografting. For nontransplant therapy, the optimal immunosuppressive regi- men, especially the desirability of sequential versus com- bined immunosuppression and the utility of added growth factors should be studied in clinical trials. Prolonged treat- ment with cyclosporine may be needed to prevent relapses. Other forms of immunosuppression, such as cyclophospha- mide, might be useful in patients refractory to ATG and cyclosporine. Monoclonal antibodies to T cells'0g and acti- vated T cells"0 have produced remissions in occasional pa- tients; specific therapy directed to the pathophysiology of aplastic anemia could include conjugated monoclonal anti- bodies to T-cell subsets and their lymphokines and competi- tion with soluble receptor molecules (Table 5) .

The remarkable achievements in our understanding and treatment of aplastic anemia are the results of clinical re- search. Further progress depends on the performance of planned trials and access to patient tissue samples. For these reasons, as well as for the experienced care available at tertiary referral hospitals, patients with aplastic anemia should continue to be referred to centers that specialize in bone marrow failure disorders.

ACKNOWLEDGMENT

Many thanks to Dr Cynthia Dunbar of the Hematology Branch and Dr Michelle Fox of the Transfusion Medicine Department for their careful reading of the manuscript in draft and to Dr Andrea Bacigalupo of Ospedale San Martino for providing recent EGBMT data before publication.

REFERENCES 1. Thomas ED, Storb R, Fefer A, Slichter SJ, Bryant JI, Buckner

CD, Neiman PE, Clift RA, Funk DD, Lerner KE: Aplastic anaemia treated by marrow transplantation. Lancet 1:284, 1972

2. Bortin MM, Gale R P , Rim AA: Allogeneic bone marrow transplantation for 144 patients with severe aplastic anemia. JAMA 2451132, 1981

3. Camitta BM, Thomas D, Nathan DG, Gale RP, Kopecky U, Rappeport JM, Santos G, Gordon-Smith EC, Storb R: A prospective study of androgens and bone marrow transplantation for treatment of severe aplastic anemia. Blood 53:504, 1979

4. Young NS, Alter B P Aplastic Anemia, Acquired and Inherited. Philadelphia, PA, Saunders, 1994

5. Annitage JO: Bone marrow transplantation. N Engl J Med 3302327, 1994

6. Deeg HJ, Klingemann H-G, Phillips CL: A Guide to Bone Marrow Transplantation. Berlin, Germany, Springer-Verlag, 1992

7. Treleaven JG, Barrett AJ (eds): Bone Marrow Transplantation in Practise. London, UK, Churchill Livingston, 1992

8. Gordon-Smith EC (ed): Aplastic Anaemia. London, UK, Bail- litre Tindall, 1989

use only.For personal at PENN STATE UNIVERSITY on February 23, 2013. bloodjournal.hematologylibrary.orgFrom

Page 9: The treatment of severe acquired aplastic anemia · 2015-07-29 · tic anemia, most particularly the decision between allogeneic transplantation and immunosuppression and the consider-

3374 YOUNG AND BARRETT

9. Schrezenmeier H, Gerok M, Heimpel H, Raghavachar A: As- sessment of frequency of hematopoietic stem cells in aplastic anemia by limiting dilution type long term marrow culture. Exp Hematol 202306, 1992 (abstr)

10. Maciejewski JP, Anderson S , Katevas P, Young NS: Pheno- typic and functional analysis of the bone marrow progenitor cell compartment in aplastic anemia. Br J Haematol 87:227, 1994

11. Kaufman DW, Kelly JP, Levy M, Shapiro S : The Drug Etiol- ogy of Agranulocytosis and Aplastic Anemia. New York, NY, Ox- ford, 1991

12. Marsh JCW, Chang J, Testa NG, Hows JM, Dexter TM: In vitro assessment of marrow ‘stem cell’ and stromal cell function in aplastic anaemia. Br J Haematol 78:258, 1991

13. Holmberg LA, Seidel K, Leisenring W, Torok-Storb B: Aplastic anemia: Analysis of stromal cell function in long-term mar- row cultures. Blood 84:3685, 1994

14. Maciejewski JP, Hibbs JR, Anderson S , Katevas P, Young NS: Bone marrow and peripheral blood lymphocyte phenotype in patients with bone marrow failure. Exp Hematol 22:1102, 1994

15. Selleri C, Anderson S , Young NS, Maciejewski JP: Inter- feron-? and tumor necrosis factor-a suppress early and late stages of hematopoiesis in vitro and induce programmed cell death. Blood 84:215a, 1994 (abstr, suppl 1)

16. Maciejewski J, Selleri C, Young NS: Fas antigen expression on CD34’ human marrow cells is induced by interferon-gamma and tumor necrosis factor-alpha and potentiates hematopoietic suppres- sion in vitro. Blood 85:3183, 1995

17. Nakao S , Yamaguchi M, Shiobara S : Interferon gamma gene expression in unstimulated bone marrow mononuclear cells predicts a response to cyclosporine therapy in aplastic anemia. Blood 79:2532, 1992

18. Nisticb A, Young NS: ?-Interferon gene expression in the bone marrow of patients with acquired aplastic anemia. Ann Intern Med 120:463, 1994

19. Nimer SD, Ireland P, Meshkinpour A, Frane M: An increased HLA DR2 frequency is seen in aplastic anemia patients. Blood 84:923, 1994

20. Nakao S , Takamatsu H, Shiobara S , Schrezenmeier H, Frickhofen N, Matsuda T, Kaneshige T, Mizoguchi H: Susceptibility to relapse of aplastic anemia after successful immunosuppressive therapy is closely associated with an HLA class I1 haplotype. Blood 84:215a, 1994 (abstr, suppl 1)

21. Storb R: Bone marrow transplantation for aplastic anemia. Cell Transplant 2:365, 1994

22. Bacigalupo A: Severe Aplastic Anaemia Working Party, in EBMT Working Parties Reports. Harrogate, UK, European Group for Bone Marrow Transplantation, 1994, p 49

23. Storb R, Weiden PL, Sullivan KM, Appelbaum FR, Beatty P, Buckner CD, Clift RA, Doney KC, Hansen J, Martin PJ, Sanders JE, Stewart P, Witherspoon RP, Thomas ED: Second marrow trans- plants in patients with aplastic anemia rejecting the first graft: Use of a conditioning regimen including cyclophosphamide and antithy- mocyte globulin. Blood 70:116, 1987

24. McCann SR, Bacigalupo A, Gluckman E, Hinterberger W, Hows J, Ljungman P, Marin P, Nissen C, van’t Veer Kerthof E, Raghavachar A, Socie G, Frickhofen N, Locasciulli A, Schrezen- meier H: Graft rejection and second bone marrow transplants for aplastic anemia: a report from the Aplastic Anaemia Working Party of the European Bone Marrow Transplant Group. Bone Marrow Transplant 13:233, 1994

25. k g HJ, Self S, Storb R, Doney K, Appelbaum F R , With- erspoon RP, Sullivan KM, Sheehan K, Sanders J, Michelson E, Thomas ED: Decreased incidence of marrow graft rejection in pa- tients with severe aplastic anemia: Changing impact of risk factors. Blood 68:1363, 1986

26. Storb R, Thomas ED, Buckner CD, Clift RA, Deeg HJ, Fefer A, Goodell BW, Sale GE, Sanders JE, Singer J, Stewart P, Weiden PL: Marrow transplantation in thirty “untransfused” patients with severe aplastic anemia. Ann Intern Med 92:30, 1980

27. Champlin RE, Horowitz MM, vanBekkum DW, Camitta BM, Elfenbein GE, Gale RP, Gluckman E, Good RA, Rimm AA, Rozman C, Speck B, Bortin MM: Graft failure following bone marrow trans- plantation for severe aplastic anemia: Risk factors and treatment results. Blood 73:606, 1989

28. Bordin JO, Heddle NM, Blajchman MA: Biologic effects of leuckocytes present in transfused cellular blood products. Blood 83:1703, 1994

29. SociC G, Henry-Amar M, Bacigalupo A, Hows J, Tichelli A, Ljungman P, McCann SR. Frickhofen N, Van’t Veer-Korthof EV, Gluckman E: Malignant tumors occurring after treatment of aplastic anemia. N Engl J Med 329:1152, 1993

30. Deeg HJ, Soci6 G , Schoch G, Henry-Amar M, Witherspoon RP, Gluckman E, Storb R: New malignancies in patients with aplatic anemia treated by marrow transplantation: Analysis of the Seattle and Paris results. Blood 84:249a, 1994 (abstr, suppl 1)

31. Smith BR, Guinan EC, Parkman R, Ferrera JM, Levey RH, Nathan DG, Rappeport JM: Efficacy of a cyclophosphamide-procar- bazine-antithymocyte serum regimen for prevention of graft rejection following bone marrow transplantation for transfused patients with aplastic anemia. Transplantation 39:67 1, 1984

32. Hows J, Palmer S , Gordon-Smith EC: Cyclosporine and graft failure following bone marrow transplantation for severe aplastic anemia. Br J Haematol 60:611, 1985

33. May WS, Sensenbrenner LL, Burns WH, Ambinder R, Carroll MP, Griffin CA, Jones RJ, Miller CB, Mellitis ED, Vogelsang GB, Wagner JE, Wingard JR, Yeager AM, Santos GW: BMT for severe aplastic anemia using cyclosporine. Bone Marrow Transplant 11:459, 1993

34. Storb R, Thomas ED, Weiden PL, Buckner CD, Clift RA, Fefer A, Fernando LP, Giblett ER, Goodell BW, Johnson FL, Lerner KG, Neiman PE, Sanders JE: Aplastic anemia treated by allogeneic bone marrow transplantation: A report on 49 new cases from Seattle. Blood 48:817, 1976

35. Storb R, Etzioni R, Anasetti C, Appelbaum FR, Buckner CD, Bensinger W, Bryant E, Clift R, Deeg HJ, Doney K, Flowers M, Hansen J, Martin P, Pepe M, Sale G , Sanders J, Singer J, Sullivan KM, Thomas ED, Witherspoon RP: Cyclophosphamide combined with antithymocyte globulin in preparation for allogeneic marrow transplants in patients with aplastic anemia. Blood 84:941, 1994

36. Ferrara JLM, Deeg HJ: Graft-versus-host disease. N Engl J Med 324:667, 1991

37. Deeg W, Cottler-Fox M: Clinical spectrum and pathophysiol- ogy of acute graft-versus-host disease, in Burakoff SJ, Deeg HJ, Ferrara J, Atkinson H (eds): Graft-vs.-Host Disease. Immunology, Pathophysiology, and Treatment. New York, NY, Marcel-Dekker, 1990, p 31 1

38. Atkinson K, Horowitz MM, Gale RP, van Bekkum DW, Gluckman E, Good RA, Jacobsen N, Kolb H, Rimm AA, Ringden 0, Ciril R, Sobocinski KA, Zwaan FE, Bortin MM: Risk factors for chronic graft-versus-host disease after HLA-identical sibling bone marrow transplantation. Blood 75:2459, 1990

39. Ochs LA, Miller WJ, Filipovich AH, Haake RJ, McGIave PB, Blazar BR, Ramsay NKC, Kersey JH, Weisdorf DJ: Predictive factors for chronic graft-versus-host disease after histocompatible sibling donor bone marrow transplantation. Bone Marrow Transplant 13:455, 1994

40. Storb R, Prentice R L , Sullivan KM, Shulman HM, Deeg HJ. Doney KC, Buckner CD, Clift RA, Witherspoon RP, Appelbaum FA, Sanders JE, Stewart PS, Thomas ED: Predictive factors in chronic graft-versus-host disease in patients with aplastic anemia

use only.For personal at PENN STATE UNIVERSITY on February 23, 2013. bloodjournal.hematologylibrary.orgFrom

Page 10: The treatment of severe acquired aplastic anemia · 2015-07-29 · tic anemia, most particularly the decision between allogeneic transplantation and immunosuppression and the consider-

TREATMENT OF SEVERE APlASTlC ANEMIA 3375

treated by marrow transplantation from HLA-identical siblings. AM Intern Med 98:461, 1983

41. Sanders JE, Storb R, Anasetti C, Deeg HJ, Doney K, Sullivan KM, Witherspoon RP, Hansen J: Marrow transplant experience for children with severe aplatic anemia. Am J Pediatr Hematol Oncol 16:43, 1994

42. Sobocinski KA, Horowitz MM, Rowlings PA, Zhang M-J, Nugent ML, Passweg JR, Armitage JO, Gale RP, Bortin MM: Bone marrow transplantation- 1994: A report from the International Bone Marrow Transplant Registry and the North American Autologous Bone Marrow Transplant Registry. J Hematother 3:95, 1994

43. Klingemann H-G, Storb R, Fefer A, Deeg HJ, Appelbaum FR, Buckner CD, Cheever MA, Greenberg PD, Stewart PS, Sullivan KM, Witherspoon RP, Thomas ED: Bone marrow transplantation in patients aged 45 years and older. Blood 67:770, 1986

44. Storb R, Deeg HJ, Pepe M, Doney K, Appelbaum F, Beatty P, Bensinger W, Buckner CD, Clift R, Hansen J, Hill R, Longton G, Anasetti C, Martin P, Loughran TP, Sanders J, Singer J, Stewart P, Sullivan KM, Witherspoon R, Thomas ED: Graft-versus-host disease prevention by methotrexate combined with cyclosporin compared to methotrexate alone in patients given marrow grafts for severe aplas- tic anaemia: Long-term follow-up of a controlled trial. Br J Haematol 72:567, 1989

45. Hows JM, Marsh JC, Yin JL, Durrant S, Swirsky D, Worsley A, Fairhead SM, Chipping PM, Palmer S, Gordon-Smith EC: Bone marrow transplantation for severe aplastic anaemia using cyclosporin: Long-term follow-up. Bone Marrow Transplant 4: 11, 1989

46. Gluckman E, Horowitz MM, Champlin RE, Hows JM, Baci- galupo A, Biggs JC, Camitta BM, Gale RP, Gordon-Smith EC, Marmont AM, Masaoka T, Ramsay NKC, Rimm AA, Rozman C, Sobocinski KA, Speck B, Bortin MM: Bone marrow transplantation for severe aplastic anemia: Influence of conditioning and graft-ver- sus-host disease prophylaxis regimens on outcome. Blood 79:269, 1992

47. Platanias L, Gascon P, Bielory L, Griffith P, Nienhuis AW, Young N: Lymphocyte subsets and lymphokines following anti- thymocyte globulin therapy in patients with aplastic anaemia. Br J Haematol 66:433, 1987

48. L6pez-Karpovitch X, Zarzosa ME, Cirdenas MR, Piedras J: Changes in peripheral blood mononuclear cell subpopulations during antithymocyte globulin therapy for severe aplastic anemia. Acta Haematol 81:176, 1989

49. Gascon P, Zoumbos N, Djeu J, Scala G, Young N: Lympho- kine abnormalities in aplastic anemia. Implications for the mecha- nism of action of ATG. Blood 65:407, 1985

50. Kawano Y, Nissen C, Gratwohl A, Speck B: Immunostimula- tory effects of different antilymphocyte globulin preparations: A possible clue to their clinical effect. Br J Haematol 68:115, 1988

51. Nimer SD, Golde DW, Kwan K, Lee K, Clark S, Champlin R: In vitro production of granulocyte-macrophage colony stimulating factor in aplastic anemia: Possible mechanisms of action of antithy- mocyte globulin. Blood 78:163, 1991

52. de Planque MM, Bacigalupo A, Wiirsch A, Hows JM, Dever- gie A, Frickhofen N, Brand A, Nissen C: Long-term follow-up of severe aplastic anaemia patients treated with antithymocyte globulin. Br J Haematol 73:121, 1989

53. Young N, Speck B: Antithymocyte and antilymphocyte glob- ulins: Clinical trials and mechanism of action, in Young NS, Levine AS, Humphries RK (eds): Aplastic Anemia. Stem Cell Biology and Advances in Treatment. New York, NY, Liss, 1984, p 221

54. Champlin R, Ho W, Gale R P Antithymocyte globulin treat- ment in patients with aplastic anemia. N Engl J Med 308:113, 1983

55. Young N, Griffith P, Brittain E, Elfenbein G, Gardner F, Huang A, Harmon D, Hewlett J, Fay J, Mangan K, Morrison F,

Sensenbrenner L, Shadduck R, Wang W, Zaroulis C, Zuckerman K: A multicenter trial of anti-thymocyte globulin in aplastic anemia and related diseases. Blood 72:1861, 1988

56. Raefsky E, Gascon P, Gratwohl A, Speck B, Young NS: Biological and biochemical characterization of anti-thymocyte glob- ulins (ATG) and anti-lymphocyte globulins (ALG). Blood 68:712, 1986

57. Smith AG, O’Reilly RJ, Hansen JA, Martin PJ: Specific anti- body-blocking activities in antilymphocyte globulin as correlates of efficacy for the treatment of aplastic anemia. Blood 66:721, 1985

58. Bonnefoy-BCrard N, Vincent C, Revillard J-P: Antibodies against functional leukocyte surface molecules in polyclonal anti- lymphocyte and antithymocyte globulins. Transplantation 5 1 :669, 1991

59. Bielory L, Gascon P, Lawley T, Nienhuis AW, Young NS, Frank M: Human serum sickness: A prospective clinical analysis of 35 patients treated with equine anti-thymocyte globulin for bone marrow failure. Medicine 67:40, 1988

60. Bayever E, Champlin R, Ho W, Lenarsky C, Storch S, La- disch S, Gale RP, Feig SA: Comparison between bone marrow trans- plantation and antithymocyte globulin in treatment of young patients with severe aplastic anemia. J Pediatr 105:920, 1984

61. Locasciulli A, van’t Veer L, Bacigalup A, Hows J, van Lint MT, Gluckman E, Nissen C, McCann S, Vossen J, Schrezenmeier A, Hinterberger W, Marin A: Treatment with marrow transplantation or immunosuppression of childhood acquired severe aplastic anemia: A report from the EBMT SAA working party. Bone Marrow Trans- plant 6:211, 1990

62. Halperin DS, Grisaru D, Freedman MH, Saunders EF: Severe acquired aplastic anemia in children: 1 I-Year experience with bone marrow transplantation and immunosuppressive therapy. Am J Pedi- atr Hematol Oncol 11:304, 1989

63. Bacigalupo A, Hows J, Gluckman E, Nissen C, Marsh J, van Lint MT, Congiu M, DePlanque MM, Emst P, McCann S, Raga- vashar A, Frickhofen N. Wursch A, Marmont AM, Gordon-Smith EC: Bone manow transplantation (BMT) versus immunosuppression for the treatment of severe aplastic anaemia (SAA): A report of the EBMT SAA working party. Br J Haematol 70:177, 1988

64. Shahidi NT, Wang WC, Shurin SB, Finlay JL, Sondel PM, Dinndorf PA: Treatment of acquired aplastic anemia with cyclosporine and androgens, in Shahidi NT (ed): Aplastic Anemia and Other Bone Marrow Failure Syndromes. London, UK, Springer- Verlag, 1990, p 155

65. Leonard EM, Raefsky E, Griffith P, Kimball J, Nienhuis AW, Young NS: Cyclosporine therapy of aplastic anaemia, congenital and acquired red cell aplasia. Br J Haematol 72:278, 1989

66. Hinterberger-Fisher M, Hocker P, Lechner K, Seewann H, Hinterberger W: Oral cyclosporin-A is effective treatment for un- treated and also for previously immunosuppressed patients with se- vere bone marrow failure. Eur J Hematol 43:136, 1989

67. Gluckman E, Esperou-Bourdeau H, Baruchel A, Boogaerts M, Briere J, Donadio D, Leverger G, Leponier M, Reiffers J, Janvier M, Michallet M, Stryckmans P: Multicenter randomized study com- paring cyclosprine-A alone and antithymocyte globulin with pred- nisone for treatment of severe aplastic anemia. Blood 79:2540, 1992

68. Leeksma OC, Thomas LLM, Van der Leslie J, Van Oers MHJ, von dem Borne AEGKr, Goudsmit R: Effectiveness of low dose cyclosporine in acquired aplastic anaemia with severe neutro- penia. Neth J Med 41:143, 1992

69. Schrezenmeier H, Schlander M, Raghavachar A: Cyclosporin A in aplastic anemia-Report of a workshop. Ann Hematol 65:33, 1992

70. Frickhofen N, Kaltwasser JP, Schrezenmeier H, Raghavachar A, Vogt HG, Henmann F, Freund M, Meusers P, Salama A, Heimpel H: Treatment of aplastic anemia with antithymocyte globulin and

use only.For personal at PENN STATE UNIVERSITY on February 23, 2013. bloodjournal.hematologylibrary.orgFrom

Page 11: The treatment of severe acquired aplastic anemia · 2015-07-29 · tic anemia, most particularly the decision between allogeneic transplantation and immunosuppression and the consider-

3376 YOUNG AND BARRETT

methylprednisolone with or without cyclosporine. N Engl J Med 324:1297, 1991

71. Park CW, Han CH, Kim CC, Kim DJ: Immunomodulation therapy for severe aplastic anemia-ALG versus ALG plus cyclosporin A. Korean J Intern Med 4:28, 1989

72. Rosenfeld S, Kimball J, Vining D, Young NS: Intensive im- munosuppression with antithymocyte globulin and cyclosporin as treatment for severe aplastic anemia. Blood 85:3058, 1995

73. Matloub YH, Bostrom B, Golembe B, Priest J, Ramsay NKC: Antithymocyte globulin, cyclosporine, and prednisone for the treat- ment of severe aplastic anemia in children. Am J Pediatr Hematol Oncol 16:104, 1994

74. Bacigalupo A, Broccia G, Corda G, Arcese W, Carotenuto M, Gallamini A, Locatelli F, Mori PG, Sarocco P, Todeschini A, Coser P, Iacopino P, van Lint MT, Gluckman E: Antilymphocyte globulin, cyclosporin and granulocyte colony stimulating factor in patients with acquired severe aplastic anemia (SAA): A pilot study of the EBMT SAA Working Party. Blood 85:1348, 1995

75. Bacigalupo A, van Lint MT, Ceni R, Giordano D, Santini G, Carella M, Damasio E, Rossi E, Risso M, Vimercati R, Podesta’ M, Durando A, Reali G, Avanzi G , Barbanti M, Marmont AM: Treat- ment of severe aplastic anemia with bolus 6-methylprednisolone and antilymphocytic globulin. Blut 41:168, 1980

76. Speck B, Gratwhol A, Nissen C, Osterwalder B, Wursch A, Tichelli A, Lori A, Reusser P, Jeannet M, Signer E: Treatment of severe aplastic anemia. Exp Hematol 14:126, 1986

77. Doney K, Pepe M, Storb R, Bryant E, Anasetti C, Appelbaum FR. Buckner CD, Sanders 3, Singer J, Sullivan K, Weiden P, Hansen JA: Immunosuppressive therapy of aplastic anemia: Results of a prospective, randomized trial of antithymocyte globulin (ATG), methylprednisolone, and oxymetholone to ATG, very high-dose methylprednisolone, and oxymetholone. Blood 79:2566, 1992

78. Marsh JCW, Zomas A, Hows JM, Chapple M, Gordon-Smith EC: Avascular necrosis after treatment of aplastic anaemia with antilymphocyte globulin and high-dose methylprednisolone. Br J Haematol 84:73 1, 1993

79. Schrezenmeier H, Marin P, Raghavachar A, McCann S, Hows J, Gluckman E, Nissen C, Van? Veer-Korthof ET, Ljungman P, Hinterberger W, van Lint MT, Frickhofen N, Bacigalupo A: Relapse of aplastic anaemia after immunosuppressive treatment: A report from the European Bone Marrow Transplantation Group SAA Work- ing Party. Br J Haematol 85:371, 1993

80. Young NS: The problem of clonality in aplastic anemia. Dr. Dameshek‘s riddle, restated. Blood 79:1385, 1992

81. Tichelli A, Gratwohl A, Wursch A, Nissen C, Speck B: Late haematological complications in severe aplastic anaemia. Br J Haematol 69:413, 1988

82. Schubert 3, Alvarado M, Uciechowski P, Zielinska-Skowro- nek M, Freund M, Vogt H, Schmidt RE: Diagnosis of paroxysmal nocturnal haemoglobinuria using immunophenotyping of peripheral blood cells. Br J Haematol 79:487, 1991

83. Shichishima T, Terasawa T, Saitoh Y, Hashimoto C, Ohto H, Maruyama Y: Diagnosis of paroxysmal nocturnal haemoglo- binuria by phenotypic analysis of erythrocytes using two-colour flow cytometry with monoclonal antibodies to DAF and CD59MACIF. Br J Haematol 85:378, 1993

84. Schrenzenmeier H, Hertenstein B, Wagner B, Raghavachar A, Heimpel H: A pathogenetic link between aplastic anemia and paroxysmal nocturnal hemoglobinuria is suggested by a high fre- quency of aplastic anemia with a deficiency of phosphatidylinositol glycan proteins. Exp Hematol 23:81, 1995

85. Narayanan MN, Geary CC, Freemont AJ, Kendra JR: Long- term follow-up of aplastic anaemia. Br J Haematol 86:837, 1994

86. Champlin RE, Nimer SD, Ireland P, Oette DH, Golde DW: Treatment of refractory aplastic anemia with recombinant human

granulocyte-macrophage-colony-stimulating factor. Blood 73:694, 1989

87. Guinan EC, Sieff CA, Oette DH, Nathan DG: A phase U I1 trial of recombinant granulocyte-macrophage colony-stimulating factor for children with aplastic anemia. Blood 76:1077, 1990

88. Kojima S , Fukuda M, Miyajima Y, Matsuyama T, Horibe K: Treatment of aplastic anemia in children with recombinant human granulocyte colony-stimulating factor. Blood 77:937, 1991

89. Sonoda Y, Ohno Y, Fujii H, Takahashi T, Nakayama S , Haru- yama H, Nasu K, Shimazaki C , Hara H, Kanamaru A, Kakishita E, Horiuchi A, Abe T, Masaoka T, Uchino H, Nagai K: Multilineage response in aplastic anemia patients following long-term administra- tion of filgrastim (recombinant human granulocyte colony stimulat- ing factor). Stem Cells 11:543, 1993

90. Ganser A, Lindemann A, Seipelt G, Ottmann OG, Eder M, Falk S , Herrmann F, Kaltwasser JP, Meusers P, Klausmann M, Frisch J, Schulz G, Mertelsman R, Hoelzer D: Effects of recombinant human interleukin-3 in aplastic anemia. Blood 76:1287, 1990

91. Nimer SD, Paquette R L , Ireland P, Resta D, Young D, Golde DW: A phase MI study of interleukin-3 in patients with aplastic anemia and myelodysplasia. Exp Hematol 222375, 1994

92. Marsh JCW, Socie G, Schrezenmeier H, Tichelli A, Gluckman E, Ljungman P, McCann SR, Raghavachar A, Marin P, Hows JM, Bacigalupo A: Haemopoietic growth factors in aplastic anaemia: A cautionary note. Lancet 344:172, 1994

93. Nemunaitis J, Anasetti C, Storb R, Bianco JA, Buckner CD, Onetto N, Martin P, Sanders J, Sullivan K, Mori M, Shannon-Dorcy K, Bowden R, Appelbaum FR, Hansen J, Singer JW: Phase 11 trial of recombinant human granulocyte-macrophage colony-stimulating factor in patients undergoing allogeneic bone marrow transplantation from unrelated donors. Blood 79:2572, 1992

94. Najean Y, Haguenauer 0: Long-term (5 to 20 years) evolution of nongrafted aplastic anemia. Blood 76:2222, 1990

95. Gardner FH, Juneja HS: Androstane therapy to treat aplastic anaemia in adults: An uncontrolled pilot study. Br J Haematol 65:295, 1987

96. Champlin RE, Ho WG, Feig SA, Winston DJ, Lenarsky C, Gale RP: Do androgens enhance the response to antithymocyte glob- ulin in patients with aplastic anemia? A prospective randomized trial. Blood 66:184, 1985

97. Bacigalupo A, Chaple M, Hows J, van Lint MT, McCann S, Milligan D, Chessels J, Goldstone AH, Ottolander J, Van’t Veer ETh, Den Ostendorf GJ, Comotti V, Coser P, Broccia G, Bosi A, Locasciulli A, Catalan0 L, Battista R, Arcese W, Carotenuto M, Marmont AM, Gordon Smith EC: Treatment of aplastic anemia (AA) with antilymphocyte globulin (ALG) and methylprednisolone (MPred) with or without androgens: A randomized trial from the EBMT SAA working party. Br J Haematol 83:145, 1993

98. Beatty PG: The use of unrelated donors bone marrow trans- plantation. Marrow Transplant Rev 1 : 1, 199 1

99. Santamaria P, Reinsmoen NL, Lindstrom AL, Boyce-Jacino MT, Barbosa JJ, Faras A J , McGlave PB, Rich SS: Frequent HLA class I and DP sequence mismatches in serologically (HLA-A, HLA- B, HLA-DR) and molecularly (HLA-DRB1, HLA-DQA1, HLA- DQBl) HLA-identical unrelated bone marrow transplant pairs. Blood 83:280, 1994

100. Gordon-Smith EC, Hows J, Bacigalupo A, Gluckman E, van Lint MT, Congiu A, James DCO, Barrett AJ, de Planque MM, Siimes MA, Toivanen A, Ringden 0, Marmont AM: Bone marrow transplantation for severe aplastic anaemia (SAA) from donors other than HLA identical siblings: A report of the EBMT working party. Semin Hematol 21:100, 1994

101. Keman NA, Bartsch G, Ash RC, Beatty PG, Champlin R, Filipovich A, Gajewski J, Hansen JA, Henslee-Downey J, McCul- lough J, McGlave P, Perkins HA, Phillips GL, Sanders J, Stroncek

use only.For personal at PENN STATE UNIVERSITY on February 23, 2013. bloodjournal.hematologylibrary.orgFrom

Page 12: The treatment of severe acquired aplastic anemia · 2015-07-29 · tic anemia, most particularly the decision between allogeneic transplantation and immunosuppression and the consider-

TREATMENT OF SEVERE APLASTIC ANEMIA 3371

D, Thomas ED, Blume K G Analysis of 462 transplantations from unrelated donors facilitated by the national marrow donor program. N Engl J Med 328:593, 1993

102. Bradley BA, Hows JM, Gore SM, Bidwell JL, Clay T, Dow- nie TR, Gluckman E, Howard M R , Laundy GJ: Current status of unrelated-donor bone marrow transplatation. Clin Transplant 91, 1992

103. Hows JM, Downie TR, Bradley BA, Gore SM: Unrelated donor BM" for acquired SAA: European experience 1989-93. Bone Marrow Transplant 143, 1994 (abstr, suppl)

104. Camitta B, Casper J, Pietryga D, Garbrecht F, Ash R, Keever C, Murray K, Baxter-Lowe L, Flomenberg N: Unrelated donor mar- row transplantation (BMT) for children with severe aplastic anemia. Blood 82:344a, 1993 (abstr, suppl 1)

105. Speck B, Tichelli A, Gratwohl A, Nissen C: Treatment of severe aplastic anemia: A 12-year follow-up of patients after bone marrow transplantation or after therapy with antilymphocyte globu- lin, in Shahidi NT (ed): Aplastic Anemia and Other Bone Marrow Failure Syndromes. London, UK, Springer-Verlag. 1990, p 96

106. Tichelli A, Gratwohl A, Nissen C, Singer E, Stebler-Gysi C, Speck B: Morphology in patients with severe aplastic anemia treated with antilymphocyte globulin. Blood 80337, 1992

107. Paquette R L , Tebyani N, Frane M, Ireland P, H0 WG, Champlin RE, Nimer SD: Long-term outcome of aplastic anemia in adults treated with antithymocyte globulin: Comparison with bone marrow transplantation. Blood 85:283, 1995

108. Crump M, Larratt LM, Maki E, Curtis JE, Minden MD, Meharchand JM, Lipton JH: Treatment of adults with severe aplastic anemia: Primary therapy with antithymocyte globulin (ATG) and rescue of ATG failures with bone marrow transplantation. Am J Med 92:596, 1992

1 0 9 . Jansen J, Gratama J W , Zwaan FE, Simonis RFA: Therapy with monoclonal antibody OKT3 in severe aplastic anemia. Exp Hematol 12:46, 1984 (suppl 15)

110. Schwinger W, Urban C, Lackner H, Mache C: Treatment of aplastic anemia with a monoclonal antibody directed against the interleukin-2 receptor. Ann Hematol 66:181, 1993

1 11 . Gluckman E, Socit G, Devergie A, Bourdeau-Esperou H, Traineau R, Cosset JM: Bone marrow transplantation in 107 patients with severe aplastic anemia using cyclophosphamide and thoraco- abdominal irradiation for conditioning: Long-term follow-up. Blood 78:2451, 1991

112. Champlin RE, Ho WG, Nimer SD, Gajewski JG, Selch M, Bumison M, Holley G, Yam P, Petz L, Winston DJ, Feig SA: Bone

marrow transplantation for severe aplastic anemia. Transplantation 49:720, 1990

113. Bortin MM, Horowitz MM, Rowlings PA, Rimm AA, So- bocinski KA, B a n g MJ, Gale RP: 1993 Progress report from the International Bone Marrow Transplant Registry. Bone Marrow Transplant 12:97, 1993 (abstr)

114. Pepe MS, Longston G, Pettinger M, Mori M, Fisher LD, Storb R: Summarizing data on survival, relapse, and chronic graft- versus-host disease after bone marrow transplantation: Motivation for and description of new methods. Br J Haematol 83502, 1993

115. Storb R, Thomas ED, Buckner CD, Appelbaum FR, Clift RA, Deeg HJ, Doney K, Hansen JA, Prentice RL, Sanders JE, Stew- art P, Sullivan KM, Witherspoon RP: Marrow transplantation for aplastic anemia. Semin Hematol21:27, 1984

116. McGlave PB, Haake R, Miller W, Kim T, Kersey J, Ramsay NKC: Therapy of severe aplastic anemia in young adults and chil- dren with allogeneic bone marrow transplantation. Blood 70:1325, 1987

117. Dupont B, O'Reilly RI, Pollack MS, Good RA: Use of HLA genotypically different donors in bone marrow transplantation. Transplant Proc 11:219, 1979

118. Gingrich R D , Ginder GD, Goeken NE, Howe CWS, Wen BC, Hussey DH, Fyfe MA: Allogeneic marrow grafling with par- tially mismatched, unrelated marrow donors. Blood 71:1375, 1988

119. Hows, JM, Yin JL, Marsh J, Swirsky D, Jones L, Apperley JF, James DCO, Smithers S , Goldman JM, Gordon-Smith EC: Histo- compatible unrelated volunteer donors compared with HLA noniden- tical family donors in marrow transplantation for aplastic anemia and leukemia. Blood 68:1322, 1986

120. Hows J, McKinnon S , Brookes P, Kaminski E, Bidwell J, Bradley B, Batchelor J, Goldman J: Matched unrelated donor trans- plantation. Transplant Proc 21:2923, 1989

121. Bacigalupo A, Hows J, Gordon-Smith EC, Gluckman E, van Lint MT, Congiu M, James DCO, Barren AJ, Gmur J, de Planque MM, Siimes MA, Toivanen A, Ringden 0, Marmont AM: Bone marrow transplantation for severe aplastic anemia from donors other than HLA identical siblings: A report of the BMT working party. Bone Marrow Transplant 3:531, 1988

122. Camitta B, Ash R, Menitove J, Lawton C, Hunter J, Casper J: Bone marrow transplantation for children with severe aplastic anemia: Use of donors other than HLA-identical siblings. Blood 741852, 1989

123. Hows JM, Szydlo R, Anasetti C, Camitta B, Gajewski J, Gluckman E: Unrelated donor transplants for severe aplastic anemia. Bone Marrow Transplant 10:102, 1992 (suppl 1)

use only.For personal at PENN STATE UNIVERSITY on February 23, 2013. bloodjournal.hematologylibrary.orgFrom