Sie sind auf Seite 1von 19

Advances in Anatomic Pathology

Vol. 10, No. 1, pp. 8–26


© 2003 Lippincott Williams & Wilkins, Inc., Philadelphia

Review Article

Bone Marrow Biopsy: Interpretive Guidelines For the


Surgical Pathologist

James D. Cotelingam

Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, U.S.A.

Summary: Ideally, the bone marrow core biopsy should be reviewed with knowledge
of the clinical history, complete blood count, and findings in the peripheral blood and
bone marrow aspirate smears. However, for a variety of reasons, the pathologist may
receive the core biopsy and aspirate clot section without all of this information.
Although this approach is not optimal, a great deal of valuable information can be
generated from these specimens. Over the past 20 years, there has been considerable
progress in the fields of flow cytometric analysis, immunohistochemistry, and mo-
lecular diagnostic studies that can be performed on smears or extracted DNA from
paraffin embedded tissue. These modalities have augmented and refined diagnostic
criteria formerly ascertained by light microscopy, cytochemistry, and cytogenetics.
This is particularly true of some myeloid and lymphoreticular neoplasms where a
collaborative and multidisciplinary approach to the diagnosis has become necessary.
Despite this growing complexity and dependence on newer methodologies, the tradi-
tional role of histopathology in evaluating the bone marrow biopsy remains as impor-
tant as it has been in the past. In this review, we focus on contemporary practices and
expectations for interpreting bone marrow biopsies and clot sections. Key Words:
Bone marrow core biopsy—Immunohistochemistry—Fluorescence in situ hybridiza-
tion—Polymerase chain reaction.

INTRODUCTION tainment from one medical facility to another, Anatomic


Pathology Services often receive only the core biopsy
Assessment of changes in the bone marrow is an im- and clot section, while other components of the bone
portant prerequisite for the care and stratification of pa- marrow specimen are submitted elsewhere for evalua-
tients with disease of the hematopoietic system. Located tion. Despite this fragmented and unsatisfactory ap-
within the labyrinth of the intertrabecular and medullary proach to total specimen interpretation, the surgical pa-
spaces of bone, this highly specialized organ with com- thologist can ascertain considerable clinically relevant
plex hematopoietic and immunologic functions provides pathologic detail by supplementing traditional histopa-
an excellent substrate for pathologic investigation in- thology on paraffin embedded tissue (PET) with state of
cluding the staging of nonhematopoietic neoplasms and the art immunohistochemistry (IHC) and molecular di-
the monitoring of response to therapy. agnostic studies.
Due to variations in logistical detail, organizational
experience, billing practices, and attempts at cost con-

Address correspondence and reprint requests to Dr. James D. ROLE OF THE BONE MARROW BIOPSY IN
Cotelingam, Department of Pathology, Louisiana State University
Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71330. PATIENT CARE
The opinions published herein are the private views of the author and
do not reflect the official position of the Louisiana State University Ever since the role of the bone marrow in hematopoi-
Health Science Center. esis was described in the German literature by Neumann

8
BONE MARROW BIOPSY 9

(1) and Bizzozero (2) in 1868, and the first biopsy was ● To explain abnormal morphologic changes observed
performed by Mosler (3) in 1876, there have been re- in the peripheral blood, such as tear drop erythrocytes,
markable improvements in the instrumentation and tech- rouleaux, myeloid and lymphocytic immaturity, and
niques of specimen acquisition, analysis, and interpreta- leukoerythroblastosis.
tion. ● To assess marrow cellularity and to monitor the prog-
Before the paper by Turkel and Bethel (4) in 1943, ress of therapy.
pathologic reports were based primarily on the micro- ● To determine the adequacy and location of stainable
scopic features of aspirated smears stained with Ro- bone marrow iron.
manowsky dyes. Subsequently, however, there has been ● To evaluate for bone marrow involvement by meta-
increasing appreciation that besides providing correla- static disease, lymphoma, plasma cell dyscrasia, my-
tion with the cytologic features of the aspirate, the core elofibrosis, and mast cell disease.
biopsy offered a reliable means to quantitate cellularity ● As part of the workup for fever of unknown origin, to
and evaluate architectural detail in situ (5). Additionally, assess for bone marrow involvement in infections,
the core biopsy enables assessment of fibrosis, the iden- granulomatous disorders, and in patients with unex-
tification of focal lesions (as in lymphoma, myeloma, plained adenopathy and hepatosplenomegaly.
metastatic tumor, and granulomata), and is a repository ● To obtain cellular bone marrow for cytochemistry,
of material for immunohistochemical and molecular di- flow cytometry, immunohistochemistry, molecular
agnostic testing. Furthermore, the biopsy provides an op- genetics, cytogenetics, microbial cultures, electron mi-
portunity to report on the condition of bony trabeculae, croscopy, and tissue culture.
blood vessels, and other nonhematopoietic stromal and ● To evaluate for bone marrow involvement in sus-
cellular components of the intertrabecular space (5,6). pected storage and collagen vascular disorders.
In our experience, medical facilities that support speci- ● To define the etiology of unexplained osteosclerosis
men acquisition, analysis, and interpretation by an orga- and other abnormalities of trabecular bone detected by
nized and interactive team of health care professionals radiologic studies.
including medical technologists, allied scientists, clini-
cians, and pathologists working in concert, offer the ad- PREPARATION OF BONE MARROW FOR
vantage of conforming to operational guidelines and HISTOPATHOLOGIC EXAMINATION
standards that cater to the needs of all specialties within
a healthcare facility at minimum cost and patient dis- Information on the types of biopsy needles, and guide-
comfort. lines for acquisition of the aspirate and core biopsy are
To maximize yield, it is recommended that before well documented in the literature (7–10). It is important
specimen procurement, the clinical picture be correlated that personnel obtaining bone marrow biopsies are fa-
with the complete blood count (CBC) appearances of the miliar with the range of tests possible on each specimen
peripheral blood smear and other existing laboratory data component, and are aware that bilateral core biopsies are
to anticipate which specimen components of the bone recommended for staging malignant lymphomas and
marrow are likely to yield information most valuable to metastatic carcinoma, and by some for the diagnosis of
the case under study. Such an ideal scenario is, however, multiple myeloma at initial work-up.
the exception rather than the rule, and in most cases, the The need to establish high expectations for quality
surgical pathologist has little control over the events an- histologic preparations cannot be overemphasized. In
tecedent to receiving slides on a bone marrow specimen fact, the cardinal sin of misdiagnosis frequently results
for sign out. from erroneous interpretation of inadequate or poorly
processed material.
Methodologies for processing the bone marrow core
INDICATIONS FOR BONE
and clot section vary considerably with institutional poli-
MARROW ANALYSIS
cies and philosophical expectations. In practice, routine
● To explain a decrease in the formed elements of the sections should be ready for interpretation within 24
blood as in anemia, leukopenia, thrombocytopenia, hours after specimens are placed on the tissue processor.
and myelodysplasia. The choices of fixative, decalcifying agent, and stains
● To determine the causes of increased formed elements vary from one institution to the other. A selected few,
of the blood as in unexplained sustained leukocytosis, which are in general use, are described below.
erythrocytosis, thrombocytosis, the myeloproliferative Fixation of the core biopsy and clot section may be
disorders, and the leukemias. achieved with a zinc formalin fixative such as B-plus Fix

Advances in Anatomic Pathology, Vol. 10, No. 1, January, 2003


10 J. D. COTELINGAM

(BBC, Stanwood, WA), B-5, or neutral buffered forma- stain imports a purple hue to the cytoplasm of plasma
lin. Thereafter, the specimen should be decalcified in a cells, prominently highlights the metachromatic granules
10% aqueous solution of nitric acid, Redecal (a 12.5% of mast cells and basophils, and enhances the tinctorial
aqueous solution of hydrochloric acid in EDTA, Stat Lab properties of neutrophilic and eosinophilic granules. Ad-
Medical Products Inc., Lewisville, TX) or RDO (APB ditional immunohistochemical stains may be necessary,
Engineering Products Corporation, Plainfield, IL), an- and are further discussed below.
other proprietary solution of hydrochloric acid in a coal Samples for molecular studies require special han-
tar base ensuring good histologic sections, without dam- dling. For instance, lesions for assessment by fluores-
age to the microtome knife. Decalcification results in the cence in-situ hybridization (FISH) may need to be local-
leaching out of some storage iron from the core biopsy, ized for the benefit of personnel in the molecular diag-
and nitric acid and hydrochloric acid have been reported nostic laboratory. Paraffin sections for FISH are
to diminish the acid fastness of mycobacteria (11,12) submitted on Superfrost Plus glass slides (Fisher Scien-
resulting in false negative results. Acid fastness is how- tific, Pittsburgh, PA). For the polymerase chain reaction
ever retained when decalcification is in formic acid- (PCR), scrolls of paraffin embedded tissue are deparaf-
sodium citrate or citric acid buffer (12). Therefore, these finized before DNA isolation in accordance with local
are the decalcifying agents of choice when the need to policies and practices.
demonstrate mycobacteria can be anticipated. Combina-
tion fixative/decalcification preparations in use are Zen- EXPECTED DIAGNOSTIC YIELD FROM
ker’s solution (13), Surgipath Decalcifier (a formic-acid COMPONENTS OF THE BONE
formalin mixture, Surgipath Medical Industries, Rich- MARROW SPECIMEN
mond, IL), and Decal Plus (an aqueous solution of for-
malin, formic acid, and methanol, Stat Lab Medical Not every item in the preceding list of indications
products, Inc. Lewisville, TX). An advantage of Zenk- prompting bone marrow analysis can be resolved by ex-
er’s solution and B-5 is a mordanting effect that en- amination of the core biopsy and clot section. Further-
hances the tinctorial properties of the Giemsa and hema- more, the optimum yield of diagnostic information from
toxylin and eosin (H & E) stains. Additionally, glacial individual components of a bone marrow specimen in-
acetic acid, the decalcifying component in Zenker’s so- nately vary, and often reflect the type of disorder being
lution counteracts the cell shrinkage resulting from fixa- investigated (21). In general, the aspirate and touch
tion. Metallic fixatives such as Zenker’s and B-5 degrade preparations provide qualitative cytologic detail, while
DNA and impair subsequent molecular diagnostic test- the core biopsy and clot section provide quantitative in-
ing. This limitation is circumvented by fixation in a zinc- formation. Because the surgical pathologist frequently
formalin fixative such as B-plus Fix. It is noteworthy that receives only PET, it is important to identify the expec-
the manufacturer’s specified ratio of specimen volume to tations and limitations of the diagnostic yield from such
fixative/decalcifying agent and treatment times be material. These are enumerated and compared to those
closely followed to achieve optimum results. from the aspirate and touch preparations in Table 1
Clot sections do not need to be decalcified. Bone dust (5,6,21–36), and are further discussed below.
and small fragments of bony trabeculae that are occa-
sionally aspirated when acquisition of the core biopsy INTERPRETATION OF PARAFFIN EMBEDDED
precedes that of the aspirate, can usually be cut through TISSUE AND DATA INTEGRATION
without decalcification. Agar embedding techniques (14)
for processing the clot section, plastic embedding (15,16) For most anatomic pathologists, a bone marrow
of the undecalcified core and electron microscopy (16– sample is often one among a number of surgicals in a run
20) of aspirated material are described in the literature; requiring interpretation and sign-out. Although the pa-
however, these techniques are not widely used for rou- tients name, age, and gender are part of the standard
tine purposes. biographic template, information concerning the clinical
Ideally, histologic sections of the core and clot section picture, CBC, peripheral blood film, and bone marrow
should be cut at 4 micron. In addition to routine hema- aspirate are often absent or inadequate, and need to be
toxylin-eosin stained sections, other stains of value in- procured to ensure a meaningful interpretation. The
clude iron (preferably performed on the nondecalcified guidelines used in evaluating a bone marrow specimen
aspirate clot section), reticulin, periodic acid-Schiff stain are understandably parochial. However, most observers
(PAS), and Giemsa. Although difficult to perform, a integrate the microscopic features of the specimen with
well-done Giemsa preparation can be very helpful. This the ancillary data listed in Table 1 into a synopsis, un-

Advances in Anatomic Pathology, Vol. 10, No. 1, January, 2003


BONE MARROW BIOPSY 11

TABLE 1. Summary of differences between aspirated and paraffin embedded bone marrow samples
Component/parameter Aspirate Touch preps of clot/biopsy Clot section Core biopsy
Fat:Cell ratio Unsuitable for estimation Recommended specimens for estimation
Myeloid: Erythroid ratio Calculated from a Count differentials Working approximation possible
differential count correlate with those
of the aspirate
Myelopoiesis Stages of maturation accurately classifiable Normal myeloblasts and promyelocytes cannot
be discriminated; however,stages thereafter,
ALIP and AML blasts may be
Erythroid component Stages of maturation can be accurately designated Identifiable, but stages cannot be specifically
classified
Megakaryocytopoiesis Stages of maturation accurately classifiable Normal megakaryoblasts not identifiable, stages
thereafter classifiable generically as
megakaryocytes
Lymphocytes Small and scattered Normal lymphocytes can be differentiated from
NRBC, T/B phenotypes can be designated by IHC
Plasma cells Readily identified Can be designated in H/E stained sections,
readily enumerated in IHC preps
Metastatic tumor Appearances characteristic of cell type Identifiable and quantifiable.
Reticulin Not quantifiable Quantifiable
Collagen Not quantifiable Quantifiable
Granulomata Seldom observed, cannot be ruled out from these Specimen components of choice to
specimen components evaluate for granulomata
Iron stores Quantitation reliable, Quantitation unreliable, Quantitation reliable, Quantitation and
best for identification RSB identifiable RSB may be dentification
of RSB identified of RSB unreliable
Monocytes/Macrophages Monocytes, sea blue histocytes and other pathologic Monocytes not identifiable, tingible body
forms can be designated, and confirmed by macrophages and pathologic forms identifiable,
cytochemistry confirmation possible with IHC
Stromal space and mesenchyme Accurate assessment not possible Can be identified Best specimen
component
to assess
Arterial/Venous Structures Capillaries may be seen Not observed Seldom apparent Routinely observed
Marrow sinusoids Not observed Normally apparent,
dilated in
fibrosing states
Trabecular bone Not part of specimen Present, may offer
diagnostic
information
Cytochemistry Specimen of choice Can be performed Selected preparations feasible
Flow cytometry Specimen of choice Not possible Possible by vortex disaggregation of unfixed material
Immunohistochemistry Possible Specimen components of choice
Molecular Diagnostics Possible
Cytogenetics Specimen of choice Not possible Feasible on vortex disaggregated material

ALIP, Abnormally located immature precursors; AML, Acute myeloid leukemia; RSB, Ring sideroblasts; NRBC, Nucleated red blood cells; IHC,
Immunohistochemistry.

derscored by a diagnostic line and a comment, if neces- ● The fat:cell ratio.


sary. To ensure that no aspect of the examination is in- ● The myeloid:erythroid ratio.
advertently omitted, and to standardize reports, a check- ● A description of the hematopoietic cell lines (i.e., my-
list (5) such as the one below is recommended: elopoiesis, erythropoiesis, and megakaryocytopoiesis).

Advances in Anatomic Pathology, Vol. 10, No. 1, January, 2003


12 J. D. COTELINGAM

● A quantitation and description of lymphocytes and evaluation of the fat:cell ratio is possible only on paraffin
plasma cells. embedded tissue, and areas with stromal hemorrhage,
● Evaluation of stainable iron. dropout, and aspiration artifact should be excluded. In
● Exclusion or description and quantitation of neoplastic cases with unexplained aplasia without peripheral blood
infiltrates such as blast forms, lymphoma, myeloma, cytopenia, it is advisable to recommend a contralateral
and metastatic tumor. core biopsy to rule out sampling error.
● Exclusion or description of infections and granuloma-
tous changes. QUANTITATION OF CELLULAR
● Exclusion or description of fibrosis. COMPONENTS IN THE BONE MARROW
● A statement, if necessary, to cover any miscellaneous
abnormality observed in the macrophage system, bone Differential counts of cellular elements in the bone
marrow stroma, vascular structures, and trabecular marrow are traditionally expressed in percentile figures
bone. (6,27,41–50). Included herein are blast forms, myelo-
● A concluding comment integrating the above with the monocytic, erythroid, megakaryocytic, plasma cellular,
results of any corroborating cytochemical, flow cyto- and lymphoid cell lines. Macrophages, stromal cells, os-
metric, immunohistochemical, molecular diagnostic, teoclasts, osteoblasts, fat cells, and metastatic tumor cells
cytogenetic, or other contributing studies. are excluded. The “hematopoietic” compartment is com-
prised of all myelomonocytic, erythroid, and megakaryo-
THE FAT:CELL RATIO cytic elements. All other forms are classified as “nonhe-
matopoietic.” Quantitation of the extent of involvement
It is the best to evaluate marrow cellularity and the by tumor does convey meaning, and should be expressed
fat:cell ratio from the core biopsy. Although less reliable, as a percentage of the involved marrow space or total
these may also be assessed from clot sections with mul- marrow cellularity.
tiple marrow particles. Variations in the ratio of fat to
cell in the bone marrow are an important index of bone MYELOID:ERYTHROID RATIO
marrow activity. At birth and during early childhood, the
bone marrow is 100% cellular and virtually devoid of fat. At birth, the myeloid:erythroid (M:E) ratio is approxi-
By the end of the first decade, the overall fat:cell ratio is mately 1.5:1. The myeloid (granulocytic) component
approximately 10:90, and in adults (Fig. 1), it varies gradually increases and peaks at approximately 6:1 by
between 30:70 and 70:30 with increasing age. Slight the end of the first week of life (51). Thereafter, values
physiologic variations of the ratio from one microscopic plateau at a physiologic range of 2.5:1 to 4:1, with little
field to another are not uncommon and review of the variation throughout life (6,20,51). Increase in either
slide with a scanning objective can enable definition of component is reported as myeloid/erythroid “predomi-
an overall ratio. Beyond the seventh decade, bone mar- nant” in the presence of a normal fat:cell ratio, and “hy-
row fat may be expected to increase by approximately perplasia” when the cellularity of the bone marrow ex-
10% with each passing decade (37–39). Despite this in- ceeds 70%. While an M:E ratio can be calculated from a
creasing hypocellularity, the CBC remains within physi- 500 cell count down to a decimal point in the aspirate,
ologic limits. However, in the presence of otherwise un- paraffin embedded tissue provides only a coarse approxi-
explained cytopenia, a fat content of greater than 70% mation of the same. However, paraffin embedded tissue
reflects hypoplasia. Quantitation of the latter is usually has the advantage of providing an overview of this ratio
expressed as mild, moderate, or severe. in situ, and in different areas of the specimen. Also,
The word “aplasia” is derived from Greek (40), and eliminated herein is any bias that may be manifest in
connotes total fatty replacement of the bone marrow. particle-depleted smears of sinusoidal blood. It is best to
Aplasia may be generalized or focal (spotty). The de- read M:E ratios on core biopsies, away from paratrabecu-
creased cellularity within the stromal space that results lar areas, which are normally myeloid predominant. Be-
from chemotherapeutic induction of acute leukemia (Fig. cause of their characteristic cytologic features, the my-
2), serous fat atrophy (Fig. 3), hypoproliferative states, eloid component beyond the promyelocyte stage can be
and fibrosis are not accompanied by increased bone mar- readily designated with a 40 x objective, right up to the
row fat. Therefore, these are examples of hypocellularity neutrophil, eosinophil, and mast cell stage. Similarly,
rather than true aplasia. Under these circumstances, it is nucleated erythroid elements beyond the early normo-
considered more meaningful to report a ratio incorporat- blast stage can be differentiated from lymphocytes by
ing the relationships of fat, cell, and stroma. Accurate their round, dense, and pyknotic nuclei. In contrast to

Advances in Anatomic Pathology, Vol. 10, No. 1, January, 2003


BONE MARROW BIOPSY 13

FIG. 1. Normocellular bone marrow biopsy. The fat:cell ratio is estimated at 60:40; H&E section (original magnification x 100).
FIG. 2. Adequate chemotherapeutic induction. Core biopsy from a patient with AML-M2 at day 14; H&E section (original magnification x 400).
FIG. 3. Serous fat atrophy. Deposits of acid mucopolysaccharide are located between fat cells. Core biopsy. (A) H&E section. (B) Alcian blue stain
(original magnification x 400).
FIG. 4. Normal M:E ratio. A PAS section of the core biopsy with an M:E ratio of approximately 4:1. Intensity of PAS activity increases with
granulocytic maturation. Erythroid precursors have deeply hematoxyphilic nuclei (original magnification x 400).
FIG. 5. Acute myeloid leukemia (AML-MO). Clot section at diagnosis. Most normal hematopoietic cells in the stromal space are replaced by the
leukemic infiltrate; H&E section (original magnification x 400).
FIG. 6. Chronic myeloid leukemia, accelerated phase. Core biopsy. Myeloblasts are less than 20% of marrow cellularity. Myeloperoxidase
immunostain (original magnification x 400).
FIG. 7. Myelodysplasia. Two morphologic features observed in core biopsies. (A) Abnormal localization of immature myeloid precursors, H&E
section (Courtesy of Dr. Kathryn Foucar). (B) Myeloblasts in stromal space estimated at less than 20% of marrow cellularity; H&E section (original
magnification x 400).
FIG. 8. Megaloblastic anemia. Note megaloblastic proliferation with interspersed myeloid component including giant metamyelocytes; H&E section
(original magnification x 400).
FIG. 9. Erythroleukemia, AML-M6B. (A) Blast forms in an H&E section. (B) Blast forms immunostained by the immunoperoxidase technique for
hemoglobin A (original magnification x 400) (Courtesy of Dr. Harold Schumacher).
FIG. 10. Dysplastic megakaryocytes including micromegakaryocytic forms from a patient with myelodysplasia. (A) H&E section. (B) FVIII RA
immunostain (original magnification x 400).
FIG. 11. Essential thrombocythemia. Core biopsy demonstrating increased megakaryocytic endoreduplication and abutment; H&E section (original
magnification x 400).
FIG. 12. Follicular lymphoma (grade I). Core biopsy. Tumor was located in paratrabecular and intertrabecular areas. (A) H&E section. (B) BCL2
preparation (original magnification x 400).
14 J. D. COTELINGAM

appearances in H & E preparations, cells of myeloid and sions of mast cell lineage can be specifically confirmed
erythroid lineage are easier to recognize in Giemsa and with mast cell tryptase and CD117 (C-Kit) preparations
PAS stains (Fig. 4), and can be designated with certainty (57–60), and TdT activity may be apparent in cases of
on myeloperoxidase (MPX) and hemoglobin peroxidase acute mixed lineage leukemia (61,62). Although the cy-
(HbPX) preparations of paraffin embedded tissue. It is tologic features of blast forms in AML on PET are not
noteworthy that monocytes and lymphocytes (also re- specific for the subtypes of AML according to the FAB
ferred to as nongranulocytes), plasma cells, and mega- (French-American-British) scheme, cases of AML-M3,
karyocytes are not included in the M:E ratio. AML-M4, and AML-M5B do tend to have “monocyt-
oid” nuclei. Also, minimal myeloid differentiation is ob-
MYELOID SERIES served in AML-M2, and an abnormal eosinophil com-
ponent is apparent in AML-M4EO. Abnormal localiza-
The myeloid component of the bone marrow arises tion of immature myeloid precursors (ALIP) (Fig. 7) in
from self-replicating stem cells, which also gives rise to the intertrabecular space is a feature of myelodysplasia
the monocytic, erythroid, and megakaryocytic cell lines. (63–65). Increased expression of PCNA antigen and
Data on the physiologic range of myeloid precursors and CD34 activity by immunostaining on PET serve to dis-
mature forms in the bone marrow (Table 2)(41–43) have tinguish hypoplastic myelodysplastic syndromes from
been derived from aspirate preparations where the cyto- acquired aplastic anemia, where these parameters are de-
logic features of each cell type are readily appreciated creased (66). While immunostaining with CD34 is of
under an oil immersion lens. Even though quantitation of value in identifying immature populations of leukemic
similar information on PET is in contrast largely subjec- cells, it does not distinguish between a myeloid and a
tive, and unlike aspirate counts lacks mathematical pre- lymphoid lineage. Without incorporating data generated
cision, it does have clinical utility. from examination of the aspirate, cytochemistry, flow
In our experience, normal myeloblasts and promyelo- cytometry, cytogenetics, and molecular diagnostic stud-
cytes are not readily differentiated from pronormoblasts ies, it may be impossible to classify myeloid, and mixed
and early normoblasts in PET with a high dry objective. lineage lesions, according to the recent recommendations
However, mature forms beyond the promyelocyte stage of the Society for Hematopathology and World Health
and pathologic shifts in compartment size as in myeloid Organization (WHO) expert committee (67–69).
hyperplasia, the myelodysplastic syndromes (MDS), the In cases where no aspirate is available due to a dry tap,
acute myeloid leukemias (AML) AML-MO (Fig. 5) vortex disaggregation of leukemic cells from the unfixed
through M6A, some of the myeloproliferative disorders, core biopsy and clot can serve as a resource of material
and chronic leukemias of myeloid origin are readily dif- for cytogenetics and flow cytometry. With the introduc-
ferentiated. Confirmation of myeloid lineage in PET may tion of newer generations of therapy such as Myelotarg
be achieved by specific esterase (Leder), MPX (Fig. 6) (70) (formerly known as CMA-676), which is specifi-
and other immunostains (28–31,52–55); and a mono- cally directed against the CD33 epitope in AML, and
cytic lineage may be supported by the identification of STI571, (71,72) which targets and inhibits bcr-abl tyro-
CD68, lysozyme, alpha-1-antitrypsin, and alpha-1- sine kinase in chronic myeloid leukemia (CML), there is
antichymotrypsin activity in AML-M4, AML-M5 and a growing expectation that laboratory identification of
chronic myelomonocytic leukemia (CMML)(56). Le- such loci will become available as part of the diagnostic
armamentarium. Furthermore, it has now become appar-
TABLE 2. Types of myeloid elements and their normal ent that in patients with CML on STI571, despite micro-
range in the bone marrow
scopic restoration of marrow cellularity with normal M:E
Cell type Range ratios, bcr-abl fusion persists (72) and may be demon-
Myeloblasts 0% to 2% strated by FISH on the core biopsy and clot section
Promyelocytes 2% to 5% (Table 3). This technology is therefore certain to influ-
Myelocytes (neutrophilic) 9% to 16% ence therapeutic protocols for STI571 in the future. Ac-
Metamyelocytes 7% to 23%
Band Forms 8% to 15% cording to the WHO expert panel and the Society for
Neutrophils 4% to 10% Hematopathology, the threshold for transition of blast
Myelocytes (eosinophilic) 0% to 2% counts from myelodysplastic syndrome (MDS) (Fig. 7)
Band 0% to 2%
Mature 0% to 2% to AML is set at 20% with the exception of lesions that
Monocytes/macrophages 0% to 3% include a t(8;21) chromosomal abnormality (67,68,73).
Basophils 0% to 1% Despite lower blast counts, this entity, which would have
Mast Cells 0% to 2%
been formerly classified as MDS, is now, because of a

Advances in Anatomic Pathology, Vol. 10, No. 1, January, 2003


BONE MARROW BIOPSY 15

TABLE 3. Lineage specific molecular cytogenetic probes* currently in use to identify myeloid lesions by fluorescence in situ
hybridization (FISH) on core biopsies of bone marrow
FISH Signal color
Disease Locus Chromosome abnormality Green Orange Yellow
AML-M0/M1,MDS EGR1 5q Deletions-interstitial (5q31) 5p15.2 (control) 5q31-EGR1 N/A
AML-M0/M1, MDS CSF1R 5q Deletions-telomeric(5q33-q34) 5p15.2 (control) 5q33-CSF1R N/A
AML-MDS D20S108 (20q12) Deletions in 20q N/A 20q12 N/A
AML/MDS D7S486 (7q31) Deletions in 7q, i(7q) Alpha-satellite 7 D7S486 (7q) N/A
(7p11.1-q11.1)
AML/MDS 8p11.1-q11.1 Trisomy 8 N/A Alpha-satellite 8 N/A
(8p11.1-q11.1)
AML-M2 AML1/ETO t(8;21)(q22;q22) 8q-ETO (CBFA2T1) 21q-AML1 (RUNX1) 8;21 Fusion signal
AML-M3 (variant) RAR␣ t(11;17)(q23;q21) and related X/RARA Abnormal-1 green Abnormal-1 orange Dual color (normal)
(split signal) (split signal)
AML-M3 PML/RAR␣ t(15;17)(q22;q21.1) 17q-RARA 15q-PML 15;17 Fusion signal
AML-M4EO CBF␤ inv(16)(p13q22) and t(16;16)(p13;q22) Abnormal-1 green Abnormal-1 orange Dual color (normal)
(split signal) (split signal)
AML-M5 and Mixed lineage MLL(ALL-1, HRX) 11q23 rearrangements- Abnormal-1 green Abnormal-1 orange Dual color (normal)
t(4;11)(q21;q23),t(9;11)(p22;q23), (split signal) (split signal)
t(11;19)(q23;p13) and variants
CML BCR/ABL t(9;22)(q34;q11.2) 22q-BCR 9q-ABL 9;22 Fusion signal

* Vysis Inc. Downers Grove, IL.


Tabulated by Mary L. Nordberg PhD.

superior long-term outlook in adults, diagnosed and and resemble the neoplastic proliferations of acute leu-
treated earlier in its natural history as AML (69,73). The kemia and large cell lymphoma (LCL). Unlike these con-
supportive role of molecular diagnostic studies, some of ditions however, myeloid elements including giant meta-
which are now available on PET (Table 3), is therefore of myelocytes are interspersed between megaloblasts. Un-
paramount importance. Additionally, it is noteworthy der these circumstances, correlation of biopsy findings
that the malignant progranulocytes of AML-M3 be in- with other laboratory data such as the appearances of the
cluded in the total blast count, as should the myeloblasts aspirate, CBC, serum LDH, folate, and B12 can be in-
of erythroleukemia AML-M6A (74,75) (traditional valuable. Arrested erythroid maturation, giant pronormo-
erythroleukemia of the FAB classification). blasts, and intranuclear inclusions are manifestations of
parvovirus B19 infection (76). Herein, viral material can
ERYTHROID SERIES be positively identified by in situ hybridization (ISH) in
PET. Multinucleated erythroid precursors may be ob-
The erythroid component of the bone marrow nor- served in CDA (77), where additional confirmatory he-
mally comprises between 15% and 37% of a differential matological testing such as the Ham and Sugar Water
cell (6,42,43,52) count that includes granulocytes, mega- tests become necessary. In cases of AML-M6B (74,75,
karyocytes, lymphocytes, and plasma cells. Maturing 78), bizarre multinuclearity of leukemic erythroid pre-
erythroid precursors include pronormoblasts, early nor- cursors may be a prominent feature and lineage identity
moblasts, intermediate normoblasts, and late normo- can be established with HbPX immunostaining (Fig. 9)
blasts. Of these, pronormoblasts and early normoblasts on PET (79). In such cases, neoplastic pronormoblasts
cannot always be distinguished with certainty from my- should be included with myeloblasts in the overall blast
eloblasts and promyelocytes in PET with a high dry ob- count, and in keeping with the recommendations of the
jective. However, later stages of erythroid maturation Society of Hematopathology and WHO, cases with a
with their characteristic round, dense and deeply baso- blast count under 20% be classified as myelodysplasia,
philic nuclei can be easily identified, and distinguished and those above as acute leukemia. Vortex disaggrega-
from interspersed lymphocytes. In circumstances where tion of cellular material from the unfixed core biopsy for
this distinction poses a problem, IHC identification of flow cytometric, cytogenetic, and molecular diagnostic
erythroid precursors with an HbPX immunostain is de- studies can be an invaluable adjunct in patients with a
finitive. This technology is also invaluable in confirming dry tap (34).
the absence of erythroid precursors in cases of total red
cell aplasia. Under normal circumstances and in regen- MEGAKARYOCYTES
erative states, clones of normoblasts tend to cluster in the Megakaryocytes are the largest of the hematopoietic
intertrabecular space. However, in florid megaloblastic cells, and comprise between 0.5% and 2.0% of all nucle-
hyperplasia (Fig. 8), marrow cellularity may be diffuse ated marrow elements (6,52,80). The earliest cell of

Advances in Anatomic Pathology, Vol. 10, No. 1, January, 2003


16 J. D. COTELINGAM

megakaryocytic lineage discernable microscopically is commercially, facilitating the evaluation of lymphocyte


the megakaryoblast, a mononucleated cell with scant subsets in PET by IHC. Beyond the third decade, benign
deeply basophilic cytoplasm rich in RNA. Megakaryo- nonparatrabecular lymphoid aggregates may be observed
blasts are slightly larger than myeloblasts and pronormo- and rarely may contain reactive germinal centers (91). In
blasts, from which they are not readily distinguished in aspirate smears, it is not uncommon to observe benign
PET with a high dry objective. Subsequent stages of polyclonal lymphoid aggregates as a monolayer of ma-
maturation are characterized by increased cell size, pro- ture lymphocytes.
gressive nuclear lobulation (endoreduplication), loss of In PET, monoclonal lymphoid infiltrates are usually
cytoplasmic basophilia, and increase in “cotton candy” characteristic of involvement by a chronic lymphoid leu-
like granules. Normal megakaryocytes are randomly dis- kemia (5) or a low or intermediate grade nonHodgkin
tributed among other hematopoietic cells, occasionally lymphoma. Rarely, follicle center cell lymphoma (FCL)
reveal mature hematopoietic cells within their cytoplasm first presents within the bone marrow in patients with
(emperipolesis), and only seldom make physical contact cytopenia but without organomegaly or adenopathy. The
with other megakaryocytes. Because megakaryocytes aspirate smears in these patients are commonly negative
normally comprise about 1:200 nucleated marrow cells, (92) but paratrabecular aggregates diagnostic of lym-
increase in the megakaryocytic compartment as in idio- phoma are found in the core (Fig. 12) and are CD20 and
pathic thrombocytopenic purpura and other reactive BCL2 positive (93). Such infiltrates may be accompa-
states can be readily appreciated with a low power ob- nied by some reactive CD3-positive T-cells. In patients
jective. Megakaryocytes are PAS positive, and in PET, with treated FCL, minimal residual (and occasionally
megakaryoblasts and megakaryocytes are FVIII related- intertrabecular) deposits of tumor can be identified by
antigen (von Willebrand factor), FXIII, CD41, and CD61 similar IHC procedures. This may correlate with the
positive by immunohistochemistry (81,82,83). This tech- presence of a combined CD19, CD10, and BCL2 popu-
nology is invaluable in establishing lineage identity in lation of lymphocytes on flow cytometric analysis. Large
acute megakaryocytic leukemia (AML M-7), where blast cell lymphoma has appearances in the bone marrow
forms may be indistinguishable from those of some other comparable to those seen in node-based biopsies (Fig.
forms of AML on H & E stains of PET (84,85). Immu- 13). The presence of BCL6 activity in LCL serves to
nohistochemistry also helps in the identification of dys- distinguish this lesion from blastic mantle cell lym-
morphic, dysplastic, and micromegakaryocytic forms phoma.
that may be observed in the myeloproliferative and my- Hodgkin lymphoma in the bone marrow (Fig. 14) is
elodysplastic disorders (Fig. 10). It is noteworthy that associated with fibrosis, accounting for the rarity with
megakaryocytes abut, cluster, sheet out, and reveal in- which Reed-Sternberg cells are observed in aspirate
creased endoreduplication in essential thrombocythemia preparations (94–98). As with the nonHodgkin lympho-
(Fig. 11). These features may be variably present in the mas, bilateral core biopsies are recommended for staging
other myeloproliferative disorders, but are less pro- of Hodgkin lymphoma (21). The true identity of mono-
nounced. Increased numbers of mature mononuclear nuclear Reed-Sternberg cells in the bone marrow can be
megakaryocytic forms may be observed in chronic my- established with antibodies specific for CD15 and CD30
eloid leukemia and the 5q− syndrome (86–89), and loss and other related immunostains (69).
of nuclear lobulation with development of small discrete In cases of bone marrow involvement by acute lym-
nuclei (“pawn ball” forms) may be apparent in myelo- phoblastic leukemia (Fig. 15) and the high-grade
dysplasia on H&E sections in PET. lymphomas (i.e., lymphoblastic lymphoma and Burkitt
lymphoma) (Fig. 15), the pattern of infiltration is invari-
LYMPHOCYTES AND ably interstitial and diffuse (6). Correlation of the mor-
LYMPHORETICULAR LESIONS phology with immunophenotypic, cytochemical, and cy-
togenetic data is invaluable (99–101). The identification
In the bone marrow of normal adults, lymphocytes are of a minimal residual population of acute lymphoblastic
small, randomly interspersed between hematopoietic leukemia (ALL) in PET can be achieved by immuno-
cells, and comprise between 8% and 24% of all nucleated staining for CD34 (Fig. 16), TdT, CD20, CD3, CD45RO,
elements (44–47). The ratio of T-cells to B-cells is ap- and additionally with molecular diagnostic studies for
proximately 3:1, and the CD4:CD8 ratio is approxi- IgH, TCR gene rearrangement, and the identification of
mately 2:1. In pediatric samples, the total lymphocyte abnormal genetic loci by PCR or FISH (Table 4). Also of
count is higher, and normal values up to 40% may be great importance is the distinction of acute lymphoblastic
observed (90). Labeled antibodies are now available leukemia from hematogones (102). The latter are B-

Advances in Anatomic Pathology, Vol. 10, No. 1, January, 2003


BONE MARROW BIOPSY 17

FIG. 13. Large cell lymphoma. Core biopsy. Diffuse involvement of the intertrabecular space. (A) H&E section. (B) CD20 preparation (original
magnification x 400).
FIG. 14. Hodgkin lymphoma. Core biopsy. Note solitary Reed-Sternberg cell and fibrosis (original magnification x 400).
FIG. 15. Acute lymphoblastic leukemia (A) and Burkitt lymphoma (B). H&E stained sections of core biopsies (original magnification x 400).
FIG. 16. Relapsed acute lymphoblastic leukemia. Clot section; CD34 immunostain (original magnification x 400).
FIG. 17. Chronic lymphocytic leukemia. Core biopsy. Interstitial pattern of infiltration. (A) H&E section and Wäldenstrom macroglobulinemia.
Core biopsy. Note Dutcher body. (B) PAS section (original magnification x 400).
FIG. 18. Adult T-cell leukemia/lymphoma. Core biopsy. Note erosion of bony trabecula adjacent to tumor; H&E section (original magnification
x 400).
FIG. 19. Anaplastic myeloma. Core biopsy; H&E section (original magnification x 400).
FIG. 20. Anaplastic myeloma. Core biopsy with kappa light chain restriction; Kappa immunostain (original magnification x 400).
FIG. 21. Metastatic carcinoma of breast. Core biopsy; H&E section (original magnification x 400).
FIG. 22. Kaposi sarcoma. Core biopsy; H&E section (original magnification x 400).
FIG. 23. Reticulin and collagen fibrosis in bone marrow core biopsies. (A) Gordon and Sweet’s reticulin stain with markedly increased reticulin.
(B) H&E section with total effacement of hematopoietic tissue by collagenous fibrosis. (C) Trichrome stain demonstrating total collagenous fibrosis
(original magnification x 400).
FIG. 24. Granulomatous changes in core biopsy of bone marrow. (A) H&E stain of epithelioid granuloma from patient infected with Mycobacterium
tuberculosis. (B) H&E stain of “ring granuloma” in patient with Q-fever (original magnification x 400).

Advances in Anatomic Pathology, Vol. 10, No. 1, January, 2003


18 J. D. COTELINGAM

TABLE 4. Lineage specific molecular cytogenetic probes* currently in use to identify lymphoid lesions by fluorescence in situ
hybridization(FISH) on core biopsies of bone marrow
FISH signal color
Chromosome
Disease Locus abnormality Green Orange Yellow
ALL TEL(ETV6)/AML1 t(12;21)(p13;q22) TEL AML1 t(12;21) Fusion
ALL BCR/ABL t(9;22)(q34;q11.2) BCR ABL 9;22 Fusion
ALL p16 del(9)(p21) Alpha-satellite 9 p16 N/A
(9p11.1-q11.1)
Burkitt Leukemia/ MYC t(8;14), t(2;8), t(8;22) Abnormal-1 green Abnormal-1 orange Dual color (normal)
Lymphoma (split signal) (split signal)
Burkitt Leukemia/ IgH/MYC t(8;14)(q24;q32) IgH MYC 8;14 Fusion signal
Lymphoma
Mantle cell lymphoma IgH/CCND1 t(11;14)(q13;q32) IgH CCND1 11;14 Fusion signal
Follicle center cell IgH/BCL2 t(14;18)(q32;q21) IgH BCL2 14;18 Fusion signal
lymphoma
Extranodal marginal zone D3Z1 +3 N/A Alpha-satellite 3 N/A
B-cell lymphoma of (3p11.1-q11.1)
mucosa-associated
lymphoid tissue (MALT
lymphoma)
Large cell lymphoma BCL6 t(?;3q27) Abnormal-1 green Abnormal-1 orange Dual color (normal)
(split signal) (split signal)
CLL, Multiple myeloma p53 del(17)(p13.1) N/A p53 N/A
CLL, Multiple myeloma D12Z3 +12 N/A Alpha-satellite 12 N/A
(12p11.1-q11.1)
CLL, Multiple myeloma RB1 del(13)(q14.3) N/A RB1 N/A
B-cell lymphoproliferative IgH t(?;14q32) Abnormal-1 green Abnormal-1 orange Dual color (normal)
disorders, including nodal (split signal) (split signal)
marginal zone B-cell
lymphoma and monocytoid
B-celllymphoma
Anaplastic large cell ALK1 t(2;5)(p23.2;q35), Abnormal-1 Abnormal- 1 orange Dual color (normal)
lymphoma# (t2;?) variants green(split signal) (split signal)

* Vysis Inc. Downers Grove, IL.


# Most T-cell monoclonalities are best determined by alternate molecular genetic techniques.
Tabulated by Mary L. Nordberg, PhD.

lymphocytic precursors that express CD10, CD19, and originating from mucosa associated lymphoid tissue
TdT, variably express CD20, usually do not appear in the (MALT), LCL, malignant lymphoma with intravascular
peripheral blood, lack sIg, do not reveal any cytogenetic infiltration, T-cell rich B-cell lymphoma, adult T-cell
or molecular abnormality, and show a characteristic pat- leukemia/lymphoma (Fig. 18), large granular lympho-
tern of antigen expression in flow cytometry histograms. cyte disorders, peripheral T-cell lymphoma, mycosis
Hematogones cannot be positively differentiated from fungoides, anaplastic large cell lymphoma, and hepato-
lymphocytes and lymphoblasts on PET. The list of locus splenic gamma/delta T-cell lymphoma can now be de-
probes that may specifically identify the molecular basis finitively ascertained (103–132). However, because of
of lymphoid lesions in PET (Table 4) is growing. While deficiencies in standardization and the known false nega-
it is unnecessary to apply such technology to cases where tive and false positive rate for PCR-based immunoglob-
histologic appearances and the corroboratory data sup- ulin gene and T-cell receptor assays to determine clonal-
port a diagnosis, these molecular genetic probes are in- ity, it seems unlikely that PCR will replace microscopy
valuable when such data is either sparse or unavailable. to detect lymphoreticular lesions of the bone marrow in
By complimenting light microscopy with the modali- the near future (133–135).
ties mentioned in Table 4, this differential and the fea-
tures characteristic of the indolent and intermediate PLASMA CELLS
grade lesions, i.e., chronic lymphocytic leukemia (Fig.
17), Waldenström’s macroglobulinemia (Fig. 17), pro- Plasma cells are absent in the bone marrow at birth,
lymphocytic leukemia, hairy cell leukemia, malignant but begin to appear during the first few years of infancy.
lymphoma-leukemic phase, splenic lymphoma with vil- In the adult, plasma cells are rare, polyclonal, comprise
lous lymphocytes, follicular lymphoma, mantle cell lym- 3% to 6% of all nucleated cells, and are occasionally
phoma, margin cell lymphoma, malignant lymphoma perivascular in distribution. The normal ratio of kappa:

Advances in Anatomic Pathology, Vol. 10, No. 1, January, 2003


BONE MARROW BIOPSY 19

lambda light chain producing cells is approximately 4:1 neuron-specific enolase, Leu-7, smooth muscle actin,
as evaluated by flow cytometry and IHC (136). Plasma desmin, vimentin, and FVIII related-antigen (143–146).
cells characteristically reveal intracytoplasmic immuno- Semiquantitation of tumor burden as a percentage of
globulin and are CD38 and CD138 positive and CD20 bone marrow cellularity or medullary space involved
negative. Increased numbers of polyclonal plasma cells conveys additional meaning in the pathologic report.
are observed in reactive states. However, when immu-
noglobulin light chain restriction is observed, a plasma RETICULIN AND COLLAGEN
cell dyscrasia such as monoclonal gammopathy of unde- PROLIFERATION
termined significance (MGUS) or multiple myeloma
(Fig. 19) should be considered (136). In such cases, Normally, a delicate filigree of reticulin (type III col-
plasma cells often cluster and demonstrate cytologic lagen) is found to pervade the stroma of the bone mar-
atypia and light chain restriction on kappa and lambda row, encircle fat cells and vascular structures, and sepa-
immunostains in PET (Fig. 20). Several locus probes for rate cellular bone marrow from adjoining bony trabecu-
the prognostication of multiple myeloma in PET are now lae. In core biopsy and aspirate clot sections, reticulin
available (Table 4). However, these may be of practical can be demonstrated with a good Wilder or Jones stain,
value only in controversial or research settings. and is reported as normal or increased. The latter may be
Correlation of plasma cell counts with clinical, radio- graded as mild, moderate, or marked (Fig. 23). Reticulin
logic, and biochemical parameters is of considerable proliferation is usually antecedent to collagen (type I
value. In patients with abnormal lymphoplasmacytic in- collagen) deposition. The latter may be suspected by its
filtrates, Waldenströms macroglobulinemia, heavy chain coarse fiber pattern in H&E sections (Fig. 23), but should
disease, and amyloidosis additionally enter the differen- be confirmed with a trichrome (Fig. 23) or other collagen
tial diagnosis (107,108,137). Plasma cells may occasion- stain. Because of its inaspirable nature, collagen fibrosis
ally reveal flame-like cytoplasmic transformation, bi- is not accurately quantifiable in aspirate clot sections and
nuclearity, and crystalline inclusions (Snapper-Schneid should be evaluated only in the core biopsy.
bodies), features that are better appreciated on aspirate Myelofibrosis may be acute or chronic, and idiopathic
smears (138–140). These changes may be observed in or secondary to myeloproliferative, lymphoproliferative,
both benign and malignant plasma cells. Because the metastatic, or granulomatous disease (147–150). Slowly
lesions of multiple myeloma are focal in the bone mar- progressive fibrosis, as evident in the early stages of
row, some authorities recommend bilateral core biopsies agnogenic myeloid metaplasia and established polycy-
at the time of initial investigation. themia vera, causes sinusoids in the bone marrow to
permanently dilate and develop intravascular hematopoi-
METASTATIC DISEASE esis. These changes may also be observed in the other
A large number of epithelial (Fig. 21) and mesenchy- myeloproliferative disorders. In contrast, rapidly pro-
mal (Fig. 22) tumors metastasize to the bone marrow gressive fibrosis replaces hematopoietic tissue, vascular
(141,142). There is, however, considerable institutional, elements, stroma, and fat, resulting in obliteration of the
biologic, and specimen related variation in the incidence marrow space.
of these lesions. Tumors that commonly metastasize to Grading of bone marrow reticulin and collagen may be
the bone marrow include carcinomas of the breast, pros- quantitated according to the Bauermeister Scale (151)
tate, lung, and gastrointestinal tract in adults, and neuro- (Table 5). The limits of normality lie between grades 0
blastoma in children. Metastatic tumor deposits in the and 2.
bone marrow may be detected in one or all of the speci-
men components submitted for review (21). Occasion- TABLE 5. The Bauermeister scale for grading bone
ally, tumor deposits in the core biopsy are necrotic. Nev- marrow reticulin and collagen
ertheless, tumor cells variably retain their protein epit- Grade Morphologic features
opes and their true nature may be confirmed by their IHC
0 No reticulin fibers demonstrable.
profile. Based on the architectural and cytologic pattern 1 Occasional fine individual fibers and foci of a fine fiber
of the tumor in question, a recommended battery of IHC network.
stains should include but not be limited to cytokeratin, 2 Fine fiber network throughout most of the section. No coarse
fibers present.
epithelial membrane antigen, carcinoembryonic antigen, 3 Diffuse fiber network with scattered thick coarse fibers. No
alpha-fetoprotein, prostate specific antigen, prostatic mature collagen present.
acid phosphatase, HMB-45, S-100, leukocyte common 4 Diffuse and often coarse fiber network with areas of
collagenization.
antigen (CD45), chromogranin, synaptophysin, CD99,

Advances in Anatomic Pathology, Vol. 10, No. 1, January, 2003


20 J. D. COTELINGAM

INFECTIONS AND also the results of a search for pathologic sideroblasts


GRANULOMATOUS CHANGES including ring forms.

A wide range of infections localize to the bone mar- DISORDERS OF THE MACROPHAGE SYSTEM
row (152), and may result in an inflammatory, necrotiz-
ing, or granulomatous response. Intact granulomas are Normally, monocytes and macrophages comprise up
seldom observed in aspirate preparations, and their pres- to 3% of cellular elements in the marrow (Table 2). In
ence cannot be excluded if the aspirate is the only speci- H&E stained sections of PET, monocytes cannot be posi-
men available for evaluation. Granulomatous changes tively identified. Nevertheless, by immunostaining for
(Fig. 24) may be specific or nonspecific (150,153), and CD68, lysozyme, alpha-1-antitrypsin, and alpha-1-
are best defined in the core biopsy and clot section. It is antichymotripsin, infiltrates of this lineage, as in AML-
necessary to work up all epithelioid granulomas with M4, AML-M5, and CMML, can be designated (56). Oc-
acid fast and fungus stains, and correlate changes with casional tingible body macrophages can be encountered
the results of microbial cultures (154). In immunocom- in cellular marrow, and do not connote a pathologic pro-
promised patients, well-defined granulomas may fail to cess. However, an abnormal macrophage component
develop. Under these circumstances, proliferation of mi- may be observed in Gaucher disease (160) (Fig. 29),
crobial-laden macrophages (as in Mycobacterium CML (161,162) (pseudo-Gaucher cells), the hemophago-
avium–intracellulare infection) may be evident (155) cytic syndromes (163), oxalosis (164–166) (Fig. 30),
(Fig. 25). The most frequently encountered granuloma in with thorotrast deposits (167) (Figure 31), sea-blue his-
clinical practice is the nonspecific lipid granuloma (156). tocytosis, and in the acquired and inherited lipidosis
The latter are not associated with hematologic disease, (168). In each of these conditions, correlation with the
and do not require work up with microbial stains. It is clinical picture and related laboratory data is invaluable.
noteworthy that the amastigote forms of leishmaniasis
and the intranuclear viral inclusions of cytomegalovirus STROMAL AND VASCULAR ABNORMALITIES
(Fig. 26), herpes simplex and parvovirus B19 infection Stromal cells and the mesenchyme of the extracellular
(Fig. 27) are readily detected with a high dry lens and matrix are located between fat cells, and surround both
may be confirmed by in situ hybridization if necessary nonhematopoietic and hematopoietic tissue (169). The
(157). latter are decreased in hypoproliferative states, and are
virtually effaced by chemotherapy, an index of adequate
therapeutic induction in acute leukemia. Replacement of
STAINABLE IRON STORES
hematopoietic tissue by mucopolysaccharide with com-
mensurate atrophy of fat cells is observed in serous fat
Quantitation of stainable iron in the bone marrow is atrophy (170). Herein, the mucopolysaccharide can be
most accurately interpreted in Prussian blue preparations delineated with an Alcian Blue stain (Fig.3). It is impor-
of the aspirate smear and clot section. The choice be- tant that the patterns of aplasia, serous fat atrophy, and
tween manual and automated stains (BioGenex Labora- adequate therapeutic induction of the acute leukemias be
tories, San Ramon, CA) is largely parochial and work- so designated.
load related. Decalcification, a prerequisite for prepara- Nutrient arteries, veins, and intervening sinusoids are
tion of the core biopsy, results in leaching out of some also located in the stromal compartment. Lesions that
storage iron. Despite this drawback, evaluation of iron in affect larger vessels, which may be incidentally ob-
the core biopsy is informative in cases where the aspirate served, are those of amyloid infiltration (171) (Fig. 32),
and clot sections were not procured or are dilute and vasculitis, (172) and thrombotic thrombocytopenic pur-
devoid of particles. Assessment on a scale of 0 to 4 plus pura (TTP) (6). It is noteworthy that sinusoidal dilatation
is both simple and reproducible (158,159). In this scale, is associated with increased stromal reticulin and colla-
stainable iron is designated as follows: 0 ⳱ absent, 1 ⳱ gen as in the cellular phase of agnogenic myeloid meta-
trace/decreased, 2 to 3 ⳱ adequate, and 4 ⳱ increased plasia, and is occasionally accompanied by intrasinusoi-
(Fig. 28). In the bone marrow, elemental iron can be dal hematopoiesis (6,173,174) (Fig. 33).
visualized in several compartments, including the cyto-
plasm of macrophages, in developing normoblasts (sid-
DISORDERS OF TRABECULAR BONE
eroblasts), and as free hemosiderin deposits within the
bone marrow stroma. Reports should include descrip- Although a core biopsy may have been procured to
tions of both the quantity and the location of iron, and evaluate hematologic disease, the bony component of

Advances in Anatomic Pathology, Vol. 10, No. 1, January, 2003


BONE MARROW BIOPSY 21

FIG. 25. Mycobacterium avium-intracellulare in the core biopsy of a terminal patient with the acquired immunodeficiency syndrome. (A) H&E
section with marked histiocytic proliferation. (B) Fite stain with numerous acid-fast bacteria within histiocytes (original magnification x 400).
FIG. 26. Cytomegalovirus inclusion. Core biopsy in patient with acquired immunodeficiency syndrome. Nature of inclusion serologically and
immunohistochemically confirmed; H&E section (original magnification x 400) (Courtesy of Dr. Diana Veillon).
FIG. 27. Parvovirus B19 inclusions in early erythroid precursors. Core biopsy. Patient with acquired immunodeficiency syndrome. Nature of
inclusions confirmed by serology and in-situ hybridization; H&E section (original magnification x 400).
FIG. 28. Increased (4+) iron stores. Core biopsy. Patient with anemia of chronic disease; Prussian blue stain (original magnification x 400).
FIG. 29. Gaucher disease. Core biopsy; H&E section (original magnification x 400).
FIG. 30. Oxalosis. Core biopsy; H&E section (original magnification x 400) (Courtesy of Dr. Richard Brunning).
FIG. 31. Thorotrast deposits. Core biopsy; H&E section (original magnification x 400).
FIG. 32. Amyloid deposits. Core biopsy. (A) Congo-red preparation. (B) Birefringent appearances of amyloid with polarized light (original
magnification x 400).
FIG. 33. Intrasinusoidal hematopoiesis. Core biopsy. Patient with agnogenic myeloid metaplasia; H&E stain (original magnification x 400).
FIG. 34. Increased osteoblastic activity in the vicinity of metastatic carcinoma of the prostate. Core biopsy; H&E section (original magnification
x 400).
FIG. 35. Renal osteodystrophy. Core biopsy; H&E section (original magnification x 400).
FIG. 36. Paget disease. Core biopsy; H&E section (original magnification x 400) (Courtesy of Dr. Tuyethoa Vinh).

Advances in Anatomic Pathology, Vol. 10, No. 1, January, 2003


22 J. D. COTELINGAM

this specimen should be routinely assessed. It has been 4. Turkel H, Bethell FH. Biopsy of bone marrow performed by a
new and simple instrument. J Lab Clin Med 1943;28:1246–1251.
estimated that the volume of cortical and trabecular bone 5. Cotelingam JD. Bone marrow interpretation: the science and the
in an iliac crest bone biopsy comprises greater than 20% art. Pathol Case Rev 2000;5:239–251.
of the specimen (175); and despite physiologic variances 6. Foucar K. Bone Marrow Pathology. 2nd ed. Chicago: American
Society of Clinical Pathologists Press; 2001.
related to gender, race, and age, the mean trabecular 7. McFarland W, Damashek W. Biopsy of the bone marrow with the
width is approximately 200 microns (175,176). Cancel- Vim-Silverman needle. JAMA 1958;166:1464–1466.
lous bone of the iliac crest is in dynamic equilibrium 8. Jamshidi K, Swaim WR. Bone marrow biopsy with unaltered
architecture. A new biopsy device. J Lab Clin Med 1971;77:
with the rest of the skeleton, and offers protection to the 2335–2342.
tissues within the labyrinthine marrow space. Trabecular 9. Islam A. Bone marrow biopsy needle with core securing device.
bone is lamellar, and is variably bordered by a slender J Clin Pathol 1982;35:359–364.
10. MananTM bone marrow/aspiration needle MD TechR [package
osteoid seam. Osteoblasts (Fig. 34) and osteoclasts are insert]. Gainsville, FL: Medical Device Technologies, Inc.; 1998.
located along the margins of trabeculae. These cells ef- 11. Anderson G, Coup AJ. Effects of decalcifying agents on the
fectively maintain the structural and physiologic integ- staining of Mycobacterium tuberculosis. J Clin Pathol 1975;28:
744–745.
rity of bone when dispersed in a ratio of approximately 12. Wong FTS, Wu PC. The influence of decalcifying fluids on the
100:1. Variations in this ratio along with changes in the demonstration of Mycobacterium tuberculosis in paraffin sec-
structure of trabecular bone may be observed in previous tions. Med Lab Sciences 1979;36:153–157.
13. Krause JR. Zenker-glacial acidic acid treatment of bone marrow
biopsy sites, renal osteodystrophy (177,178) (Fig. 35), biopsies. In: Bone Marrow Biopsy. New York: Churchhill Liv-
osteopenia, osteomalacia, (179) osteoporosis, (180) ingston; 1981;209.
Paget disease, (181) (Fig. 36) and osteopetrosis (182). 14. Arber DA, Johnson RM, Rainer PA, et al. The bone marrow agar
section: a morphologic and immunohistochemical evaluation.
These lesions should be appropriately described and des- Mod Pathol 1993;6:592–598.
ignated in the pathology report. 15. Islam A. Proposal for a classification of acute myeloid leukemia
based on plastic embedded bone marrow biopsy sections. Leuk
Res 1993;17:421–427.
SUMMARY 16. Burns WA, Yook CR. Plastic sections and ultrastructural tech-
niques in the evaluation of bone marrow pathology. Hematol
Reporting takes many forms, and is understandably Oncol Clin North Am 1988;2:525–536.
17. Breton-Gorius J, Reyes F. Ultrastructure of human bone marrow
parochial. Whatever the choice, accuracy, clarity, brev- cell maturation. Int Rev Cytol 1976;46:251–321.
ity, and timeliness are paramount. A summary comment 18. Ghadially FN. Ultrastruct Pathol of the Cell and Matrix. 3rd ed.
following the microscopic description and diagnostic Boston: Butterworths; 1988.
19. Zucker-Franklin D, Greaves MF, Grossi CE, Marmont AM. Atlas
line which ties together other ancillary aspirate related, of Blood Cells. 2nd ed. Philadelphia: Lea & Febiger; 1988.
cytochemical, flow cytometric, molecular diagnostic, 20. Wickramasinghe SN. Bone marrow. In: Histology for Patholo-
and other clinicopathologic data gleaned during the work gists. 2nd ed. Philadelphia: Lippincott-Raven; 1997:707–742.
21. Barekman CL, Fair KP, Cotelingam JD. Comparative utility of
up conveys depth to the report. This is invaluable for diagnostic bone marrow components: a 10-year study. Am J
purposes of prognostication, teaching, coding, retrieval, Hematol 1997;56:37–41.
and research in keeping with local needs and philoso- 22. Aboul-Nasr R, Estey EH, Kantarjian HM, et al. Comparison of
touch imprints with aspirate smears for evaluating bone marrow
phies. It is our practice to promptly appraise clinical specimens. Am J Clin Pathol 1999;111:753–758.
personnel by telephone of lesions and changes that are 23. Custer RP. An Atlas of the Blood and Bone Marrow. 2nd ed.
unexpected or have immediate therapeutic impact. This Philadelphia: WB Saunders; 1974.
24. Beckstead JH. The bone marrow biopsy: a diagnostic strategy.
brings excellence in communication and service and also Arch Pathol Lab Med 1986;110:175–179.
keeps the pathologist abreast of clinical events. 25. Brynes RK, McKenna RW, Sundberg RD. Bone marrow aspira-
tion and trephine biopsy: an approach to a thorough study. Am J
Clin Pathol 1987;70:753–759.
ACKNOWLEDGMENT 26. Hyun BH, Gulati GL, Ashton JK. Bone marrow examination:
techniques and interpretation. Hematol Oncol Clin N Am 1988;
2:513–523.
The author thanks Ms. Bettie Crisler for secretarial 27. Naeim F. Atlas of Bone Marrow and Blood Pathology. Philadel-
support. phia: WB Saunders; 2001.
28. Li C-Y, Yam LT. Cytochemistry and immunohistochemistry in
hematologic diagnosis. Hematol Oncol Clin North Am 1994;8:
REFERENCES 665–681.
29. Pileri SA, Roncador G, Ceccarelli C, et al. Immunohistochemistry
1. Neumann E. Uber die Bedetung des Knochenmarks fur die Blut- of bone-marrow biopsy. Leuk Lymphoma 1997;26 (Suppl
building. Centralblatt Med Wiss 1868;6:689. 1):69–75.
2. Bizzozero G. Sulla fungione ematopoietica del midollo delle 30. Erber WN, Willis JI, Hoffman GJ. An enhanced immunohisto-
ossa. Zentralbl Med Wissensch 1868;6:885. chemical method for staining bone marrow trephine sections. J
3. Mosler F. Klinische Symptome und Therapie der Medullalären Clin Pathol 1997;50:389–393.
Leukemi. Berl Klin Wochenschr 1876;13:233. 31. van der Valk P, Mullink H, Huijens PC, et al. Immunohistochem-

Advances in Anatomic Pathology, Vol. 10, No. 1, January, 2003


BONE MARROW BIOPSY 23

istry in bone marrow diagnosis. Value of a panel of monoclonal 53. Pinkus GS, Pinkus JL. Myeloperoxidase: a specific marker for
antibodies on routinely processed bone marrow biopsies. Am J myeloid cells in paraffin sections. Mod Pathol 1991;4:733–741.
Surg Pathol 1989;13:97–106. 54. Horny HP, Campbell M, Steinke B, et al. Acute myeloid leuke-
32. D’Onofrio G, Zini G, Tommasi M, et al. Automated analysis of mia: immunohistochemical findings in paraffin-embedded bone
bone marrow: routine implementation and differences from pe- marrow specimens. Hum Pathol 1990;21:648–655.
ripheral blood. Lab Hematol 1998;4:71–79. 55. Dunphy CH, Polski JM, Evans HL, et al. Evaluation of bone
33. Fan G, Alvares C, Ismail S, et al. Quantitative and qualitative marrow specimens with acute myelogenous leukemia for CD34,
bone marrow analysis by the Cell-Dyn 4000 Hematologic Ana- CD15, CD117, and myeloperoxidase. Arch Pathol Lab Med
lyzer. Mod Pathol 1999;12:190A. 2001;125:1063–1069.
34. Vos JA, Simurdak JH, Davis BJ, et al. Vortex disaggregation for 56. Warnke RA, Pulford KA, Pallesen G, et al. Diagnosis of myelo-
flow cytometry: a tissue preserving method. Mod Pathol 1999; monocytic and macrophage neoplasms in routinely processed tis-
12:147A. sue biopsies with monoclonal antibody KP1. Am J Pathol 1989;
35. McCoy J, Keren DF. Current practices in clinical flow cytometry: 135:1089–1095.
a practice survey by the American Society of Clinical Patholo- 57. Horny HP, Parwaresch MR, Lennert K. Bone marrow findings in
gists. Am J Clin Pathol 1999;111:156–160. systemic mastocytosis. Hum Pathol 1985;16:808–814.
36. Willman CL. Acute leukemias: a paradigm for the integration of 58. Li WV, Kapadia SB, Sonmez-Alpan E, et al. Immunohistochem-
new technologies in diagnosis and classification. Mod Pathol ical characterization of mast cell disease in paraffin sections using
1999;12:218–228. tryptase, CD68, myeloperoxidase, lysozyme, and CD20 antibod-
37. Hartsock R, Smith EB, Petty CS. Normal variations with aging in ies. Mod Pathol 1996;9:982–988.
the amount of hematopoietic tissue in bone marrow from the 59. Horny HP, Sillaber C, Menke D, et al. Diagnostic utility of stain-
anterior iliac crest. Am J Clin Pathol 1965;43:326–331. ing for tryptase in patients with mastocytosis. Am J Surg Pathol
38. Gruppo RA, Lampkin BC, Granger S. Bone marrow cellularity 1998;22:1132–1140.
determination: comparison of the biopsy, aspirate, and buffy coat. 60. Natkunam Y, Rouse RV. Utility of paraffin section immunohis-
Blood 1977;49:29–31. tochemistry for C-KIT (CD117) in the differential diagnosis of
39. Fong TP, Okafor LA, Schmitz TH, et al. An evaluation of cellu- systemic mast cell disease involving the bone marrow. Am J Surg
larity in various types of bone marrow specimens. Am J Clin Pathol 2000;24:81–91.
61. Hanson CA, Abaza M, Sheldon S, et al. Acute biphenotypic
Pathol 1979;72:812–816.
leukemia: immunophenotype and cytogenetic analysis. Br J Hae-
40. Vaquez MH, Aubertin C. L’anémie pernicieuse d’après concep-
matol 1993;84:49–60.
tions actuelles. Bull Mem Soc Med Hop Paris 1904;21:288.
62. Gaffney RL, Lowrey MC, Sciotto CG. Acute lymphoblastic leu-
41. Osgood EE, Seaman AJ. The cellular composition of bone mar-
kemia relapsing as acute myelogenous leukemia: the role of the
row as obtained by sternal puncture. Physiol Rev 1939;24:105– mixed lineage leukemia (MLL) gene. ASCP Check Sample HP-
114. 99, 1999.
42. Wickramasinghe SN. Normal hematopoiesis. Cellular composi- 63. Mangi MH, Salisbury JR, Mufti GJ. Abnormal localization of
tion of normal bone marrow. In: Blood and Bone Marrow. Sys- immature precursors in the bone marrow of myelodysplastic syn-
temic Pathology. 3rd ed. Edinburgh: Churchill Livingstone; dromes: current state of knowledge and future directions. Leuk
1991;70. Res 1991;15:627–639.
43. Perkins SL. Examination of blood and bone marrow. In: Lee GR, 64. Heaney ML, Golde DW. Myelodysplasia. N Engl J Med 1999;
Foerster J, Lukens J, Paraskevas F, Greer JP, Rodgers GM, eds. 340:1649–1660.
Wintrobe’s Clinical Hematology. 10th ed. Baltimore: Williams & 65. Reddey VB. Topics in bone marrow biopsy pathology: role of
Wilkins; 1999:1;9. marrow topography in myelodysplastic syndromes and evaluation
44. Clark P, Normansell DE, Innes DJ, et al. Lymphocyte subsets in of posttreatment and postbone marrow transplant biopsies. Ann
normal bone marrow. Blood 1986;67:1600–1606. Diagn Pathol 2001;5:110–120.
45. Horny HP, Wehrmann M, Griesser H, et al. Investigation of bone 66. Orazi A, Albitar M, Heerema NA, et al. Hypoplastic myelodys-
marrow lymphocyte subsets in normal, reactive, and neoplastic plastic syndromes can be distinguished from acquired aplastic
states using paraffin-embedded biopsy specimens. Am J Clin anemia by CD34 and PCNA immunostaining of bone marrow
Pathol 1993;99:142–149. biopsy specimens. Am J Clin Pathol 1997;107:268–274.
46. O’Donnell LR, Alder SL, Balis UJ, et al. Immunohistochemical 67. Brunning R. The proposed WHO classification of MDS and
reference ranges for B lymphocytes in bone marrow biopsy par- MDS-related acute leukemias. Mod Pathol 1999;12:102.
affin sections. Am J Clin Pathol 1995;104:517–523. 68. Vardiman JW. Myelodysplastic/myeloproliferative disorders.
47. Algino KM, Thomason RW, King DE, et al. CD20 (Pan-B cell Mod Pathol 1999;12:104–105.
antigen) expression on bone marrow derived T-cells. Am J Clin 69. Jaffe ES, Harris NL, Stein H, et al. Tumors of Hematopoietic and
Pathol 1996;106:78–81. Lymphoid Tissue (WHO Classification of Tumors: Pathology and
48. Sternberg PA, Hill RJ. Platelets and megakaryocytes. In: Lee GR, Genetics). Lyon: IARC Press; 2001.
Foerster J, Lukens J, Paraskevas F, Greer JP, Rodgers GM, eds. 70. van Der Valden VH, te Marvelde JG, Hoogeveen PG, et al. Tar-
Wintrobe’s Clinical Hematology. 10th ed. Baltimore: Williams & geting of the CD33-calicheamicin immunoconjugate Myelotarg
Wilkins; 1999:1;615–660. (CMA-676) in acute myeloid leukemia: in vivo and in vitro satu-
49. Wolf BC, Brady K, O’Murchadha MT, et al. An evaluation of ration internalization by leukemic and normal myeloid cells.
immunohistologic stains for immunoglobulin light chains in bone Blood 2001;97:3197–3204.
marrow biopsies in benign and malignant plasma cell prolifera- 71. Druker BJ, Talpaz M, Resta D, et al. Efficacy and safety of a
tions. Am J Clin Pathol 1990;94:742–746. specific inhibitor of the BCR-ABL tyrosine kinase in chronic
50. Harada H, Asaoku H, Kuramoto A, et al. Phenotypic difference of myeloid leukemia. N Eng J Med 2001;334:1031–1037.
normal plasma cells from mature myeloma cells. Blood 1993;81: 72. Hasserjian RP, Boecklin F, Parker S, et al. ST1571 (Imatinib
2658–2663. Mesylate) reduces bone marrow cellularity and normalizes mor-
51. Oski FA, Naiman JL. The bone marrow. In: Oski FA, Naiman JL, phologic features irrespective of cytogenetic response. Am J Clin
eds. Hematologic Problems in the Newborn. 3rd ed. Philadelphia: Pathol 2002;117:360–367.
WB Saunders Co; 1982;20. 73. Applebaum FR, Downing J, Willman C. The biology and therapy
52. Naeim F. Pathology of Bone Marrow. 2nd ed. Baltimore: Wil- of acute myelogenous leukemia. Molecular detection and moni-
liams & Wilkins; 1997. toring of AML 1/ETO transcripts in AML with t(8;21). In: Kaus-

Advances in Anatomic Pathology, Vol. 10, No. 1, January, 2003


24 J. D. COTELINGAM

hansky K, McArthur JR, eds. Hematology Education Program, initially in the bone marrow of patients infected with the human
American Society of Hematology. Seattle: Smith, Bucklin & As- immunodeficiency virus. Cancer 1993;71:1235–1238.
sociates; 1995;23–35. 97. Naughton MJ, Hess JL, Zutter MM, et al. Bone marrow staging
74. Kowal-Vern A, Cotelingam JD, Schumacher HR. The prognostic in patients with nonHodgkin’s lymphoma. Is flow cytometry a
significance of pronormoblasts in acute leukemia. Am J Clin useful test? Cancer 1998;82:1154–1159.
Pathol 1992;98:34–40. 98. Munker R, Hasencleaver D, Brosteanu O, et al. Bone marrow
75. Mazzella FM, Cotelingam JD, Kowal-Vern A, et al. Proposed involvement in Hodgkin’s disease: an analysis of 135 consecutive
WHO classification of acute myeloid leukemia and myelodys- cases. J Clin Oncol 1995;13:403–409.
plastic syndromes. Mod Pathol 2000;13:101–102. 99. Schumacher HR. Acute Leukemia: Difficult, Controversial and
76. Strauchen JA. B19 Parvovirus infection. In: Diagnostic Histopa- Automated Analysis. Baltimore: Williams & Wilkins; 1997.
thology of the Bone Marrow. New York: Oxford University 100. Orazi A, Cotton J, Cattoretti G, et al. Terminal deoxynucleotidyl
Press; 1996;282–285. transferase staining in acute leukemia and normal bone marrow in
77. Lee GR. Congenital dyserythropoietic anemias. In: Lee GR, routinely processed paraffin sections. Am J Clin Pathol 1994;102:
Foerster J, Lukens J, Paraskevas F, Greer JP, Rodgers GM, eds. 640–645.
Wintrobe’s Clinical Hematology. 10th ed. Baltimore: Williams & 101. Kheiri SA, MacKerrell T, Bonagura VR, et al. Flow cytometry
Wilkins; 1999:2;1489–1496. with or without cytochemistry for the diagnosis of acute leuke-
78. Mazzela FM, Kowal-Vern A, Shrit A, et al. Erythroleukemia. mias. Cytometry 1998;34:82–86.
Evaluation of 48 cases with reference to classification, cell pro- 102. Davis RE, Longacre TA, Cornbleet PJ. Hematogones in the bone
liferation, cytogenetics and prognosis. Am J Clin Pathol 1998; marrow of adults. Immunophenotypic features, clinical settings
110:590–598. and differential diagnosis. Am J Clin Pathol 1994;102:202–211.
79. Crocker J, Gyde O, Jenkins R. Demonstration of normoblasts in 103. Foucar K. Chronic lymphoid leukemias and lymphoproliferative
tissue sections by means of an immunohistochemical technique disorders. Mod Pathol 1999;12:141–150.
for haemoglobin. J Clin Pathol 1984;37:1312–1313. 104. Gala J-L, Guiot Y, Delannoy A, et al. Use of image analysis and
80. Sternberg PA. Hill RJ. Platelets and megakaryocytes. In: Lee GR, immunostaining of bone marrow trephine biopsy specimens to
Foerster J, Lukens J, Paraskevas F, Greer JP, Rodgers GM, eds. quantify residual disease in patients with B-cell chronic lympho-
Wintrobe’s Clinical Hematology. 10th ed. Baltimore: Williams & cytic leukemia. Mod Pathol 1999;12:391–399.
Wilkins; 1999:1;615–660.
105. Shapiro JL, Miller ML, Pohlman B, et al. CD5− B-cell lympho-
81. Innes DJ, Mills SE, Walker GK. Megakaryocytic leukemia: iden-
proliferative disorders presenting in blood and bone marrow. A
tification using antifactor VIII immunoperoxidase. Am J Clin
clinicopathologic study of 40 patients. Am J Clin Pathol 1999;
Pathol 1982;77:107–110.
111:477–487.
82. Calapso P, Vitarelli E, Crisafulli C, et al. Immunocytochemical
106. Tworek JA, Singleton TP, Schnitzer B, et al. Flow cytometric and
detection of megakaryocytes by endothelial markers: a compara-
immunohistochemical analysis of small lymphocytic lymphoma,
tive study. Pathologica 1992;84:215–223.
mantle cell lymphoma, and plasmacytoid small lymphocytic lym-
83. Chuang SS, Yung YC, Li CY. Von Willebrand factor is the most
phoma. Am J Clin Pathol 1998;110:582–589.
reliable immunohistochemical marker for megakaryocytes of my-
elodysplastic syndrome and chronic myeloproliferative disorders. 107. Andriko JA, Aguilera NS, Chu WS, et al. Waldenström’s mac-
Am J Clin Pathol 2000;113:506–511. roglobulinemia: a clinicopathologic study of 22 cases. Cancer
84. Huang MJ, Li CY, Nichols WL, et al. Acute leukemia with mega- 1997;80:1926–1935.
karyocytic differentiation: a study of 12 cases identified immu- 108. Owen RG, Barrans SL, Richards SJ, et al. Waldenströms macro-
nocytochemically. Blood 1984;64:427–439. globulinemia: development of diagnostic criteria and identifica-
85. San Miguel JF, Gonzalez M, Canizo MC, et al. Leukemias with tion of prognostic factors. Am J Clin Pathol 2001;116:420–428.
megakaryoblastic involvement: clinical, hematologic, and immu- 109. Schlette EJ, Buesco-Ramos CE, Medeiros LJ. B-cell prolympho-
nologic characteristics. Blood 1988;72:402–407. cytic leukemia. Pathol Case Rev 2000;5:274–280.
86. Ivanyi JL, Kiss A, Telek B, et al. Megakaryocyte markers in 110. Schumacher HR, Cotelingam JD. T-Prolymphocytic leukemia.
myeloproliferative disorders. Acta Histochem 1993;95:79–88. In: Chronic Leukemia: Approach to Diagnosis. New York: Igaku-
87. Jacobson S, Wadenvik H, Kutti J, et al. Low megakaryocyte Shoin; 1993;269–280.
ploidy in Ph+ chronic myeloid leukemia measured by flow cy- 111. Hounieu H, Chittal SM, AL Saati T, et al. Hairy cell leukemia.
tometry. Am J Clin Pathol 1999;111:185–190. Diagnosis of bone marrow involvement in paraffin-embedded
88. Boultwood J, Lewis S, Waincoat JS. The 5q− syndrome. Blood sections with monoclonal antibody DBA 44. Am J Clin Pathol
1994;84:3253–3260. 1992;98:26–33.
89. van den Berghe H, Michaux L. 5q−, twenty five years later:a 112. Pittaluga S, Tierens A, Dodoo YL, et al. How reliable is histo-
synopsis. Cancer Genet Cytogenet 1997;94:1–7. logic examination of bone marrow trephine biopsy specimens for
90. Rosse C, Kraemer MJ, Dillon TL, et al. Bone marrow cell popu- the staging of nonHodgkin’s lymphoma? A study of hairy cell
lations of normal infants: the predominance of lymphocytes. J leukemia and mantle cell lymphoma involvement of the bone
Lab Clin Med 1977;89:1225–1240. marrow trephine specimen by histologic immunohistochemical,
91. Farhi DC. Germinal centers in bone marrow. Hematol Pathol and polymerase chain reaction techniques. Am J Clin Pathol
1989;3:133–136. 1999;111:179–184.
92. Lambertenghi-Deliliers G, Annaloro C, Soligo D, et al. Incidence 113. Troussard X, Valensi F, Duchayne E, et al. Splenic lymphoma
and histologic features of bone marrow involvement in malignant with villous lymphocytes: clinical presentation, biology and prog-
lymphomas. Ann Hematol 1992;65:61–65. nostic factors in a series of 100 patients. Br J Haematol 1996;
93. Ben-Ezra JM, King BE, Harris AC, et al. Staining for Bcl-2 93:731–736.
protein helps to distinguish benign from malignant lymphoid ag- 114. McCluggage WG, Bharucha H, el-Agnaf M, et al. B-cell signet
gregates in bone marrow biopsies. Mod Pathol 1994;7:560–564. ring cell lymphoma of bone marrow. J Clin Pathol 1995;48:275–
94. O’Carroll DI, McKenna RW, Brunning RD. Bone marrow mani- 278.
festations of Hodgkin’s disease. Cancer 1976;38:1717–1728. 115. Vasef MA, Medeiros LJ, Koo C, et al. Cyclin D1 immunohisto-
95. Bartl R, Frisch B, Burnhardt R, et al. Assessment of bone marrow chemical staining is useful in distinguishing mantle cell lym-
histology in Hodgkin’s disease: correlation with clinical factors. phoma from other low grade B-cell neoplasms in bone marrow.
Br J Haematol 1982;51:345–360. Am J Clin Pathol 1997;108:302–307.
96. Karcher DS. Clinically unsuspected Hodgkin disease presenting 116. Kent SA, Variakojis D, Peterson LC. Comparative study of mar-

Advances in Anatomic Pathology, Vol. 10, No. 1, January, 2003


BONE MARROW BIOPSY 25

gin zone lymphoma involving bone marrow. Am J Clin Pathol marrow sections, smears, and immunohistologic staining for im-
2002;117:698–708. munoglobulin light chains in the diagnosis of benign and malig-
117. Traweek ST, Sheibani K. Monocytoid B-cell lymphoma with nant plasma cell proliferation. Histopathology 1993;22:423–428.
bone marrow and peripheral blood involvement at presentation. 139. Snapper J, Schneid B. On the influence of stilbamidine upon
Am J Clin Pathol 1990;94:117–118. myeloma cells. Blood 1946;1:534–536.
118. Hodges GF, Lenhardt TM, Cotelingam JD. Bone marrow in- 140. Roberts GT, Khalil SH. Multiple myeloma with bone marrow
volvement in large cell lymphoma. Prognostic implications of crystal deposition and marrow fibrosis. Ann Saudi Med 1998;18:
discordant disease. Am J Clin Pathol 1994;101:305–311. 432–436.
119. Jones D. Large B-cell lymphoma in the bone marrow. Pathol 141. Singh G, Krause JR, Breitfeld V. Bone marrow examination for
Case Rev 2000;5:281–286. metastatic tumor: aspirate and biopsy. Cancer 1977;40:2317–
120. Franco V. Florena AM, Campesi G. Intrasinusoidal bone marrow 2321.
infiltration: a possible hallmark of splenic lymphoma. Histopa- 142. Jatoi A, Dallal GE, Nguyen PL. False negative rates of tumor
thology 1996;29:571–575. metastases in the histologic examination of bone marrow. Mod
121. Tucker TJ, Bardales RH, Miranda RN. Intravascular lymphoma- Pathol 1999;12:29–32.
tosis with bone marrow involvement. Arch Pathol Lab Med 1999; 143. Mansi JL, Berger U, Wilson P, et al. Detection of tumor cells in
123:952–956. bone marrow of patients with prostatic carcinoma by immunocy-
122. Khalidi HS, Brynes RK, Browne P, et al. Intravascular large tochemical techniques. J Urol 1988;139:545–548.
B-cell lymphoma: the CD5 antigen is expressed by a subset of 144. Lazda EJ, Berry PJ. Bone marrow metastasis in Ewing’s sarcoma
cases. Mod Pathol 1998;11:983–988. and peripheral primitive neuroectodermal tumor: an immunohis-
123. Estalilla OC, Koo CH, Brynes RK, et al. Intravascular large B- tochemical study. Pediatric & Developmental Pathol 1998;1:
cell lymphoma: a report of five cases initially diagnosed by bone 125–130.
marrow biopsy. Am J Clin Pathol 1999;112:255. 145. Mathieu MC, Friedman S, Bosq J, et al. Immunohistochemical
124. Skinnider BF, Connors JM, Gascoyne RD. Bone marrow involve- staining of bone marrow biopsies for detection of occult metas-
ment in T-cell rich B-cell lymphoma. Am J Clin Pathol 1997; tasis in breast cancer. Breast Cancer Res Treat 1990;15:21–26.
108:570–578. 146. Dabbs DJ. Diagnostic Immunohistochemistry. New York: Chur-
125. Ogata M, Ogata Y, Kohno K, et al. Eosinophilia associated with chill Livingstone; 2002.
adult T-cell leukemia: role of interleukin-5 and granulocyte- 147. Hruban RH, Kuhajda FP, Mann RB. Acute myelofibrosis. Immu-
macrophage colony-stimulating factor. Am J Hematol 1995;59: nohistochemical study of four cases and comparison with acute
242–245. megakaryoblastic leukemia. Am J Clin Pathol 1987;88:578–588.
126. Evans HL, Burks E, Viswantatha D, et al. Utility of immunohis- 148. Rywlin AM. Myelofibrosis and osteosclerosis. In: Histopathol-
tochemistry in bone marrow evaluation of T-lineage large granu- ogy of the Bone Marrow. Boston; Little Brown & Co.; 1976;153.
lar lymphocyte leukemia. Hum Pathol 2000;31:1266–1273. 149. Burthem J, Cawley JC. Bone marrow fibrosis of hairy cell leu-
127. Hanson CA, Brunning RD, Gajl-Peczalska KJ, et al. Bone mar- kemia is caused by synthesis and assembly of fibronectin matrix
row manifestations of peripheral T-cell lymphoma. A study of 30 by hairy cells. Blood 1994;15:479–504.
cases. Am J Clin Pathol 1986;86:449–460. 150. Bhargava V, Farhi DC. Bone marrow granulomas. Clinicopath-
128. Gaulard P, Kanavaros P, Farcet JP, et al. Bone marrow histologic ologic findings in 72 cases and review of the literature. Hematol
and immunohistochemical findings in peripheral T-cell lym- Pathol 1988;2:43–50.
phoma: a study of 38 cases. Hum Pathol 1991;22:331–338. 151. Bauermeister DE. Quantitation of bone marrow reticulin: a nor-
129. Graham SJ, Sharpe RW, Steinberg SM, et al. The prognostic mal range. Am J Clin Pathol 1971;56:24–31.
implications of a bone marrow histopathologic classification sys- 152. Diebold J, Molina T, Camilleri-Broët S, et al. Bone marrow mani-
tem in mycosis fungoides and the Sézary syndrome. Cancer festations of infectious and systemic diseases observed in bone
1993;72:726–734. marrow trephine biopsy. Histopathology 2000;37:199–211.
130. Salhany KE, Greer JP, Cousar JB, et al. Marrow involvement in 153. Tefferi A, Li CY. Bone marrow granulomas associated with
cutaneous T-cell lymphoma. Am J Clin Pathol 1989:92;747–754. chronic natural killer cell lymphocytosis. Am J Hematol 1997;54:
131. Fraga M, Brosset P, Schaifer D, et al. Bone marrow involvement 258–262.
in anaplastic large cell lymphoma. Immunohistochemical detec- 154. Volk EE, Miller ML, Kirkley BA, et al. The diagnostic usefulness
tion of minimal disease and it’s prognostic significance. Am J of bone marrow cultures in patients with fever of unknown origin.
Clin Pathol 1995;103:82–89. Am J Clin Pathol 1998;110:150–153.
132. Vega F, Medeiros LJ, Bueso-Ramos C, et al. Hepatosplenic 155. Farhi DC, Mason III UG, Horsburgh Jr CR. The bone marrow in
gamma/delta T-cell lymphoma in bone marrow. A sinusoidal neo- disseminated MAI infection. Am J Clin Pathol 1985;83:463–468.
plasm with blastic cytologic features. Am J Clin Pathol 2001; 156. Rywlin AM, Ortega R. Lipid granulomas of the bone marrow. Am
116:410–419. J Clin Pathol 1972;57:457–462.
133. Coad JE, Olson DJ, Christensen DR, et al. Correlation of PCR- 157. Conner DH, Chandler FW, Schwartz DA, et al. Pathology of
detected clonal gene rearrangements with bone marrow morphol- Infectious Disease. Stamford: Appleton and Lange; 1997:1;255.
ogy in patients with B-lineage lymphoma. Am J Surg Pathol 158. Strauchen JA. Interpretation of iron stores in the bone marrow. In:
1997;21:1047–1056. Diagnostic Histopathology of the Bone Marrow. Oxford: Oxford
134. Medeiros LJ, Carr J. Overview of the role of molecular methods University Press; 1996;16.
in the diagnosis of malignant lymphoma. Arch Path Lab Med 159. Rywlin AM. Bone marrow hemosiderin. In: Histopathology of
1999;123:1189–1207. the Bone Marrow. Boston: Little Brown & Co.; 1976;53–58.
135. Fader S, Kurzrock R, Estrov Z. Minimal residual disease in he- 160. Rector JT, Cotelingam JD. Gaucher’s disease. ASCP Check Path
matologic disorders. Arch Pathol Lab Med 1999;123:1030–1034. (Hematopathology) 1996; QAH 96–4.
136. Wolf BC, Brady K, O’Murchadha MT, et al. An evaluation of 161. Lee RE, Ellis LD. The storage cells of chronic myeloid leukemia.
immunohistologic stains for immunoglobulin light chains in bone Lab Invest 1971;24:261–264.
marrow biopsies in benign and malignant plasma cell prolifera- 162. Ulirsch RC. Sea Blue histiocytes in chronic myeloid leukemia.
tions. Am J Clin Pathol 1990;94:742–746. ASCP Check Sample 1985; H-4.
137. Harada H, Asaoku H, Kuramoto A, et al. Phenotypic difference of 163. Brunning RD, McKenna RW. Infection associated hemophago-
normal plasma cells from mature myeloma cells. Blood 1993;81: cytic syndrome. In: Rosai J, Sobin LH, eds. Atlas of Tumor Pa-
2658–2663. thology, Bone Marrow. AFIP Fasicle. Washington DC; Armed
138. Thiry A, Delvenne P, Fontaine MA, et al. Comparison of bone Forces Institute of Pathology; 1994;9:447–452.

Advances in Anatomic Pathology, Vol. 10, No. 1, January, 2003


26 J. D. COTELINGAM

164. McKenna RW, Dehner LP. Oxalosis: an unusual cause of my- syndrome: a review of literature and histomorphology. Adv Biosci
elophthisis in childhood. Am J Clin Pathol 1976;66:991–997. 1975;16:9–56.
165. Boquist L, Lindquist B, Ostberg Y, et al. Primary oxalosis. Am J 174. Wolf BC, Neiman RS. Myelofibrosis with myeloid metaplasia:
Med 1973;54:673–681. pathophysiologic implications of the correlation between bone
166. Gherardi G, Poggi A, Sisca S, et al. Bone oxalosis and renal marrow changes and progression of splenomegaly. Blood 1985;
osteodystrophy. Arch Pathol Lab Med 1980;104:105–111. 65:803–809.
167. Graham SJ, Heaton RB, Garvin DF, Cotelingam JD. Whole body 175. Vigorita VJ. The bone biopsy protocol for evaluating osteoporo-
pathologic analysis of a patient with thorotrast induced myelo- sis and osteomalacia. Am J Surg Pathol 1984;8:925–930.
dysplasia. Health Phys 1992;63:20–26. 176. Frisch B, Bartl R. Biopsy Interpretation of Bone and Bone Mar-
168. Kolodny EH, Tenembaum AL. Bone marrow involvement in stor- row. London: Arnold-Oxford University Press; 1999;27.
age diseases. In: Nathan DG, Orkin SH, eds. Nathan and Oski’s
177. Teitelbaum SL. Renal osteodystrophy. Human Pathol 1984;15:
Hematology of Infancy and Childhood. 4th ed. Philadelphia: WB 306–323.
Saunders Co; 1992;1452–1492.
178. Wittels B. Renal osteodystrophy. In: Bennington JL, ed. Surgical
169. Wilkins BS, Jones DB. Immunophenotypic characterization of
Pathology of Bone Marrow: Core Biopsy Diagnosis. Philadel-
stromal cells in aspirated human bone marrow samples. Exp He-
phia: WB Saunders Co; 1985;133–136.
matol 1998;26:1061–1067.
170. Mehta K, Gascon P, Robboy S. The gelatinous bone marrow 179. Bain BJ, Clark DM, Lampert IA. Osteomalacia. In: Bone Marrow
(serous atrophy) in patients with AIDS, evidence of excess sul- Pathology. London: Blackwell Scientific Publications; 1993;258–
phated glycosaminoglycan. Arch Pathol Lab Med 1992;116:504– 259.
508. 180. Becks JS, Nordin BEC. Histological assessment of osteoporosis
171. Wolf BC, Kumar A, Vera JC, et al. Bone marrow morphology by the iliac crest biopsy. J Pathol Bacteriol 1960;80:391–397.
and immunology in systemic amyloidosis. Am J Clin Pathol 181. Frisch B, Lewis SM, Burkhardt R, et al. Paget’s disease of bone.
1986;86:84–88. In: Biopsy Pathology of Bone and Bone Marrow. New York:
172. Rosenthal NS, Farhi DC. Bone marrow findings in connective Raven Press; 1985;97–100.
tissue disease. Am J Clin Pathol 1989;92:650–654. 182. Milgram JW, Jasty M. Osteopetrosis. A morphologic study of
173. Burkhardt R, Bartl R, Beil E, et al. Myelofibrosis-osteosclerosis twenty-one cases. J Bone Joint Surg Am 1982;64:912–929.

Advances in Anatomic Pathology, Vol. 10, No. 1, January, 2003

Das könnte Ihnen auch gefallen