NO 6 15, - Semantic ScholarBLOOD VOL 79, NO 6 The Jouml of The American Society of Hematology MARCH...

9
BLOOD VOL 79, NO 6 The Jouml of The American Society of Hematology MARCH 15, 1992 PERSPECTIVE The Problem of Clonality in Aplastic Anemia: Dr Dameshek’s Riddle, Restated By Neal S. Young WENTY-FIVE YEARS AGO, William Dameshek, T the founder of this journal and one of the most creative minds in American hematology, raised a provoca- tive question: what do aplastic anemia (AA), paroxysmal nocturnal hemoglobinuria (PNH), and acute leukemia have in common?’ His question was prompted by three observa- tions: (1) the frequency of development of PNH in his own patients with AA, (2) the overlap between the syndromes of aplasia and PNH; and (3) the similar high prevalence of both AA and PNH in the Orient. Dameshek speculated that PNH erythropoiesis was “ecologically advantageous,” and, furthermore, that both AA and PNH might represent different responses to marrow insults (of which he listed chemicals, ionizing radiation, and viruses). In the interven- ing decades since Dameshek’s editorial, the prospect of survival for patients with AA has been greatly improved; we have a much better understanding of the pathophysiology of AA at the cellular level, of PNH biochemically, and of the myelodysplasia syndromes genetically. Hematopoietic cell clonality can be measured in clinical specimens and after sophisticated manipulations in animal experiments. Provisional solutions to Dameshek’s riddle may be sug- gested, with some temerity, in light of these new data. CLONAL HEMATOPOIESIS IN APLASTIC ANEMIA Clonal hematopoiesis has been related to AA by three important clinical observations, listed here chronologically. PNHlaplasia syndrome. First, there are patients, whose bone marrow failure syndrome is difficult to classify, in whom there are features of classical aplasia-diminished marrow cellularity-and evidence of clonal hematopoiesis; these include especially the AA/PNH syndrome and also hypoplastic myelodysplasia. Lewis and Dacie described a positive Ham test in 7 of 46 of their AA patients, and 15 of 60 patients with PNH developed AA during their course.’ In a recently published Duke series of younger patients with PNH, the rate of AA was even higher, 58%.3The acid or sucrose hemolysis test can be positive on presentation or during the course of otherwise typical idiopathic AA and in patients with drug4or benzene-associateds AA. The finding of a positive Hams test in a pancytopenic patient or marrow acellularity in a patient with a positive Hams test can present considerable diagnostic confusion. PNH, like AA, is also relatively common in the Far East, where, as in younger American patients, it is more usually associated with AA and less so with thromboses.6 Second, long-term observa- tions by European investigators of patients seemingly cured of their aplasia by antilymphocyte globulin show that many appear to evolve to clonal hematopoiesis and clonal hema- tologic disease. In a series of 103 patients treated by immunosuppression in Basel, 13 developed PNH and 8 developed myelodysplasia or acute leukemia; the actuarial risk of PNH was estimated at 57% at 8 years.’ Of 223 long-term survivors after immunosuppression followed-up by the European Cooperative Group for Bone Marrow Transplantation, 19 developed PNH (13% risk at 7 years), 11 had myelodysplasia, and 5 of them later manifested acute myelogenous leukemia (combined risk, 15% at 7 years).8 Myelodysplasia after aplasia may have a particu- larly high risk of leukemic transformation? Third, as reported recently in these pages, some proportion of patients with a diagnosis of AA show evidence of clonal hematopoiesis on molecular analysis. One older woman of 7 aplastic cases in the recently published English study showed a monoclonal pattern with the M27p probe against whole blood and granulocytes, a proportion not significantly different from normal.” In contrast, in a Dutch study of aplastic anemia using three X-linked probes (hypoxanthine phosphoribosyl- transferase [HPRT], phosphoglycerate kinase [PGK], and M27P), 13 of 19 (72%) showed a monoclonal pattern, including 4 of 4 cases on presentation.” Patients with monoclonal patterns of hematopoiesis could respond to antithymocyte globulin (ATG) and one even recovered spontaneously! (The reason for the difference in results between the two studies is not apparent; most of the Dutch cases were informative for the M27P probe alone, which for technical reasons might lead to overestimation of monoclon- ality, as described below.) Late clonal disease in AA. Clonaliq on presentation. From the Cell Biology Section, Clinical Hematology Branch, Submitted December 30, 1991; accepted January 2, 1992. Address reprint requests to Neal S. Young, MD, ChieJ Cell Biology 0 1992 by The American Society of Hematology. National Heart, Lung, and Blood Institute, Bethesda, MD. Section, NHLBI, Bldg IO, Rm 7C103, NIH, Bethesda MD 20892. 0006-497119217906-0036$3.00/0 Mood, Vol79, No 6 (March 15), 1992: pp 1385-1392 1385 For personal use only. on August 30, 2017. by guest www.bloodjournal.org From

Transcript of NO 6 15, - Semantic ScholarBLOOD VOL 79, NO 6 The Jouml of The American Society of Hematology MARCH...

BLOOD VOL 79, NO 6

The Jouml of The American Society of Hematology

MARCH 15, 1992

PERSPECTIVE

The Problem of Clonality in Aplastic Anemia: Dr Dameshek’s Riddle, Restated

By Neal S. Young

WENTY-FIVE YEARS AGO, William Dameshek, T the founder of this journal and one of the most creative minds in American hematology, raised a provoca- tive question: what do aplastic anemia (AA), paroxysmal nocturnal hemoglobinuria (PNH), and acute leukemia have in common?’ His question was prompted by three observa- tions: (1) the frequency of development of PNH in his own patients with AA, (2) the overlap between the syndromes of aplasia and PNH; and (3) the similar high prevalence of both AA and PNH in the Orient. Dameshek speculated that PNH erythropoiesis was “ecologically advantageous,” and, furthermore, that both AA and PNH might represent different responses to marrow insults (of which he listed chemicals, ionizing radiation, and viruses). In the interven- ing decades since Dameshek’s editorial, the prospect of survival for patients with AA has been greatly improved; we have a much better understanding of the pathophysiology of AA at the cellular level, of PNH biochemically, and of the myelodysplasia syndromes genetically. Hematopoietic cell clonality can be measured in clinical specimens and after sophisticated manipulations in animal experiments. Provisional solutions to Dameshek’s riddle may be sug- gested, with some temerity, in light of these new data.

CLONAL HEMATOPOIESIS IN APLASTIC ANEMIA

Clonal hematopoiesis has been related to AA by three important clinical observations, listed here chronologically.

PNHlaplasia syndrome. First, there are patients, whose bone marrow failure syndrome is difficult to classify, in whom there are features of classical aplasia-diminished marrow cellularity-and evidence of clonal hematopoiesis; these include especially the AA/PNH syndrome and also hypoplastic myelodysplasia. Lewis and Dacie described a positive Ham test in 7 of 46 of their AA patients, and 15 of 60 patients with PNH developed AA during their course.’ In a recently published Duke series of younger patients with PNH, the rate of AA was even higher, 58%.3 The acid or sucrose hemolysis test can be positive on presentation or during the course of otherwise typical idiopathic AA and in patients with drug4 or benzene-associateds AA. The finding of a positive Hams test in a pancytopenic patient or marrow acellularity in a patient with a positive Hams test can present considerable diagnostic confusion. PNH, like AA, is also relatively common in the Far East, where, as in

younger American patients, it is more usually associated with AA and less so with thromboses.6

Second, long-term observa- tions by European investigators of patients seemingly cured of their aplasia by antilymphocyte globulin show that many appear to evolve to clonal hematopoiesis and clonal hema- tologic disease. In a series of 103 patients treated by immunosuppression in Basel, 13 developed PNH and 8 developed myelodysplasia or acute leukemia; the actuarial risk of PNH was estimated at 57% at 8 years.’ Of 223 long-term survivors after immunosuppression followed-up by the European Cooperative Group for Bone Marrow Transplantation, 19 developed PNH (13% risk at 7 years), 11 had myelodysplasia, and 5 of them later manifested acute myelogenous leukemia (combined risk, 15% at 7 years).8 Myelodysplasia after aplasia may have a particu- larly high risk of leukemic transformation?

Third, as reported recently in these pages, some proportion of patients with a diagnosis of AA show evidence of clonal hematopoiesis on molecular analysis. One older woman of 7 aplastic cases in the recently published English study showed a monoclonal pattern with the M27p probe against whole blood and granulocytes, a proportion not significantly different from normal.” In contrast, in a Dutch study of aplastic anemia using three X-linked probes (hypoxanthine phosphoribosyl- transferase [HPRT], phosphoglycerate kinase [PGK], and M27P), 13 of 19 (72%) showed a monoclonal pattern, including 4 of 4 cases on presentation.” Patients with monoclonal patterns of hematopoiesis could respond to antithymocyte globulin (ATG) and one even recovered spontaneously! (The reason for the difference in results between the two studies is not apparent; most of the Dutch cases were informative for the M27P probe alone, which for technical reasons might lead to overestimation of monoclon- ality, as described below.)

Late clonal disease in AA.

Clonaliq on presentation.

From the Cell Biology Section, Clinical Hematology Branch,

Submitted December 30, 1991; accepted January 2, 1992. Address reprint requests to Neal S. Young, MD, ChieJ Cell Biology

0 1992 by The American Society of Hematology.

National Heart, Lung, and Blood Institute, Bethesda, MD.

Section, NHLBI, Bldg IO, Rm 7C103, NIH, Bethesda MD 20892.

0006-4971 19217906-0036$3.00/0

Mood, Vol79, No 6 (March 15), 1992: pp 1385-1392 1385

For personal use only.on August 30, 2017. by guest www.bloodjournal.orgFrom

1386 NEAL S. YOUNG

Clonality for hematologists implies malignant or prema- lignant disease, as in the acute and chronic myelogenous leukemias, lymphoma, and myelodysplasia. The observa- tion of clonal hematopoiesis in classical AA has suggested to some observers that AA is also fundamentally a premalig- nant disorder12 and that immunosuppressive therapy only postpones its inevitable progre~sion.‘~ Indeed, before the dramatic improvement in survival after immunosuppres- sion, leukemia was an unusual complication of AA, esti- mated at less than 1%” and sufficiently rare to have warranted case reports of its occurrence after androgen therapy,14 marrow transplantation,” and immunosuppres- sionI6; less than 2% of patients with acute myelogenous leukemia have a history of aplastic anemia.17 However, the laboratory and clinical evidence that AA is an autoimmune disease are very strongI8; a majority of patients, perhaps more than 70%,’9 respond to aggressive treatment that suppresses the immune system. Even more striking is that patients with hypoplastic myelodysplasia can respond to immunosuppressive therapy (see below). How can clonality be interpreted in the context of immunologically mediated marrow disease?

LABORATORY DETERMINATION OF CLONALITY

Current clonality techniques rely on the natural mosa- icism of women for X-chromosome gene expression, due to the inactivation of most of one sex chromosome at an early stage of embryogenesis. Which X-chromosome is inacti- vated is randomly determined in the embryonic progenitor cell and then well preserved among its progeny that ultimately constitute an organ like the bone marrow. Fialkow first used polymorphism at the glucosed-phos- phate dehydrogenase (G6PD) allele to study blood cells from women heterozygous for enzyme types A and B, which are easily distinguished by electrophoresis.m For example, Oni et aP’ first demonstrated that PNH was a clonal disease in an African woman. Their red blood cells showed the expected mixture of A and B G6PD types, but complement lysis of the PNH erythrocytes released only G6PD type B.*’ Similarly, in a G6PD heterozygous patient with myelodyspla- sia, skin and T cells showed both isozyme types while erythrocytes, platelets, granulocytes, and B cells all con- tained type B only,22 showing clonality at the level of a stem cell for hematopoietic and B lymphocytes.”

An obvious limitation of G6PD expression analysis-the limited numbers of heterozygotes available-was overcome by the application of restriction enzyme digestion to X-chro- mosome DNA. Restriction enzymes cleave DNA into smaller fragments at specific nucleotide sequences; differ- ences in the length of fragments of DNA can occur because some base substitutions alter the recognition sequence for an enzyme. Polymorphisms have been identified in X-chro- mosome genes like HPRT and PGK for which specific probes are available. To exploit polymorphisms for clonal- ity studies, DNA is first digested with an enzyme to distinguish if the paternal and maternal X-chromosomes differ in fragment size of the gene. X-chromosome inactiva- tion is usually accompanied by methylation of cytosine residues, and if two fragment length polymorphisms are

present, the inactivated fragment can be distinguished by a second digestion with an enzyme sensitive to the presence of methyl groups such as Hpa II.23 In tissue of polyclonal origin, Southern analysis will show nearly equal quantities of both sizes of restriction fragments after this two-step digestion process, indicating derivation from a population of cells in which both X-chromosomes are active; if the tissue is monoclonal in origin, only a single restriction fragment length will be detected. Restriction fragment length polymorphism analysis can be applied to the major- ity of females, especially if combinations of probes are used: on HPRT anhlysis 29% of females are heterozygous; for PGK, 33%; and for two sequences called the variable copy number tandem repeat, characterized by multiallelic varia- tion and detected using a probe called M27P, the heterozy- gosity rate is greater than 80%. Restriction fragment length polymorphism analysis of myelodysplasia has demonstrated monoclonality in virtually every case.”,zs

Despite its inherent logic and simplicity, this method is prone to certain errors. Monoclonality may be missed because of contamination with uninvolved cells or tissue. Not all base substitutions are neutral in their effect on the cell, and cells expressing some alleles may face a selective disadvantage, resulting in pseudoclonality, as in HPRT deficiency, G6PD deficiency, and Wiskott-Aldrich syn- drome. The pattern of methylation may be aberrant and not strictly dependent on X-chromosome inactivation, as oc- curs in certain tumors or with certain probes (like the pXUT23-2.1 marke? or the M27P probe,” which are not capable of distinguishing all active from inactive X-chromo- somes). Aberrant results in the experimental tissue may become apparent on comparison with uninvolved tissue, like skin, but often the controls for putative clonally derived tissue is the same type of tissue obtained from normal donors.

Clonality of hematopoietic tissue is detected in some proportion of normal persons, perhaps a significant propor- tion: 6 of 42 (14%) by G6PD analysism; 3 of 81 (4%)= to 15 of 65 (23%)27 by HPRT or PGK fragment polymorphisms; 3 of 18 (17%) by M27P analysis.28 Are the cells of these individuals truly monoclonal in a pathologic sense, or do they represent random chance selection of a limited num- ber of clones, or is selection operating on X-chromosome expression? The results stress the importance of comparing assays on suspect monoclonal tissue in disease with analysis of the same tissue in normal populations and of uninvolved tissue in the same subject.

CLONALITY AS CONSEQUENCE: THE LIMITED STEM CELL POOL

Regulation of the limited number of primitive stem cells is required to avoid depletion of the compartment over the life span of the animal. Kay sug- gested in 1965 that variation over time in the entry of primitive stem cells into mitosis could effect an ordered release of mature differentiated cells, a process termed clonal succe~sion.2~ The clonal succession model predicted that a very small percentage of primitive stem cells would be in mitosis at any one time, and that individual stem cells,

Clonal succession.

For personal use only.on August 30, 2017. by guest www.bloodjournal.orgFrom

CLONALITY IN APLASTIC ANEMIA I 387

stem cell pool4

‘ sdudion

CTL - A

A

selection I

CTL /‘ -

b CTL - 1 PNH

Fig 1. Some possible mechanisms of marrow failure resulting in clonal hematopoiesis. The stem cell compartment is represented as a triangle. Generational age is indicated by the narrow arrow and efflux to the committed progenitor pool by the broad arrow. The triangular model of the stem cell pool emphasizes the hierarchical nature of the compartment.w The most primitive cells, shown at the apex, give rise by mitosis to the most mature stem cells, seen at the base; clones are denoted by colors. More mature cells have a greater probability of leaving the compartment to terminally differentiate because they are more numerous (a stochastic process) and because they are altered as a result of repeated cell division (a determinative process). As described in the text, clonality might be the result of a shrunken stem cell compartment with loss of a large number of stem cell clones or aborted maturation of primitive stem cells within the compartment. With immune attack on the marrow, stem cells normally lacking a ligand for cytotoxic lymphocytes (0) might be selected, resulting in PNH or a positive Hams test in AA. Finally, and most speculatively, under some circumstances cytotoxic lymphocyte (CTL) attack may induce genetic damage in the target cell (€3). manifested as altered cell proliferation associated with chromosomal deletions, and the clinical diagnosis of myelodysplasia (MDS); a similar process presumably occurs after irradiation or alkylating drugs, with early stem cell death (aplasia) followed by late unveiling of accumulated mutational damage (dysplasia, leukemia).

once triggered to mitosis, will produce progeny and become extinct. Clonal succession has been demonstrated under special experimental circumstances, as after introduction of marrow cells marked in vitro with integrated retroviral s eq~ences~””~ and in short-term analysis after transplanta- tion into irradiated recipients of relatively small numbers of marrow cells using X-linked red cell enzyme markers in mice” and cats.34

However, studies performed under more physiologic conditions have failed to demonstrate clonal succession. After transplantation of congenic mice that differed in enzyme isotype” and in analysis of hemoglobin types in mice obtained from chimeric embryos,% the small statistical variances in the proportion of enzyme or hemoglobin types in serially sampled reticulocytes imply continuous activity of large numbers of pluripotent stem cells many months after infusion?’ Reconstitution of hematopoiesis after trans- plant in humans is almost always p~lyclonal.”’~ By infer-

ence from the distribution of G6PD phenotype in heterozy- gous female carriers of chronic granulomatous disease, the number of pluripotent stem cells maintaining hematopoie- sis in adults was estimated to be greater than 400.”

The influence of stem cell number on apparent clonality of hematopoiesis has been most dramatic after transplanta- tion of highly purified repopulating cells. When a small number of donor stem cells (with cell surface phenotype Thy-1’” Lin- Sca-1’) were injected into congenic recipient mice:” the majority of the pluripotent clones disappeared early, with only one-third showing evidence of long term self-renewal. While repopulation was oligoclonal with injec- tion of limiting numbers of stem cells, the proportion of clones actively contributing to hematopoiesis increased in proportion to the size of the donor inoculum. These results would suggest that hematopoiesis may be oligoclonal only when stem cell numbers are limiting, an important argu- ment that is directly applicable to the interpretation of

For personal use only.on August 30, 2017. by guest www.bloodjournal.orgFrom

1388 NEAL S. YOUNG

clonality in patients with AA. In steady state normal, unstressed hematopoiesis, a larger number of stem cell clones, probably several hundred in humans, would support blood cell production. Nonetheless, these cells still would represent only a small proportion of the total primitive stem cell compartment.

Another and related paradox is the difference in stem cell self- renewal inferred from observations of marrow physiology compared with experimental repopulation. Only a small proportion of stem cells needs to be active because a repopulating cell has an enormous proliferative capacity. All the experiments cited above agree that a single stem cell can support hematopoiesis in a mouse for several months. The hierarchical structure of the stem cell compartment can be demonstrated experimentally by the enrichment for primitive stem cells produced by treatment with a cycle- active drug like 5 - f l ~ o r o u r a c i 1 ~ ~ ~ ~ indicating that the cells of the youngest generational age have the highest proliferative and self-renewal capacity (by implication, as stem cells pass through mitoses they lose proliferative capacity and in- crease in probability of terminal differentiation).

Observations of intact hematopoiesis and theoretical calculations of the number and capacity of the stem cell compartment suggest that animals are more than ade- quately endowed with hematopoietic stem cells of very high proliferative and self-renewal capacity. Normal hematopoi- esis is maintained from relatively small numbers of repopu- lating cells for the life-span of the animal, and there is very little evidence for aging of hematopoiesis-indeed, elderly mice make quite as good bone marrow donors as young 0nes.4~'~ Normal stem cells serially transplanted into W / W animals can maintain normal blood counts through several life spans: and marrow cells from old donors repopulate at least as well as cells from young and even fetal donors.46 However, hematopoietic stress profoundly affects stem cell regulation. When spleen colonies are injected into second- ary and tertiary irradiated recipients, the number and size of the new spleen colonies that form are markedly re- d u ~ e d . 4 ' ~ ~ ~ In the shielded limb of an irradiated animal49 or after a single bone marrow transplantation, stem cell proliferative capacity declines more than over a lifetime of normal f u n c t i ~ n . ~ ~ . ~ ' The same phenomenon can be ob- served with parabiosis instead of physical removal of donor cells, and in W / W mice (which are only mildly anemic) as in the irradiated host, and independently of the size of the donor inoculum.52 Similar permanent loss of self-renewal capacity can be observed at the stressful initiation of a long-term bone marrow culture53 and after cell irradiation in vitro.54 A likely mechanism of loss of repopulating cells under hematopoietic stress may be their premature recruit- ment into mitotic cycle, as illustrated experimentally by the devastating effect on functional reconstituting ability of marrow cells exposed to two doses of 5-fluorouracil-the first treatment recruits a large proportion of primitive stem cells into cycle, which are made susceptible to a cycle-active agent.55 The discordance between stem cell function under physiologic conditions and after serial transplantation par-

Serial transplantation and loss of selfrenewal.

allels the difference in clonal recruitment when stem cell number is abundant or limited. Hematopoietic stress can apparently lead to qualitative dysregulation of stem cell function in addition to and perhaps disproportionate to quantitative stem cell number loss.

Clonality in most AA patients is probably due to a reduced and dysregulated stem cell pool (Fig 1). Under conditions of hematopoietic stress, progeny of individual or very few clones can be detected when the blood is sampled. A good control for studies of clonality in bone marrow failure diseases is patients with normal marrows who are receiving cytotoxic chemotherapy. Studies of lymphoma patients after chemotherapy have been reported in ab- stract: 11 of 19 showed clonal hematopoiesis when studied with the M27B probe,= results analogous to studies of G6PD expression in heterozygous cats after chemother- apy?6 (In another recent British study, a high rate of clonality was observed in both postchemotherapy cases [treated for hematologic disease] and normal women.27) Serial studies of marrow failure patients might be expected to show rapid clonal succession in many severely pancyto- penic patients and reversion to polyclonal hematopoiesis with hematologic recovery.

CLONALITY AS ESCAPE: PNH?

The concept of PNH as a clonal disorder, based on the observation of G6PD type in the affected erythrocytes of a single woman:' has since been confirmed by restriction enzyme analysis of DNA in other patients, 5 of 5 cases in the recent series from England tested with the M27P probe." The pathophysiologic basis of PNH is now under- stood as a defect in a class of membrane proteins that attach to the cell surface by a glycolipid anchor, a phosphati- dylinositol moiety that is introduced into the lipid portion of the membrane by the fatty acids of diacyl glycer01.S~ Failure to express glycolipid bound proteins explains the abnormal complement sensitivity of PNH because several complement-inactivating proteins are phosphoinositol linked to the erythrocyte surface membrane.

Hematopoietic progenitor number is severely decreased in patients with cytopenias and PNH, even when the marrow is ce l I~Iar .S~.~~ Yet, normal syngeneic cells eclipse defective cells in twin transplantation.@' What membrane defect could explain marrow progenitor hypoproliferation? None of the phosphoinositol-linked proteins identified so far are related to hematopoietic growth factors, their receptors, or general regulators of cell proliferation. How- ever, several molecules important in the immune system proteins are affected by the PNH defect: the type I11 Fc receptor: monocyte antigen CD14, of unknown function:' and lymphocyte function-associated antigen-3 (LFA-3), which is the ligand for the T-cell glycoprotein CD2.63 LFA-3 is of special interest because it mediates adhesion of cytotoxic lymphocytes to target cells.".65

Absence of a recognition site for cytotoxic lymphocytes would certainly provide a growth advantage in a hematopoi- etic system under immune system attack and allow emer- gence of an ordinarily handicapped type of cell (Fig 1).

For personal use only.on August 30, 2017. by guest www.bloodjournal.orgFrom

CLONALITY IN APLASTIC ANEMIA 1389

Somatic mutations of the phosphoinositol-linkage pathway are probably common and relatively benign, as a small percentage of PNH cells (about 1%) can be detected in some normal individuals. Studies of fractionated erythro- cytes@ and progenitor cell cultures6’ from patients with AA show that complement-sensitive cells are increased in a high proportion of cases of otherwise typical AA on presen- tation. Rotoli and Luzzatto hypothesized that marrow failure was primary to both aplasia and PNH, the degree of selection of PNH clones in bone marrow failure determin- ing the disease’ clinical appearance as pure aplasia (no clones emerge), the mixed syndrome (some clones with limited proliferative capacity emerge), or typical PNH (multiple clones or high proliferative capacity emerge).6s Aplastic patients with positive Hams tests can respond to immunosuppressive therapy like antithymocyte globulin and c y c l ~ s p o r i n . ~ ~ ~ ~ ~ ” The relative balance among T-cell attack, LFA-3 loss, and selection of PNH clones would determine the clinical character of a fundamentally immune- mediated disease. In addition, the absence of LFA-3 on the stem cell surface would also explain the susceptibility of these cells to leukemic transformation because of loss of immune surveillance!

CLONALITY AS ETIOLOGY: MYELODYSPLASIA?

Clonality has been implicated as the seminal event in AA.”.” But hypocellular myelodysplasia is even more prob- lematic. How can a clone with a growth disadvantage dominate the marrow?

Misdiagnosis? In contrast to myelodysplasia and myelog- enous leukemia, cytogenetic abnormalities are rare in acquired AA. In one large series, chromosomal abnormali- ties were found in only 4% of 183 cases, and those seen-deletions in chromosomes 5, monosomy 7, and trisomy &are so frequently observed in dysplasia as to suggest misdiagnosis of hypoplastic myelodysplasia.’1 Simi- larly, in one patient with pancytopenia and clonal hemato- poiesis, as determined by G6PD analysis, multiple physical anomalies, only moderate blood count depression, and preserved erythropoiesis in the bone marrow suggest a case not typical of AA.72

An inhibitory stem cell clone? Many molecules normally produced in the marrow, like transforming growth factor+ (TGF-f3),’3 inhibi~~,’~ macrophage inflammatory protein/ stem cell inhibit~r,’~ or macrophage colony-stimulating factor (M-CSF)? or present there in pathologic states, like y-interferon” and tumor necrosis factor-a,” can suppress hematopoiesis. These cytokines are assumed to play homeo- static regulatory roles or to suppress marrow in disease. Inhibitory activity for in vitro hematopoiesis has been reported in leukemian and myelodysplasia.” However, it is not only difficult to conceive of a stem cell with a growth disadvantage and inhibitory activity dominating bone mar- row, but there is no evidence that inhibitory molecules are specifically produced by hematopoietic cells or of hemato- poietic cells with inhibitory activity for their neighbors’ proliferation in hypocellular states.

A common insult for aplasia and myelodysplasia? Early

marrow failure and late clonal disease do occur in two well-defined syndromes, irradiation injury and Fanconi’s anemia, marked by the introduction of DNA damage in the first and inadequate repair of damaged DNA in the second. In both cases, pancytopenia due to AA is an early event and leukemia occurs much later; global stem cell destruction is the result of massive disruption of the dividing cell’s genetic integrity and abnormal proliferation of a single clone is the result of accumulated mutational events.

Stem cell destruction from radiation is an example of apoptosis. Apoptosis, or programmed cell death, is distin- guished from accidental cell death by specific histologic” and functional” features. In necrosis, cells and their cyto- plasmic organelles swell, release their contents, and elicit a local inflammatory response. In apoptosis, chromatin aggre- gation, cellular volume loss, and the formation of membrane- bound apoptotic bodies are characteristic, scattered cells are pyknotic, and there is no local inflammatory response. In apoptosis, cell death originates in the nucleus and can be detected in vitro by the release of ordered nucleotide fragments; DNA fragmentation precedes cell membrane lysis, the opposite sequence from cytotoxicity as a result of membrane attack by complement, antibodies, natural killer cells, or physical disruption. DNA cleavage in apoptosis is the result of endogenous endonuclease activation, and because these enzyme pathways may not be identical in all cells, cellular damage may vary in different tiss~es.8~ Apop- tosis can be triggered by a number of stimulants, including not only irradiations4 but also hematopoietic growth factor ~ithdrawal‘~”~ and cytotoxic lymphocyte attack.= Apoptosis has been hypothesized as a mechanism for the destruction of intracellular viral DNA and the prevention of its release when the infected cell is disr~pted.8’*~’

On the assumption that few biologic pheomena are absolute, some cells may survive cytotoxic lymphocyte attack and have resulting residual DNA injury (Fig 1). If true, two clinical predictions could be made. First, cytotoxic lymphocyte attack should induce dysplastic as well as aplastic marrow changes. In support of this hypothesis, variable degrees of cellular atypia are not uncommon in classical AA, some patients with myelodysplasia share immunologic abnormalities with AA and some appear to respond to immunosuppressive therapy like antithymocyte g l o b ~ l i n ’ ~ , ~ ~ and c y c l ~ s p o r i n e ~ ~ . ~ ~ (and our personal experi- ence), nor does dysplasia on presentation correlate with re- sponse to ATG therapy?5* Perhaps myelodysplasia is more likely than aplasia in the older patient with a blunted immune response. Second, late clonal abnormalities in AA, some myelodysplasia and acute leukemia, would be the result of the initial cytotoxic lymphocyte insult and induced chromosomal abnormalities. In our experience and in that of others, some patients whose relapsed pancytopenia is

*I have also observed characteristic but transient myelodysplasia in the bone marrow of a patient treated with an experimental cardiac drug (marketed as Arkin-Z in Japan) that ordinarily produces agranulocytosis at high rate (1% to 2%); the pathologic changes resolved with discontinuation of the offending medication.

For personal use only.on August 30, 2017. by guest www.bloodjournal.orgFrom

1390 NEAL S. YOUNG

associated with marrow cellularity respond well to a second course of immunosuppression. Immune attack, like radia- tion exposure, would thus have a dual effect on hematopoi- esis, inducing both acute marrow failure and long-term genetic injury.

IMPLICATIONS FOR THERAPY

The pessimistic tone of recent comment on AA, due to the development of late clonal disease in some patients, may be premature. Immune attack on hematopoietic stem cells may be chronic and may require more intensive initial therapy or longer duration of treatment. Immune system abnormalities of lymphocyte activation and lymphokine release often persist, despite seemingly effective immuno- suppressive therapy, just as progenitor number and marrow cellularity remain depressed despite adequate blood counts. Formal trials of immunosuppression of clonal diseases, like PNH and myelodysplasia, also seem warranted. Immunosup- pression is far less expensive and far less traumatic than bone marrow transplantation for AA. Even ATG, a rela- tively crude preparation, has produced survival results

equivalent to marrow replacement in most large series. When considered as an autoimmune disease, AA’s therapy compares well with that of other autoimmune diseases- rheumatoid arthritis, ulcerative colitis, iritis, type I diabetes mellitus-considering the vital importance of the organ system damaged. Newer immunosuppressives and the abil- ity to more specifically manipulate the immune response offer promise to improve immediate hematologic responses and perhaps to eliminate late clonal disease as well.

CONCLUSION

Dameshek concluded his 1967 editorial with a recogni- tion of the need for hard clinical categorization but an appeal for the value of a “vague” approach to thinking about pathogenesis. His final statement, “That a single ‘insult’ to the marrow may be responsible for bringing about different kinds of abnormalities . . . deserves consideration, not only from the conceptual standpoint but from the experimental approach as well,” may finally be realized in both the clinic and laboratory.

REFERENCES 1. Dameshek W Riddle: What do aplastic anemia, paroxysmal

nocturnal hemoglobinuria (PNH) and “hypoplastic” leukemia have in common? Blood 30:251,1967

2. Lewis SM, Dacie Jv: The aplastic anaemia-Paroxysmal nocturnal haemoglobinuria syndrome. Br J Haematol13:236, 1967

3. Ware RE, Halt SE, Rosse WF: Paroxysmal nocturnal hemo- globinuria with onset in childhood and adolescence. N Engl J Med 325:991,1991

4. Quagliana JM, Cartwright GE, Wintrobe MM: Paroxysmal nocturnal hemoglobinuria following drug-induced aplastic ane- mias. Ann Intern Med 61:1045,1964

5. Aksoy M: Different types of malignancies due to occupational exposure to benzene: A review of recent observations in Turkey. Environ Res 23:181,1980

6. Kruatrachue M, Wasi P, Na-Nakorn S: Paroxysmal nocturnal haemoglobinuria in Thailand with special reference to an associa- tion with aplastic anaemia. Br J Haematol39:267,1978

7. Tichelli A, Gratwohl A, Wursch A, Nissen C, Speck B: Late haematological complications in severe aplastic anaemia. Br J Haematol69:413,1988

8. de Planque MM, Bacigalupo A, Wiirsch A, Hows JM, Devergie A, Frickhofen N, Brand A, Nissen C: Long-term fol- low-up of severe aplastic anaemia patients treated with antithymo- cyte globulin. Br J Haematol73:121,1989

9. de Planque MM, Kluin-Nelemans HC, van Krieken HJM, Kluin PM, Brand A, Beverstock GC, Willemze R, van Rood JJ: Evolution of acquired severe aplastic anaemia to myelodysplasia and subsequent leukaemia in adults. Br J Haematol70:55,1988

10. Josten KM, Tooze JA, Borthwick-Clarke C, Gordon-Smith EC, Rutherford TR: Acquired aplastic anemia and paroxysmal nocturnal hemoglobinuria: Studies on clonality. Blood 78:3162, 1991

11. VanKamp H, Landegent JE, Jansen RPM, Willemze R, Fibbe WE: Clonal hematopoiesis in patients with acquired aplastic anemia. Blood 78:3209,1991

12. Marsh JC, Geary CG: Annotation-Is aplastic anaemia a pre-leukaemic disorder? Br J Haematol77:447,1991

13. Moore MA, Castro-Malaspina H: Immunosuppression in

aplastic anemia-postponing the inevitable? [editorial]. N En@ J Med 324:1358,1991

14. King JB, Burns D G Aplastic anaemia, oxymetholone and acute myeloid leukaemia. S Afr Med J 46:1622,1972

15. Hughes RT, Milligan DW, Smith GM, Leyland MJ, Gordon- Smith EC. A second bone marrow transplant for acute myeloid leukaemia after transplantation for aplastic anaemia [letter]. Br J Haematol68:391,1988

16. Tichelli A, Gratwohl A, Wiirsch A, Nissen C, Speck B: Secondary leukemia after severe aplastic anemia. Blut 56:79, 1988

17. Orlandi E, Alessandrino EP, Caldera D, Bemasconi C: Adult leukemia developing after aplastic anemia: Report of 8 cases. Acta Haematol79:174,1988

18. Frickhofen N, Liu JM, Young NS: Etiologic mechanisms of hematopoietic failure. Am J Ped Hematol Oncol12385,1990

19. Frickhofen N, Kaltwasser JP, Schrezenmeier H, Raghava- char A, Vogt HG, Henmann F, Freund M, Meusers P, Salama A, Heimpel H: Treatment of aplastic anemia with antithymocyte globulin and methylprednisolone with or without cyclosporine. N Engl J Med 324:1297,1991

20. Fialkow P: Primordial cell pool size and lineage relation- ships of five human cell types. Ann Hum Genet 37:39,1973

21. Oni SB, Osunkoya BO, Luzzatto L Paroxysmal noctural hemoglobinuria: Evidence for monoclonal origin of abnormal red cells. Blood 36:145, 1970

22. Raskind WH, Tirumali N, Jacobson R, Singer J, Fialkow PJ: Evidence for a multistep pathogenesis of a myelodysplastic syn- drome. Blood 63:1318,1984

23. Vogelstein B, Fearon ER, Hamilton SR, Preisinger AC, Willard HF, Michelson AM, Riggs AD, Orkin S H Clonal analysis using recombinant DNA probes from the X-chromosome 1. Can- cer Res 47:4806,1987

24. Janssen JWG, Buschle M, Layton M, Drexler HG, Lyons J, Van den Berghe H, Heimpel H, Kubanek B, Kleihauer E, Mufti GJ, Bartram CR: Clonal analysis of myelodysplastic syndromes: Evidence of multipotent stem cell origin. Blood 73:248,1989

25. Dunbar CE, Nienhuis AW: The myelodysplastic syndromes, in Handin R, Lux S, Stossel T (eds): Blood, Principles and Practice of Hematology. Philadelphia, PA, Lippincott (in press)

For personal use only.on August 30, 2017. by guest www.bloodjournal.orgFrom

CLONALITY IN APLASTIC ANEMIA 1391

26. Hodges E, Howell WM, Boyd Y, Smith J L Variable X-chro- mosome DNA methylation patterns detected with probe M27 beta in a series of lymphoid and myeloid malignancies. Br J Haematol 77315,1991

27. Gale RE, Wheadon H, Linch D C X-chromosome inactiva- tion patterns using HPRT and PGK polymorphisms in haematolog- ically normal and post-chemotherapy females. Br J Haematol 79:193,1991

28. Cachia PG, Culligan DJ, Whittaker JA, Padua RA, Jacobs A: Clonal haemopoiesis in patients in stable remission from lymphoma. Blood 78:327a, 1991 (abstr, suppl 1)

29. Kay HEM: How many cell generations? Lancet 2418,1965 30. Capel B, Hawley RG, Mintz B: Long- and short-lived murine

hematopoietic stem cell clones individually identified with retrovi- ral integration markers. Blood 752267,1990

31. Jordon CT, Lemischka I R Clonal and systemic analysis of long-term hematopoiesis in the mouse. Genes Dev 4220,1990

32. Snodgrass R, Keller G: Clonal fluctuation within the hae- matopoietic system of mice reconstituted with retrovirus-infected stem cells. EMBO J 6:3955,1987

33. Nakano,T; Waki N, Asai H, Kitamura Y Long-term mono- clonal reconstitution of erythropoiesis in genetically anemic W/W" mice by injection of 5-fluorouracil-treated bone marrow cells of Pgk-lb/Pgk-la mice. Blood 70:1758,1987

34. Abkowitz JL, Linenberger ML, Newton MA, Shelton GH, Ott RL, Guttorp P: Evidence for the maintenance of hematopoie- sis in a large animal by the sequential activation of stem-cell clones. Proc Natl Acad Sci USA 87:9062,1990

35. Harrison DE, Astle CM, Lemer C Number and continuous proliferative pattern of transplanted primitive immunohematopoi- etic stem cells. Pkoc Natl Acad Sci USA 85:822,1988

36. Harrison DE, Lerner C, Hoppe PC, Carlson GA, Alling D: Large numbers of primitive stem cells are active simultlneously in aggregated embryo chimeric mice. Blood 69:773,1987

37. Nash R, Storb R, Neiman P: Polyclonal reconstitution of human marrow after allogeneic bone marrow transplantation. Blood 72:2031,1988

38. Turhan AG, Humphries RK, Phillips GL, Eaves AC, Eaves CT: Clonal hematopoiesis demonstrated by X-linked DNA polymor- phisms after allogeneic bone marrow transplantation. N Engl J Med 3201655,1989

39. Buescher ES, Alling DW, Gallin JI: Use of an X-linked human neutrophil marker to estimate timing of lybnization and size of the dividing stem cell pool. J Clin Invest 76:1581,1985

40. Smith LG, Weissman IL, Heimfeld S: Clonal analysis of heaatopoietic stem-cell differentiation in vivo. Proc Natl Acad Sci USA 88:2788,1991

41. Rosendaal M, Hodgson GS, Bradley TR: Haemopoietic stem cells are organized for use on the basis of their generation- age. Nature 26468,1976

42. Lemer C, Harrison D E 5-Fluorouracil spares hemopoietic stem cells responsible for long-term repopulation. Exp Hematol 18:114,1990

43. Harrison D E Normal function of transplanted marrow cell lines from aged mice. J Gerontol30279,1975

44. Hellman S, Botnick LE, Hannon EC, Vigneulle RM: Prolif- erative capacity of murine hematopoietic stem cells. Proc Natl Acad Sci USA 75:490,1978

45. Harrison DE: Normal production of erythrocytes by mouse marrow continuous for 73 months. Proc Natl Acad Sci USA 703184,1973

46. Harrison DE, Astle CM, Lerner C Ultimate erythropoietic repopulating abilities of fetal, young adult, and old adult cells compared using repeated irradiation. J Exp Med 160759,1984

47. Ross EAM, Anderson N, Micklem HS: Serial depletion and regeneration of the murine hematopoietic system. Implications for hematopoietic organization and the study of cellular aging. J Exp Med 155:432,1982

48. Harrison DE, Stone M, Astle C M Effects of transplantation on the primitive immunohematopoietic stem cell. J Exp Med 172431,1990

49. Mauch P, Rosenblatt M, Hellman S: Permanent loss in stem cell self renewal capacity following stress to the marrow. Blood 72:1193, 1988

50. Harrison DE, Astle CM, Delaittre J A Loss of proliferative capacity in immunohemopoietic stem cells caused by serial trans- plantation rather than aging. J Exp Med 147:1526,1978

51. Mauch P, Hellman S: Loss of hematopoietic stem cell self-renewal after bone marrow transplantation. Blood 74:872, 1989

52. Harrison DE, Astle CM: Loss of stem cell repopulating ability upon transplantation. Effects of donor age, cell number, and transplantation procedure. J Exp Med 1561767,1982

53. MacMillan JR, Wolf NS: Depletion of reserve in the hemopoietic system. 11. Decline in CFU-S self-renewal capacity following prolonged cell cycling. Stem Cells 2:45, 1982

54. Reincke U, Hannon EC, Hellman S : Residual radiation injury exhibited in long-term bone marrow cultures. J Cell Physiol 112:345, 1982

55. Harrison DE, Lerner CP: Most primitive hematopoietic stem cells are stimulated to cyyle rapidly after treatment with 5-fluorouracil. Blood 78:1237,1991

56. Abkowitz JL, Ott RM, Holly RD, Adamson JW: Clonal evolution following chemotherapy-induced stem cell depletion in cats heterozygous for glucose-6-phosphate dehydrogenase. Blood 71:1687,1988

57. Rosse WF: Phosphatidylifiositol-linked proteins and parox- ysmal nocturnal hemoglobinuria. Blood 751595,1990

58. Moore JG, Humphries RK, Frank MM, Young N: Character- ization of the Bematopoietic defect in paroxysmal nocturnal hemoglobinuria. Exp Hematoll4222,1986

59. Rotoli B, Robledo R, Luzzatto L Decreased number of circulating BFU-Es in paroxysmal nocturnal hemoglobinuria. Blood 60157,1982

60. Fefer A, Freeman H, Storb R, Hill J, Singer J, Edwards A, Thomas E Paroxysmal nocturnal hemoglobinuria and marrow failure treated by infusion of marrow from an identical twin. Ann Intern Med 84692,1976

61. Selvaraj P, Rosse WF, Silber R, Springer T A The major Fc receptor in blood has a phosphatidylinositol anchor and is deficient in paroxysmal nocturnal haemoglobinuria. Nature 333565,1988

62. Simmons DL, Tan S, Tenen DG, Nicholson-Weller A, Seed B: Monocyte antigen CD14 is a phospholipid anchored membrane protein. Blood 73:284,1989

63. Selvaraj P, Dustin ML, Silber R, Low MG, Springer TA Deficiency of lymphocyte function-associated antigen 3 (LFA-3) in paroxysmal nocturnal heffioglobinuria. Functional correlates and evidence for a phosphatidylinositol membrane anchor. J Exp Med 166:1011,1987

64. Selvaraj P, Plunkett ML, Dustin M, Sanders ME, Shaw S, Springer T A The T lymphocyte glycoprotein CD2 binds the cell surface ligand LFA-3. NBture 326:400,1987

65. Shaw S, Ginther Luce GE, Quinones R, Gress RE, Springer TA, Sanders ME: Two antigen-dependent adhesion pathways used by human cytotoxic T cell clones. Nature 323:262,1986

66. Ben-Bassat I, Brok-Simoni F, Ramot B: Complement- sensitive red cells in aplastic anemia. Blood 46:357,1975

67. Nissen C, Gratwohl A, Speck B, Wiirsch A, Moser Y, Weis J:

For personal use only.on August 30, 2017. by guest www.bloodjournal.orgFrom

1392 NEAL S. YOUNG

Acquired aplastic anaemia: A PNH-like disease? Br J Haematol 64:355,1986

68. Rotoli B, Luzzatto L Paroxysmal nocturnal hemoglobinuria. Semin Hematol26:201,1989

69. 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

70. Kusminsky GD, Barazzutti L, Korin JD, Blasetti A, Tartas NE, Avalos JCS: Complete response to antilymphocyte globulin in a case of aplastic anemia-paroxysmal nocturnal hemoglobinuria syndrome [letter]. Am J Hematol29:123,1988

71. Appelbaum FR, Barrall J, Storb R, Ramberg R, Doney K, Sale GE, Thomas ED: Clonal cytogenetic abnormalities in patients with otherwise typical aplastic anemia. Exp Hematol 15:1134,1987

72. Abkowitz JL, Fialkow PJ, Niebrugge DJ, Raskind WH, Adamson J W Pancytopenia as a clonal disorder of a multipotent hematopoietic stem cell. J Clin Invest 73:258,1984

73. Eaves CJ, Cashman JD, Kay RJ, Dougherty GJ, Otsuka T, Gaboury LA, Hogge DE, Lansdorp PM, Eaves AC, Humphries R K Mechanisms that regulate the cell cycle status of very primitive hematopoietic cells in long-term human marrow cultures. 11. Analysis of positive and negative regulators produced by stromal cellswithin the adherent layer. Blood 78:110, 1991

74. Yu J, Shao L-E, Lemas V, Yu A, Vaughan J, Rivier J, Vale W Importance of FSH-releasing protein and inhibin in erythrodif- ferentiation. Nature 330765,1987

75. Maciejewski JP, Liu JM, Green SW, Walsh CE, Plumb M, Pragnell IB, Young NS: Expression of a stem cell inhibitor (SCI/LD78) gene in patients with bone marrow failure. Exp Hematol (in press)

76. Mayani H, Guilbert LJ, Clark SC, Janowska-Wieczorek A Inhibition of hematopoiesis in normal human long-term marrow cultures treated with recombinant human macrophage colony- stimulating factor. Blood 78:651,1991

77. Zoumbos N, Gascon P, Djeu J, Young NS: Interferon is a mediator of hematopoietic suppression in aplastic anemia in vitro and possibly in vivo. Proc Natl Acad Sci USA 82188,1985

78. Caux C, Favre C, Saeland S, Duvert V, Durand I, Mannoni P, Banchereau J: Potentiation of early hematopoiesis by tumor necrosis factor-alpha is followed by inhibition of granulopoietic differentiation and proliferation. Blood 78:635,1991

79. Homans CA, Cohen U, Barker EB, Mazur EM: Aplastic presentation of acute lymphoblastic leukemia: Evidence for cellu- lar inhibition of normal hematopoietic progenitors. Am J Ped Hematol Oncol4456,1989

80. Ohmori M, Ohmori S, Ueda Y, Tohyama K, Yoshida Y, Uchino H: Myelodysplastic syndrome (MDS)-associated inhibitory

activity on haemopoietic progenitor cells. Br J Haematol 74:179, 1990

81. Wyllie AH, Kerr JFR, Currie A R Cell death: The signifi- cance of apoptosis. Int Rev Cytol68:251,1980

82. &hen JJ: Programmed cell death in the immune system. Adv Immunol50:55,1991

83. Sellins KS, Cohen JJ: Cytotoxic T lymphocytes induce different types of DNA damage in target cells of different origins. J Immunol147:795,1991

84. Sellins KS, Cohen JJ: Gene induction by gamma-irradiation leads to DNA fragmentation in lymphocytes. J Immunol 139:3199, 1987

85. Williams GT, Smith CA, Spooncer E, Dexter TM, Taylor DR Haemopoietic colony stimulating factors promote cell survival by suppressing apoptosis. Nature 343:76,1990

86. Distelhorst CW: Glucocorticosteroids induce DNA fragmen- tation in human lymphoid leukemia cells. Blood 72:1305,1988

87. Koury MJ, Bondurant M C Erythropoietin retards DNA breakdown and prevents programmed death in erythroid progeni- tor cells. Science 248:378, 1990

88. Schmid DS, Tite JP, Ruddle NH: DNA fragmentation: Manifestation of target cell destruction mediated by cytotoxic T-cell lines, lymphotoxin-secreting helper T-cell clones, and cell- free lymphotoxin-containing supernatant. Proc Natl Acad Sci USA 83:1881,1986

89. Clouston WM, Kerr JFR: Apoptosis, lymphocytotoxicity and the containment of viral infections. Med Hypotheses 18:399,1985

90. Martz E, Howell DM: CTL: Virus control cells first and cytolytic cells second? Immunol Today 10:79,1989

91. Sellins KS, Cohen JJ: Polyomavirus DNA is damaged in target cells during cytotoxic T-lymphocyte-mediated killing. J Virol 63:572,1989

92. Sulecki M, Shadduck RK, Zeigler Z Anti-thymocyte globu- lin for hypoplastic myelodysplastic syndrome. Blood 72:229a, 1988 (abstr, suppl 1)

93. Litzow MR, Kyle RA: Multiple responses of aplastic anemia to low-dose cyclosporine therapy despite development of a myelo- dysplastic syndrome. Am J Hematol32:226,1989

94. Miecher PA, Favre H, Beris P: Autoimmune myelodyspla- sias. Semin Hematol28:322,1991

95. de Planque MM, van Krieken JHJM, Kluin-Nelemans HC, Colla LPJM, van der Burgh F, Brand A, Kluin PM: Bone marrow histopathology of patients with severe aplastic anaemia before treatment and at follow-up. Br J Haematol72:439,1989

96. Young NS: The pathogenesis and pathophysiology of aplas- tic anemia, in Hoffman R, Benz EJ Jr, Shattil SJ, Furie B, Cohen HJ (eds): Hematology. Basic Principles and Practice. New York, NY, Churchill Livingstone, 1991, p 122

For personal use only.on August 30, 2017. by guest www.bloodjournal.orgFrom

1992 79: 1385-1392  

NS Young restated [see comments]The problem of clonality in aplastic anemia: Dr Dameshek's riddle, 

http://www.bloodjournal.org/content/79/6/1385.citation.full.htmlUpdated information and services can be found at:

Articles on similar topics can be found in the following Blood collections

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

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

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

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

For personal use only.on August 30, 2017. by guest www.bloodjournal.orgFrom