You are on page 1of 19

International Journal of Laboratory Hematology

The Official journal of the International Society for Laboratory Hematology

REVIEW INTERNAT IONAL JOURNAL OF LABORATO RY HEMATO LOGY

ICSH guidelines for the standardization of bone marrow


immunohistochemistry
E. E. TORLAKOVIC*, R. K. BRYNES † , E. HYJEK ‡ , S.-H. LEE § , H. KREIPE ¶ , M. KREMER**, R. MCKENNA † † ,
Y. SADAHIRA ‡ ‡ , A. TZANKOV § § , M. REIS ¶ ¶ , A. PORWIT* , ***, FOR THE INTERNATIONAL COUNCIL FOR
STANDARDIZATION IN HAEMATOLOGY

*Department of Laboratory S U M M A RY
Hematology, University Health
Network, University of Toronto, Bone marrow (BM) tissue biopsy evaluation, including trephine
Toronto, ON, Canada biopsy and clot section, is an integral part of BM investigation and

Department of Pathology, Keck
School of Medicine, University is often followed by ancillary studies, in particular immunohisto-
of Southern California, Los chemistry (IHC). IHC provides in situ coupling of morphological
Angeles, CA, USA assessment and immunophenotype. The number of different IHC

Department of Pathology,
tests that can be applied to BM trephine biopsies and the number
University of Chicago, Chicago,
IL, USA of indications for IHC testing is increasing concurrently with the
§
Department of Haematology, St development of flow cytometry and molecular diagnostic methods.
George Hospital, SEALS Central, An international Working Party for the Standardization of Bone
Sydney, NSW, Australia

Department of Pathology,
Marrow IHC was formed by the International Council for Standard-
Hannover Medical School, ization in Hematology (ICSH) to prepare a set of guidelines for the
Hannover, Germany standardization of BM IHC based on currently available published
**Munich Municipal Hospital, evidence and modern understanding of quality assurance principles
Institute of Pathology, Munich,
Germany as applied to IHC in general. The guidelines were discussed at the
††
Special Hematology, ICSH General Assemblies and reviewed by an international panel
Department of Laboratory of experts to achieve further consensus and represent further
Medicine and Pathology,
development of the previously published ICSH guidelines for the
University of Minnesota,
Minneapolis, MN, USA standardization of BM specimens handling and reports.
‡‡
Department of Pathology,
Kawasaki Medical School,
Kurashiki, Japan
§§
Institute of Pathology,
University Hospital Basel, Basel,
Switzerland
¶¶
Department of Clinical
Pathology, Sunnybrook Health
Sciences Centre, Toronto, ON,
Canada
***Department of Pathology,
Karolinska Institute, Stockholm,
Sweden

© 2015 John Wiley & Sons Ltd, Int. Jnl. Lab. Hem. 2015, 37, 431–449 431
432 E. E. TORLAKOVIC ET AL. | STANDARDIZATION OF BONE MARROW IMMUNOHISTOCHEMISTRY

Correspondence:
Emina Emilia Torlakovic, Table of Contents
Department of Laboratory 1. Background information, rationale, and aims
Hematology, University Health 2. Recommendations
Network, University of Toronto, 2.1. IHC test classification
11th Floor/R411, 200 Elizabeth 2.2. Pre-analytical standards
Street, Toronto, ON M5G 2C4, 2.3. Analytical standards
Canada. 2.4. Postanalytical standards
Tel.: +1 416 340 4932; 2.5. Quality assurance standards
Fax: +1 416 340 5543; 3. Special considerations for pre-analytical phase
E-mail: emina.torlakovic@uhn.ca 4. Special considerations for analytical phase
5. Special considerations for postanalytical phase
6. Special considerations for quality assurance
doi:10.1111/ijlh.12365

Received 14 November 2014;


accepted for publication 2
March 2015

Keywords
Standardization, immunohisto-
chemistry, bone marrow, tre-
phine biopsy

for external quality assurance/proficiency testing


1 . BAC K G R O U N D I N F O R M AT I O N,
(EQA/PT) often coupled to requirements for labora-
R AT I O N A L E , A N D A I M S
tory accreditation. However, bone marrow (BM)
Immunohistochemistry (IHC) standards have been IHC, as applied to BM core biopsy specimens and
evolving during the last few decades as the clinical BM aspirate clot sections, is largely unaffected by
needs for standardization for IHC laboratory testing this trend. In contrast to anatomic pathology, where
have been rising. In particular, IHC testing that almost all laboratories fix tissues in formalin (most
provides prognostic and predictive information often in 10% buffered formaldehyde), many labora-
important for stratification of patients for specific tories have developed their unique protocols for tis-
therapies is at the forefront of IHC standardization sue processing of BM core biopsies as well as
[1–5]. Although no systematic studies are available aspirate clot preparations [8–12]. Pre-analytical con-
to date, it is generally assumed to be correct that ditions are critical for the IHC outcomes. As long as
false IHC results, related to technical and interpreta- we cannot standardize pre-analytical conditions,
tive pitfalls, may lead to incorrect diagnosis [4, 6]. complete standardization of BM IHC will continue
The risks to patient safety are lower when IHC tests’ to be impossible. The use of different fixatives,
results are interpreted as a part of the panel and in unique and variable fixation times, as well as the
conjunction with other information (clinical history, use of various decalcifying reagents and variation in
morphology, flow cytometry, biochemical testing, time for decalcification multiply this problem in BM
etc.) [7]. The need for standardization has been fur- tissue processing [8]. It is clear from published liter-
ther supported and enhanced by evolving programs ature, extensive global experience, and this group’s

© 2015 John Wiley & Sons Ltd, Int. Jnl. Lab. Hem. 2015, 37, 431–449
E. E. TORLAKOVIC ET AL. | STANDARDIZATION OF BONE MARROW IMMUNOHISTOCHEMISTRY 433

experimental results (partly included in Appendix)


2 . R E C O M M E N DAT I O N S
that there is more than one tissue processing proto-
col (various combinations of various fixatives and
2.1. IHC test classification
decalcifying reagents with significant time range for
both fixation and decalcification) that may produce Inspired by The US Food and Drug Administration
excellent results for IHC testing, as well as good (FDA) classification of IHC reagents and kits based on
quality of DNA and RNA. However, the large num- the ‘risk assessment’ and ‘level of concern’, the Cana-
ber of protocols prevents standardization, may create dian Association of Pathologists (CAP-ACP) developed
difficulties for external consultation/review, and hin- classification of IHC tests based on the ‘risk assess-
ders development of EQA [8]. This is similar to ana- ment’ and ‘level of concern’, which was published in
tomic pathology (AP) where fixatives other than 2010 [3]. If the IHC results are to be used by the
formalin are likely to show good or even better per- pathologists, they are classified as Class I (lesser risk to
formance, but the AP community worldwide selects the patient safety) and, if they are to be used by clini-
to use 10% buffered formalin as their primary cians (higher risk to the patient safety) for stratifica-
choice of fixative. Therefore, narrowing choices for tion for different targeted therapies or any other
fixation and decalcification appears to be one of the clinical decision (e.g., the result of IHC testing war-
most important mandates in the process of develop- rants further genetic testing), they are classified as
ing BM IHC standards. Class II [3].
It is the lack of standardization of the pre-analytical The relationship between FDA classification of
component that prevents building EQA/PT programs IHC devices (Class I–III) and CAP-ACP classification
for BM IHC because EQA programs cannot provide of IHC tests (Class I and II) was recently addressed
large number of tissue samples with different pre-ana- by Torlakovic et al. [16]. Although both classifica-
lytical conditions that would parallel those methods tions are based on their intended use and on
used in clinical IHC laboratories. patient safety/potential to harm principles, the
The published literature was the primary basis for CAP-ACP classification emphasizes that the patient
the preparation of the recommendations in this safety/potential to harm are ultimately based on not
study. When such literature was not available, the how the industry markets their products, but on
recommendations were based on the expert consen- how pathologists and other physicians use the
sus. Guidelines summarized in this article do not results of IHC testing irrespective of FDA ruling, as
include proposed IHC panels for workup of various follows:
BM diseases/lesions, but rather are focused on tech- Class I: IHC test results are both interpreted and
nical and other general issues related to BM IHC. used by (hemato)pathologists:
Guidelines that include proposed panels for different
disorders primarily focus on biology of disease and
• Qualitative IHC tests, which are used for diagnostic
purposes; often used for determination of cell lineage.
their diagnostic sensitivity and specificity [9–15];
indirectly, such recommendations also depend on
• Validation design is determined by the medical
director of the IHC laboratory.
numerous technical parameters that are addressed
here.
• Test performance characteristics are descriptive, usu-
ally produce categorical data/results and are generally
Although standardization of BM IHC is not fully
interpreted as ‘positive’ or ‘negative’.
achievable at this time, it is entirely relevant to define
which components can be standardized at present and
which components can be harmonized now and possi- Class II: IHC test results are interpreted by (hemato)
bly standardized in the future. pathologists; however, these tests have either prog-
The aim of these guidelines for BM IHC standardi- nostic or predictive nature and their results are used
zation is to define BM IHC parameters relevant to by treating physician (oncologists or other nonpathol-
standardization, set the stage for development of BM ogist physicians, who use this information for stratify-
IHC EQA/PT, and to develop relevant framework for ing the patients for appropriate therapies or other
BM IHC performance characteristics. significant clinical decision-making that may result

© 2015 John Wiley & Sons Ltd, Int. Jnl. Lab. Hem. 2015, 37, 431–449
434 E. E. TORLAKOVIC ET AL. | STANDARDIZATION OF BONE MARROW IMMUNOHISTOCHEMISTRY

in either different treatment approach or different ment why calibrated controls are not used. It is rec-
further workup of the patient). ommended that (calibrated) controls are placed on
the same slide as the patient’s sample (so-called
• Qualitative and quantitative IHC used as prognostic
‘on-slide’ controls).
and/or predictive markers.
Recommendation 5. Reagent negative controls are
• Validation design based on published guidelines for generally not recommended for Class I IHC tests
each marker.
and are recommended for Class II IHC tests. How-
• Test performance characteristics include sensitivity, ever, published guidelines for the use of negative
specificity, reproducibility, and repeatability, and the
controls should be followed whenever possible
results may be interpreted as descriptive/qualitative
[16]. Regent negative controls are run on a sepa-
(positive vs. negative) or (semi) quantitative (% posi-
rate slide without specific primary antibody (Ab) as
tivity or H-score or other results of a specific scoring
per reference 16.
system are reported to treating physicians).
• At this time, only a few Class II IHC markers may
occasionally be relevant to BM IHC including breast 2.2. Pre-analytical standards
cancer markers in metastatic breast cancer, ALK-1 in
metastatic lung cancer, or NPM-1 when molecular Pre-analytical standards include standards relevant to
studies may not be available [2, 17, 18]. However, the pre-analytical phase. The pre-analytical phase
consideration of IHC test class is important for test starts at the time of procurement of the BM trephine
development by the medical director in the clinical and aspirate samples and ends by cutting the paraffin-
laboratory, test development for clinical trials, as well embedded (or plastic-embedded) tissue onto glass
as researchers using IHC testing of BM samples. slides. This phase includes following components: (i)
ischemic time, (ii) type of clot sample preparation,
Recommendation 1. CAP-ACP classification of IHC (iii) fixative and fixation time, (iv) type of decalcifying
tests into Class I and Class II is recommended to reagent and time of decalcification, (v) embedding
properly design and follow QA requirements rele- media, and (vi) unstained glass slides with tissue.
vant to patient safety. Due to higher risk for patient See Section 3 for special considerations relevant to
safety, Class II IHC tests need higher level of QA pre-analytical phase.
measures, as specifically recommended below.
Recommendation 2. All Class II IHC tests that are
Recommendations relevant to ischemic time
used for stratification of patients for definitive tar-
geted therapy need to follow national and/or inter- See Section 3 for special considerations relevant to
national guidelines for recommended protocol ischemic time.
validation, re-validation, and daily QC, if available. Recommendation 1. Containers with fixative need to
Recommendation 3. If national and/or international be included in BM procedure sets so that the sample
guidelines are not available, it is recommended that can be placed in the container with fixative imme-
the medical director prepare design for validation diately. It is imperative that the ischemic time be
and re-validation of Class II markers, according to predictable and very short, and therefore, the
published guidelines for validation [4]. samples need to be placed in fixative at the bedside.
Recommendation 4. If national and/or international Recommendation 2. It is recommended that BM
guidelines are not available, the Class II IHC tests biopsy imprints (touch preps) be prepared at the
need to be run with calibrated positive control and bedside by trained professional (a trained profes-
reagent negative controls. Calibrated positive sional assisting the procedure or physician who
controls need to include negative tissues, weakly performs the procedure) before the sample is put in
positive tissues, and strongly positive tissues. For the container with the fixative.
rare disease or marker of low frequency in tissues, Recommendation 3. If transportation of unfixed
it may be difficult or impossible to obtain sufficient sample is required, the transportation time of the
tissues for finely calibrated controls; when this is sample to the laboratory needs to be monitored,
the case, it is a role of medical director to docu- and it should be as short as possible.

© 2015 John Wiley & Sons Ltd, Int. Jnl. Lab. Hem. 2015, 37, 431–449
E. E. TORLAKOVIC ET AL. | STANDARDIZATION OF BONE MARROW IMMUNOHISTOCHEMISTRY 435

Recommendation 4. During transportation, dehydra-


Recommendations for fixative and fixation time
tion of the BM trephine biopsy needs to be pre-
vented by transporting the sample in a closed See Table 1 for summarized recommendations. It is
container with physiological saline, PBS, or RPMI. suggested that for practical reasons, TAT be defined as
Kendall Telfaâ (Covidien, MN, USA) or similar short, intermediate, and standard. This approach is
nonstick pads are recommended to be placed in the entirely applicable even for future purposes when
container with the sample. No other transportation new reagents of higher quality may replace currently
medium is recommended. used reagents. See Section 3 for special considerations
Recommendation 5. If vented hoods are used for relevant to fixation.
preparation of BM trephine biopsy imprints (of the Recommendation 1. Heating and stirring both
transported sample), dehydration and loss of the improve fixation and should always be considered.
sample in vented hoods due to vacuum forces used Recommendation 2. Either 10% buffered formalin
for increased air flow need to be prevented. (6–72 h of fixation before decalcification) or AZF
(Glacial acetic acid, <1%; formaldehyde, 5.6% zinc
chloride, 3%) (2–72 h of fixation before decalcifica-
Recommendations relevant for preparation of clot
tion) are recommended.
sample
Recommendation 3. Selection of fixative depends on
Recommendation 1. The least possible amount of blood the desirable turn-around-time (TAT).
should be included in the clot sample. This allows for
more rapid penetration of the fixative and also
Recommendations for decalcification
facilitates cutting of the embedded samples. Although
methods of clot section sample preparation may vary See Table 1 for summarized recommendations. See Sec-
in the total amount of clotted blood included in the tion 3 for special considerations relevant to decalcification.
sample and the clot-/gel-forming medium, only those Recommendation 1. Heating and stirring both improve
methods that enable ‘clean’ collection of BM particles decalcification and should always be considered.
are recommended (e.g., hour-glass dish with manual Recommendation 2. It is recommended that the selec-
collection of BM fragments) [19]. tion of the type of the decalcifying reagent is based

Table 1. Recommended protocols for bone marrow (BM) fixation and decalcification

Turnaround Fixation Decal


time (TAT)* Fixative time Decalcification time Comments

Very Acetic 2–72 h† ShandonTM TBD-1TM 30–40 Whenever possible,


short TAT acid–zinc–formalin Decalcifier min† longer fixation (within
(AZF) the range) is preferred
Intermediate AZF Overnight Gooding and Stewart’s 6h So-called ‘Hammersmith
TAT decalcification fluid Protocol’
(10% formic acid and 5%
formaldehyde)‡
Standard 10% buffered 8–72 h 14% EDTA 16–24 h† Preferred protocol for BM
TAT formalin = 3.7% (overnight biopsy fixation and
formaldehyde fixation is decalcification
preferred)†

*Consideration of agitation and warming to 37 °C of the decalcifying solutions are recommended for each protocol.
Ultrasonic decalcification may also be employed. These methods were shown to significantly shorten TAT.
†The timing may vary based on ancillary use of stirrers, ultrasound energization, microwave or other heating methods,
or their combination.
‡Although decalcifying fixative is not recommended to be used alone, decalcifying fixative can produce superior results
when used after the BM biopsy was already properly fixed in AZF or formalin.

© 2015 John Wiley & Sons Ltd, Int. Jnl. Lab. Hem. 2015, 37, 431–449
436 E. E. TORLAKOVIC ET AL. | STANDARDIZATION OF BONE MARROW IMMUNOHISTOCHEMISTRY

on allowable TAT as per Table 1. However, shorter including cytological evaluation using an oil-
TAT methods are not recommended if additional immersion lens with 9 100 magnification [19, 23,
samples for special studies are not available (e.g., 24].
separate sample for flow cytometry, molecular Recommendation 2. Cutting at a right angle to the
studies, and cytogenetic studies) (Figure 1). long side of the BM cores may decrease some arti-
Recommendation 3. Decalcification should be fol- facts due to inconsistencies of successive bony tra-
lowed by careful rinsing of about 10 min to remove beculae and marrow spaces compared with cutting
decalcification reagent [20, 21]. parallel to the long side.
Recommendation 4. Combined decalcifying fixative Recommendation 3. Unstained slides should be
solutions are not recommended [21, 22]. stained as soon as possible or stored at 80 °C indi-
vidually wrapped in aluminum foil to prevent anti-
gen deterioration at room temperature [25]. For
Recommendation for embedding
detailed recommendations regarding storage and
See Section 3 for special considerations relevant to shipment of unstained slides, see reference 25.
embedding.
Recommendation 1. Embedding in paraffin is recom-
2.3. Analytical standards
mended.
The analytical phase pertains to protocols used for IHC
staining including antigen retrieval, primary antibody
Recommendations for cutting
incubation, incubation with detection system, color
Recommendation 1. Whatever embedding and cutting development for visualization of immunological
methods are employed, sections must be suffi- reaction(s), counterstaining, and it ends with cover-
ciently and evenly, thin (generally 2–3 lm) to slipping. The desired results drive antibody clone
allow high-quality morphological assessment selection as well as calibration of the entire protocol by

Figure 1. Bone marrow sampling and selection of tissue processing protocol for BM tissue biopsy. Depending on the
type of BM samples, short protocol with suboptimal preservation of DNA, RNA, and protein may be acceptable.
The selection of the method (short BM TAT vs. regular BM TAT) depends on the institutional practice. If BM tissue
biopsy +/ smears for morphology are not regularly complemented by additional samples for molecular,
cytogenetic, and flow cytometry studies, short BM TAT is not acceptable as diagnostic DNA and RNA testing may
not be possible on tissues exposed to harsh acid decalcification. If such additional samples are collected and
available to hematopathologists, short TAT protocols may be acceptable.

© 2015 John Wiley & Sons Ltd, Int. Jnl. Lab. Hem. 2015, 37, 431–449
E. E. TORLAKOVIC ET AL. | STANDARDIZATION OF BONE MARROW IMMUNOHISTOCHEMISTRY 437

tweaking various parameters amenable to modification


Table 2. SOPs components required to be specified
(temperature, time, pH, etc.). As the pre-analytical for each bone marrow immunohistochemistry tests
phase cannot be absolutely controlled and therefore
cannot be entirely standardized (acceptable ischemic SOP component Descriptors
time is defined as maximum time with a range of Primary antibody Monoclonal vs. polyclonal, clone or
T0–Tmax, and similarly, there is also an acceptable time (Ab) type lot name/number, source,
range for fixation and decalcification), and this com- concentrated vs. prediluted, and
bined with multiple options for selecting other reagents dilution (if concentrated Ab),
and equipment as well as changes in equipment perfor- incubation time, temperature
Antigen retrieval Type, pH, concentration
mance or primary antibody deterioration may lead to
method (for enzyme-based methods),
changes in IHC protocols to maintain desired sensitivity temperature, time, and source
and specificity of the results. See Section 4 for special Detection system Type, name, temperature, time,
considerations relevant to the analytical phase. source
Recommendation 1. Immunohistochemistry protocols Amplification Type, temperature, time, source
Chromogen Type, time
need to be validated, that is, designed as such to
Enhancement Type, time
reflect optimal calibration of sensitivity and specific- Automated stainer Name, source
ity of the IHC test for particular use depending on the platform
biology of the tested IHC marker and its ultimate use
[4]. This assumes that performance characteristics of Recommendation 1. All BM aspirate results should be
each IHC test, irrespective of its class, are defined available to (hemato)pathologists directly, that is,
before introduction of the protocol to clinical use. they need to evaluate them and sign them out, or
Recommendation 2. Monitoring of protocol perfor- indirectly, that is, results of cytological evaluation
mance characteristics using appropriate calibrated of BM smears, molecular studies, and cytogenetic
controls is recommended. studies are submitted to (hemato)pathologists who
Recommendation 3. Separate standard operating pro- are evaluating BM tissue biopsy optimally before
cedures (SOPs) for each IHC test that is performed they are made available to other physicians.
in the laboratory need to be developed, as every Recommendation 2. Interpretation of IHC results
IHC test is a different test. All essential components should be performed in consideration of the sample
of IHC protocols for each IHC test need to be adequacy. Proper sampling of the BM cannot be
included in the SOPs (Table 2). This information overemphasized. Although IHC is a powerful
must be readily available for review for all IHC lab- technique and may help detect pathological cells
oratory staff. The protocols must be regularly even when they are present in small numbers and
updated. Updating of SOPs is required at the time not apparent morphologically, standardization and
any changes in the protocol are introduced. In addi- excellence in BM IHC cannot replace proper BM
tion, periodic review of SOPs is recommended as sampling [26]. If the BM sample does not contain
per laboratory accreditation requirements. the lesion, no special studies could compensate
Recommendation 4. To facilitate methodology trans- for the deficiency of proper sampling [27–30]. There-
fer, when BM IHC results are published (research, fore, general guidelines for BM biopsy should be fol-
case reports, etc.), all information included in SOPs lowed in the first place [19, 23, 24, 31–33].
(as shown in Table 2) is recommended to be Recommendation 3. It is recommended that the inter-
included in the ‘Methods’ section. pretation starts with the evaluation of the external
control(s), by which it is determined that the
proper antibody was applied and the test is prop-
2.4. Postanalytical standards
erly calibrated.
Postanalytical standards pertain to the interpretation Recommendation 4. Evaluation of the patient sample
of the IHC results by the (hemato)pathologist. See starts with detection and evaluation of an internal
Section 5 for special considerations relevant to the positive control if such is present. Detection of non-
postanalytical phase. specific staining should be observed if present.

© 2015 John Wiley & Sons Ltd, Int. Jnl. Lab. Hem. 2015, 37, 431–449
438 E. E. TORLAKOVIC ET AL. | STANDARDIZATION OF BONE MARROW IMMUNOHISTOCHEMISTRY

Recommendation 6. Whenever possible, external


2.5. Quality assurance standards
controls (multitissue or single tissue) should be
Quality assurance (QA) standards pertain to the fol- mounted on the same slide as patients’ samples
lowing: positive and negative controls, participation in (so-called ‘on-slide controls’).
PT provided by EQA programs, use of flow cytometry Recommendation 7. BM tissue biopsy often contains
(FCM) to validate IHC, and training and education. elements of normal hematopoiesis and includes
See Section 6 for special considerations relevant to many epitopes that are evaluated in the lesional
QA standards. tissue (internal positive control). Internal positive
controls should be evaluated before interpretation
of IHC test is conducted for the diseased tissue
Positive and negative controls
whenever present.
Published guidelines relevant to external control Recommendation 8. When internal controls are pres-
selection, design, and use should be followed when- ent in the BM tissue biopsy, they cannot fully sub-
ever possible [34]. stitute for external controls for some epitopes.
Recommendation 1. Selection of external control When the expression is normally present, the levels
samples needs to reflect desired level of analytical of epitope expression in benign BM tissue are usu-
sensitivity and specificity (calibration), which ally predictable (useful), but the range is usually
depends on definitions set by the end user (hemat- limited (not useful).
o)pathologist and/or medical director. Recommendation 9. Negative reagent controls are
Recommendation 2. External positive controls need obligatory when biotin-based detection systems are
to contain areas with at least two different levels of used [16].
expression of the target epitope (weak + strong) as Recommendation 10. Negative reagent controls are
well as expected negative areas. Tissue microarray recommended to be ordered by (hemato)patholo-
(TMA)-based positive external controls can be used, gist: (i) when the patient’s sample contains internal
but are not required for daily QC. pigments that may interfere with interpretation of
Recommendation 3. Multitissue controls or TMA con- results; (ii) when there is tissue necrosis or dense
trols may include a mixture of cores from human tissue fibrosis, and (iii) when only one IHC test has
tissues, cell-line cell blocks or xenograft tissue. If been ordered. Standards relevant to negative con-
cell lines are used, positive cell lines could be trols for IHC are detailed elsewhere [16].
mixed with negative cell lines to provide expected
negative areas.
Participation in proficiency testing (PT), provided by
Recommendation 4. Any exceptions and alterations
external quality assurance programs (EQA)
in the use of positive controls that deviate from
general principles of control design and use need to Recommendation 1. It is recommended that laborato-
be qualified and approved by the medical director. ries participate in PT schemes for BM IHC only if
Recommendation 5. Tissue processing of control the EQA program provides samples with identical
samples needs to be controlled to closely replicate or nearly identical BM tissue processing. It is not
tissue processing of the patients’ samples. Impor- recommended that laboratories participate in PT for
tantly, a decalcification step is required for control BM IHC if the EQA provider does not provide sam-
samples even if they do not contain any bony tissue. ples with identical or nearly identical BM tissue
Patients’ samples have defined range of acceptable processing.
fixation and decalcification times. As it is not possi-
ble to replicate all potential variations for the accept-
Use of flow cytometry to validate IHC
able time range in tissue processing, controls could
be processed using most frequently used time of fixa- Recommendation 1. If FCM is available and the dis-
tion and decalcification, but there should be no devi- ease state is such that it enables straight forward
ation in the type of fixative and decalcifying reagent. interpretation of IHC results (i.e., diffuse involve-

© 2015 John Wiley & Sons Ltd, Int. Jnl. Lab. Hem. 2015, 37, 431–449
E. E. TORLAKOVIC ET AL. | STANDARDIZATION OF BONE MARROW IMMUNOHISTOCHEMISTRY 439

ment of the BM trephine biopsy by lymphoma or analytical parameters, but also on pre-analytical
leukemia), corresponding IHC testing can be evalu- parameters. Many published studies report IHC results
ated for QA purposes to compare the number of on BM tissue biopsy introducing new analytical
cells and the levels of expression between the two parameters (e.g., new application of previously widely
methods. Therefore, when possible, it is recom- used antibody, novel antibody (Ab) clones, new anti-
mended to utilize FCM for QA of BM IHC for com- gen retrieval, or new detection systems, etc.) per-
monly used markers. formed on BM tissue biopsy samples that were
processed by their unique pre-analytical conditions
that cannot directly apply to many diagnostic IHC lab-
Education and training in IHC
oratories [36–38]. Because the pre-analytical condi-
Recommendation 1. Education for both technologists/ tions are critical for IHC testing, each diagnostic IHC
laboratory scientists and pathologists alike should laboratory that would like to develop such new test-
include (i) learning the theory of IHC methods and ing often cannot use published methods for methodol-
the theory of antigen distribution in normal and ogy transfer, but need to develop protocols
diseased tissues, (ii) practical laboratory aspects of independently.
assays by manual and automated methods, and (iii) These guidelines make an attempt to reconcile TAT
practical aspects of interpretation of IHC results in demands and acceptable quality of the BM tissue
control tissues and patients’ samples. All of the biopsy samples. Members of the ICSH BM IHC Stan-
above are required components of education in dardization Working Party agreed that for the great
IHC for both technologists/laboratory scientists and majority of BM core biopsy samples, there is no clini-
pathologists, but different areas are emphasized by cal need for rapid TAT. Further, for most BM core
each group. biopsy samples, shorter protocols that are not optimal
for DNA or RNA preservation might be acceptable
when other types of samples (for FCM, cytogenetics/
3 . S P E C I A L C O N S I D E R AT I O N S F O R P R E -
FISH, and molecular studies) are available (Figure 1).
A N A LY T I C A L P H A S E
Therefore, recommendations for fixation and decalcifi-
Pre-analytical standards are relevant to both, immu- cation are dependent on acceptable TAT as well as
nohistochemical methods as well as histochemical institutional practices regarding collection of samples
methods used for evaluation of BM biopsies. The for molecular, cytogenetic, and FCM studies at the
importance of BM pre-analytical conditions for evalu- time of BM tissue biopsy. If the most optimal BM
ation of BM core biopsy was highlighted by Buesche work up is desirable, including very short TAT, BM
et al. by showing that pre-analytical conditions of fixa- sampling needs to include additional aspirate samples
tion, decalcification, embedding, and marrow tissue for FCM, molecular, and cytogenetic studies, and
shrinkage during biopsy processing, and even the these studies need to be under the constituency (con-
thickness of marrow sections significantly impacted trol and oversight) of hematopathologists. This may
results of diagnosis and quantification of myelofibrosis not be achievable even if there is institutional com-
by Gomori silver impregnation in patients with mitment to provide samples of this type because: (i)
primary myelofibrosis [35]. These guidelines empha- samples for FCM, molecular, and cytogenetic evalua-
size the need for radical reduction in BM tissue pro- tion may have been submitted to immunologists,
cessing methods because: (i) there is no evidence that molecular scientists, and cytogeneticists, who do not
many different methods are required for tissue pro- report to hematopathologists, or the results of these
cessing of BM samples; (ii) standardized tissue pro- studies are instead independently released to or evalu-
cessing of BM tissue could minimize potential ated by hematologists instead of hematopathologists,
technical problems with IHC testing of external BM or (ii) there is BM fibrosis or other pathological condi-
samples (second opinion, oncology reviews, etc.), and tions that results in so-called ‘dry-tap’. When this
(iii) a limited number of applied methods will enable happens, BM tissue biopsy/core biopsies that have not
development of BM IHC EQA/PT. Methodology trans- been fixed in formalin and decalcified in EDTA may
fer of IHC testing also depends not only on published not be optimal (see Figure 1).

© 2015 John Wiley & Sons Ltd, Int. Jnl. Lab. Hem. 2015, 37, 431–449
440 E. E. TORLAKOVIC ET AL. | STANDARDIZATION OF BONE MARROW IMMUNOHISTOCHEMISTRY

mized for BM trephine biopsies that are fixed in Bou-


Ischemic time
in’s solution. However, Bouin’s solution has very low
The effects of cold ischemic time are important for pH, contains picric acid, which can be explosive, and
IHC results even if decalcification is not performed is sensitive to friction and shock when dry. Further, a
like in clot sections [39–43]. Recommended approach, recent study showed that fixation in Bouin’s solution
with partial tissue processing of the BM aspirate and did not provide IHC results comparable to those
biopsy sample at the bed site, assumes availability of obtained with formalin [50]. Fixatives containing
technical help and expertize for preparation of BM mercuric chloride like Zenker’s fixative and B5 were
biopsy imprints and BM aspirate smears at the bed- also widely shown to be suitable for IHC studies
side. This is the optimal approach and ensures preser- [51–57], but are toxic; their disposal as hazardous
vation of sample integrity. Due to practice protocols waste is costly, and these reagents are not allowed in
or lack of funds in some institutions, the transport of many countries at this time.
unfixed samples to the laboratory for further process-
ing, of both trephine biopsy and aspirate samples, is
Decalcification
being used as an alternative approach. This is a subop-
timal approach that introduces ischemic time. Degra- It was already emphasized in 1987 that knowledge of
dation of protein epitopes relevant to IHC as well as the effects of the various decalcifying agents on the
degradation of RNA and DNA integrity is created by immunoreactivity of cellular antigens is essential for
delayed fixation as a significant parameter in labora- IHC analysis of lesions in calcified tissue [58]. This
tory testing potentially leading to false-negative results early study showed that overall EDTA-based decalcifi-
not only by IHC, but also for molecular diagnostics. cation was better for IHC analysis, but also that sev-
eral differentiation antigens can be detected even in
specimens decalcified in strong acid solutions.
Fixation
Strong mineral acids, such as 10% hydrogen chlo-
Insufficient/incomplete/uneven fixation may lead to ride (HCl), or weak organic acids, such as 5–10% for-
zoning phenomenon, resulting in areas with absent or mic acid (HCOOH), form soluble calcium salts in an
suboptimal staining; zoning phenomenon should be ion exchange that moves calcium in the decalcifica-
considered and recognized at the interpretation stage tion solution. Similarly, 10–14% ethylenediaminetet-
(postanalytical phase) to prevent misdiagnosis. raacetic acid (EDTA) sequesters calcium in aqueous
Although 10% buffered formalin is widely avail- solutions as a chelating factor [20, 21]. Although deg-
able and its fixation effects on protein, DNA, and RNA radation of proteins that are detected by IHC may
are well understood, AZF was found to provide some have different kinetics than that of DNA, and RNA,
greater flexibility for workflow, eliminate labor-inten- current state of practice requires that recommenda-
sive steps, save processing time, and improve turn- tions for the IHC pre-analytical methods also take into
around time [44–47]. It is essential that fixation time account DNA and RNA integrity. Published evidence
needs to be adjusted to the type of fixative chosen; suggests that various fixatives and decalcifying agents
10% buffered formalin optimally requires 18 h of fix- may be acceptable for preservation of protein anti-
ation, but good results for IHC may also be achieved gens, DNA and RNA. However, proper timing of fixa-
with shorter fixation even for unstable markers [45, tion and decalcification is important, and some agents
47, 48]. Fixatives with protein precipitant formulation appear to be ultimately superior to others in achieving
require shorter time of fixation [46]. optimal preservation of the tissue, but the time they
There are large numbers of fixatives for BM tre- require to achieve this may not be compatible with
phine biopsy and clot preparation that continue to be local patient care requirements. Decalcification is best
in use in various institutions [8]. There are also sev- achieved by 14% EDTA that sequesters metallic ions,
eral studies showing that some or many of IHC tests including calcium, in aqueous solutions as a chelating
could be performed in BM tissue biopsy fixed in dif- factor for 16–24 h [38, 59–62]. Although a recent
ferent fixatives. For example, Gala et al. [49] showed study showed that EDTA and formic acid both per-
that there is number of antibodies that can be opti- form equally regarding preservation of DNA and RNA,

© 2015 John Wiley & Sons Ltd, Int. Jnl. Lab. Hem. 2015, 37, 431–449
E. E. TORLAKOVIC ET AL. | STANDARDIZATION OF BONE MARROW IMMUNOHISTOCHEMISTRY 441

only effects achieved after 2 h of decalcification with Hammersmith Protocol [44]. Commercial short decal-
formic acid were studied, which is insufficient for clin- cifying solutions with decalcification of 1 h or less
ical practice [63]. Similarly, another study evaluated were found acceptable for frequently used IHC tests
effects of SurgiPath Decalcifier II after 24 h, while this (Appendix). In one study, greater success of FISH was
particular reagent is more likely to be used for 2–4 h achieved when a rapid decalcifier was combined with
of decalcification in clinical practice as very small sam- B5 fixative vs. formalin (77% vs. 53%) [74]. Although
ples can be decalcified in <4 h [64]. B5 fixative provides good morphology and protein
Therefore, if institutional TAT consideration allows preservation, B5 fixative cannot be not recommended
for longer procedures, 14% disodium-EDTA dihydrate primarily because of environmental concerns, and
is recommended as tissues decalcified in such manner nucleic acid studies are not recommended on B5-fixed
are superior and allow obtaining high-quality protein, biopsies [75, 76]. Therefore, AZF that allows greater
DNA, and RNA. The timing may vary based on ancil- flexibility in fixation times, eliminates labor-intensive
lary use of stirrers, ultrasound energization, micro- steps required for B5 processing is recommended for
wave or other heating methods, or their combination. short TAT protocols instead of either B5 or formalin
In 1958, a systematic study of rate of decalcification [46]. AZF fixation was shown to have staining and
using different decalcifying reagents including 5% morphologic detail comparable to B5 and achieved
nitric acid, trichloroacetic acid, and 20% formic acid equivalent or superior antigen preservation for IHC
showed that increasing temperature from room tem- studies in the previous studies as well as in the cur-
perature to about 37–40 °C shortens the time about rent study (see Appendix). Although 10% buffered
30–40% and using agitation at RT decalcification is formalin is an excellent fixative, it is not recom-
again shortened about 30% [65]. Similarly, newer mended for short protocols; optimal fixation will not
studies show that excellent results are achieved by be achieved with fixation of <8 h, and decalcification
shortening EDTA decalcification using a magnetic stir- of suboptimal fixed BM will further contribute to loss
rer in a microwave or hot plate at 37–40 °C [66–69]. of epitopes (see Appendix).
Acceleration of the decalcifying process can be Shorter protocols for decalcification are more
achieved by using ultrasound energization at lower aggressive, which requires that the BM tissues be prop-
temperatures (e.g. 18 °C), which reduces the decalcifi- erly fixed before decalcification and that the time of
cation time to approximately 6–12 h or, in another decalcification is precisely monitored not to exceed the
study to as short as 2 h [70–72]. Confirmatory studies time that is validated for this purpose. Also, in short
of the significantly reduced EDTA decalcification time protocols, it is critical that all reagents are fresh (e.g.,
by magnetic stirring in a hot plate or microwave, or commercially available rapid decalcifying reagents will
ultrasonic energization applied to human bone mar- not perform optimally if diluted by large number of
row in diagnostic setting are rare [60]. Currently, if samples or if re-used; supplier recommendations
institutional policies ask for 1-day TAT, decalcification regarding recommended volumes and the possibility of
of the BM could be carried out after short fixation by re-use should be followed closely). If the core biopsy is
AZF using strong mineral acids, such as 10% hydro- not properly decalcified, so-called ‘surface decalcifica-
chloric acid (HCl), or weak organic acids, such as tion’ or ‘decalcification on block’ is likely to be
5–10% formic acid (HCOOH), or similar commercially performed by histotechnologist during cutting of the
available solutions; the acids form soluble calcium block, the results of which cannot be fully predicted or
salts in an ion exchange that moves calcium in the controlled, but can be deleterious to some epitopes [20,
decalcification solution. Formic acid was found suit- 21, 77–80]. Decalcification with simultaneous fixation
able for FISH and CGH studies [73]. However, the acid is not recommended. In most of such combinations,
strength and a time of decalcification matters, and 5% acid starts working before the tissue is properly fixed
formic acid was better than 10% formic acid, and time [21, 22]. However, new decalcifying solutions are being
of <24 h was better than decalcification in excess of developed, and some may be less deleterious to bone
24 h [73]. Also, 10% formic acid was found to pre- marrow tissue even after short fixation [31].
serve protein antigens, DNA, and RNA when used After fixation and decalcification, the tissue needs
after acetic acid–zinc–formalin fixative as a part of the to be further processed before embedding in paraffin.

© 2015 John Wiley & Sons Ltd, Int. Jnl. Lab. Hem. 2015, 37, 431–449
442 E. E. TORLAKOVIC ET AL. | STANDARDIZATION OF BONE MARROW IMMUNOHISTOCHEMISTRY

The most important problem is inadequate tissue specifically for how the results of the particular IHC
dehydration and clearing prior to paraffin embedding. test are used, which drives selection of appropriate
This can be prevented by preparing all solutions clones as well as calibration of the protocols (e.g.,
freshly every week, depending on the volume of tis- ALK-1 IHC in anaplastic large cell lymphoma vs. ALK-
sue processed [47]. 1 IHC in lung cancer). Therefore, protocols need to be
tailored to biologically unique applications. Most IHC
tests, including BM IHC, are laboratory-developed
Embedding
tests. As a consequence, each laboratory that is using
BM trephine biopsy could be embedded in paraffin or IHC for clinical purposes needs to define test perfor-
in plastic. Both methods have certain advantages and mance characteristics that are relevant to IHC.
disadvantages (reviewed in references 76 and 77). Optimal calibration (analytical sensitivity and spec-
Under optimal circumstances, morphology is reported ificity) is descriptive for Class I IHC tests. As example,
as superior in plastic-embedded tissue [81, 82]. How- calibration of the CD117 IHC test is descriptive as fol-
ever, there is experiential evidence of excellent mor- lows: CD117 needs to be clearly detectable in mye-
phology with appropriate fixation time and thin loid and proerythroblasts, and it needs to
sectioning (2 lm) of tissue for formalin, B5, and AZF demonstrate very strong membranous staining in
fixatives, and the need for embedding in plastic was mast cells. For Class II tests, calculation of sensitivity
challenged in 1987 by Gatter et al. [83]. With proper and specificity needs to be performed on the number
optimization, most special in situ and molecular stud- of samples recommended by national or international
ies are at least equally amenable to both embedding guidelines. Therefore, when breast cancer markers
media as shown in many studies [84–91]. Plastic are performed in the BM core biopsy, the reported
embedding techniques may obviate the need for the results need to specify if these IHC protocols were
decalcification step and thus eliminate one of the fully validated according to international guidelines
major variables impacting pre-analytical bone marrow for this particular use or not. At this time, such
IHC quality. However, only a few centers still culti- guidelines are available for breast cancer markers, but
vate expertise required for plastic embedding. Without are lacking for other markers that are generally used
a resurgence of interest and developmental efforts for BM IHC [2].
leading to broader use of plastic embedding, paraffin Although tweaking of the IHC protocols by chang-
embedding will likely remain the method of choice in ing primary antibody concentration, incubation time,
the great majority of laboratories. If plastic embedding change of detection systems, etc. may improve IHC
could be automated and cost-effective, it could testing results, no change in antigen retrieval or other
become a preferred choice for embedding of all types protocol components can re-establish epitopes lost to
of tissue biopsies, not only bone marrow samples. suboptimal pre-analytical conditions related to pro-
longed ischemic time, drying, or excessive decalcifica-
tion [41–43, 92–95]. Therefore, analytical parameters
4 . S P E C I A L C O N S I D E R AT I O N S F O R
are very important and may compensate to some
A N A LY T I C A L P H A S E
extent for suboptimal pre-analytical parameters, but
The ICSH BM IHC Working Party testing showed that, often even optimal highly sensitive protocols are not
although many pre-analytical factors show definite able to completely compensate for suboptimal pre-
effects on preservation of epitopes and success of BM analytical parameters.
IHC, superior IHC protocols may compensate, at least Clinical IHC laboratories often need to develop sep-
partly, for unfavorable pre-analytical conditions (see arate protocols for markers that are used for evalua-
Appendix). tion of tissues with special tissue processing, which
prominently includes BM tissue biopsy. Therefore,
CD34 test for BM may have different protocol than
Protocol design
CD34 test for other tissues that were not decalcified
Development of IHC protocols has to be designed not (skin, lymph nodes, other). In addition, due to the
only for bare detection of the epitope of interest, but broad range of antigen abundance, it can be useful to

© 2015 John Wiley & Sons Ltd, Int. Jnl. Lab. Hem. 2015, 37, 431–449
E. E. TORLAKOVIC ET AL. | STANDARDIZATION OF BONE MARROW IMMUNOHISTOCHEMISTRY 443

use different protocols (usually different antibody con- Therefore, it is a duty of the medical director to
centrations or different antigen retrieval method) select/design external control samples based on desir-
even in the same BM Bx depending on how the test able test performance characteristics, which are either
is being used (e.g., low-sensitivity and high-sensitivity described in published literature or, when such
protocols for Ig light chains depending if surface or published literature is not available, based on the
cytoplasmic Ig is of interest). ‘fit-for-use’ principles.
Internal controls have limitations. For example,
myeloperoxidase is very strongly expressed in maturing
5 . S P E C I A L C O N S I D E R AT I O N S F O R
granulocytes, but even very low levels are also diagnos-
P O S TA N A LY T I C A L P H A S E
tic of myeloid differentiation. In such circumstances,
IHC test results can be interpreted only after evalua- diseased tissue with low levels of expression is a better
tion of the external and internal controls, which will choice for test calibration and monitoring. When this is
indicate if the IHC test is properly calibrated and that not possible, cell types that show high levels of expres-
no internal tissue factors prevent interpretation of spe- sion of evaluated marker (e.g., myeloperoxidase)
cific IHC staining results. The expected results of vari- should demonstrate expected high intensity of IHC
ous IHC tests for BM evaluation are published and staining. If only weak reactions are demonstrated, the
updated in peer-reviewed literature as methodology results are not acceptable. For those markers for which
improves as well as our understanding of disease both weak and strong internal control is present, cell
develops [96–107, and http://www.ncbi.nlm.nih.gov]. types that normally show low levels of expression of
The overall diagnostic performance of BM samples certain markers (CD117 in proerythroblasts and myelo-
requires close co-operation between the physician and blasts) should be demonstrated and the results are not
the laboratory technologist/clinical laboratory scien- acceptable if only highly expressing cells are detected
tist, as various different types of samples may be col- (mast cells show strong expression of CD117 and are
lected, and they each may require different tissue not sufficient evidence of appropriate calibration and
processing, which is often more complex and elabo- sensitivity of the CD117 tests).
rate than that for usual anatomic pathology samples
[37]. Therefore, ideally, interpretation of the BM IHC
External quality assurance and proficiency testing
results should be performed by the (hemato)patholo-
gist familiar with the complexities of the bone marrow External quality assurance (EQA) program that would
tissue processing and their potential effects for the provide PT samples for BM IHC does not exist in the
IHC results, who is able to recognize various artifacts great majority of countries. This is secondary to large
that may results in either false-negative or false-posi- number of unique tissue processing protocols that are
tive staining. used for BM biopsy and is linked to large numbers of
IHC protocols that are optimized and validated for such
tissue biopsy protocols. This document recommends
6 . S P E C I A L C O N S I D E R AT I O N S F O R Q UA L I T Y
three methods of tissue fixation and decalcification
ASSURANCE
sorted by TAT, which in any combination should
Design of protocols is facilitated and its performance achieve comparable results for IHC (Table 1). Once tis-
monitored and optimally maintained by the use of sue processing is standardized, meaningful BM biopsy
suitable positive and negative controls depending on PT for IHC and other methods (histochemistry, in situ
the test application [3, 16]. hybridization, or other) will theoretically be possible.

Positive and negative controls Use of flow cytometry to validate IHC


Recently published guidelines recommend standardi- Theoretically, QA components in BM IHC are largely
zation of both, positive and negative controls for IHC comparable to anatomic pathology IHC as they are
[16, 34]. Although general principles apply, controls both elements of laboratory medicine quality systems.
for specific IHC test for BM IHC are not standardized. However, there are some differences that need to be

© 2015 John Wiley & Sons Ltd, Int. Jnl. Lab. Hem. 2015, 37, 431–449
444 E. E. TORLAKOVIC ET AL. | STANDARDIZATION OF BONE MARROW IMMUNOHISTOCHEMISTRY

considered; immunophenotyping using FCM fre- be decided by the medical director of IHC in collabo-
quently accompanies BM evaluation, which results in: ration with the medical director of FCM.
(i) decreased need for IHC and (ii) an additional
potentially powerful tool to compare results of IHC to
AC K N OW L E D G M E N T S
FCM. The latter has been largely underutilized for
QA. This is partly due to logistics issues as FCM and This guideline was supported and endorsed by the
biopsy IHC are performed in some countries at differ- International Council for Standardization in Haema-
ent laboratories and interpreted by different special- tology (ICSH). The Working Party gratefully acknowl-
ists. In this document, FCM is also included as a part edges the following reviewers for their expert
of BM IHC QA. Thorough understanding of the biol- comments: Prof. Dr. med. Falko Fend, University Hos-
ogy of either benign tissue or neoplastic tissues is pital Tuebingen, Tuebingen, Germany; Ass. Prof. Tracy
required for this exercise as interpretation of the I. George, University of New Mexico HSC, Albuquer-
results is dependent on ‘expected reactivities’. This is que, NM, USA; Prof. LoAnn C. Peterson, Feinberg
particularly important because these are genuinely Medical School of Northwestern University, Chicago,
different samples, and the number of positive cells IL, USA; Prof. Dr. Stefano Pileri, Bologna University
and the intensity of staining may not fully correlate in School of Medicine, Bologna, Italy; Prof. Clive R. Tay-
the two samples even if both protocols are working lor, Keck School of Medicine, University of Southern
optimally. It has been described that in some condi- California, Los Angeles, CA, USA; and Dr. Jon Van
tions, IHC and FCM may not be concordant and on der Walt, St. Thomas’ Hospital, London, UK.
occasion IHC may be more informative than FCM
[108–113]. Most common discrepancies are seen
DISCLAIMER
when FCM shows weak expression and there is nega-
tive staining by IHC. Therefore, the assessment of bio- While the advice and information in these guidelines
logically expected correlation needs to be weighted for are believed to be true and accurate at the time of
parameters related to tissue sampling and primary going to press, neither the authors, nor ICSH, nor can
antibody design. Which samples and which tests are publishers accept any legal responsibility for the con-
amenable for this type of correlative QA study should tent of these guidelines.

REFERENCES 3. Canadian Association of Pathologists- Pathologists Pathology and Laboratory


Association canadienne des pathologistes Quality Center. Arch Pathol Lab Med
1. McCourt CM, Boyle D, James J, Salto- National Standards Committee, Torlako- 2014;138:1432–43.
Tellez M. Immunohistochemistry in the vic EE, Riddell R, Banerjee D, El-Zimaity 5. Howat WJ, Lewis A, Jones P, Kampf C,
era of personalized medicine. J Clin H, Pilavdzic D, Dawe P, Magliocco A, Ponten F, der van Loos CM, Gray N, Wo-
Pathol 2013;66:58–61. Barnes P, Berendt R, Cook D, Gilks B, mack C, Warford A. Antibody validation
2. Hammond ME, Hayes DF, Dowsett M, Williams G, Perez-Ordonez B, Wehrli B, of immunohistochemistry for biomarker
Allred DC, Hagerty KL, Badve S, Fitzgib- Swanson PE, Otis CN, Nielsen S, Vyberg discovery: recommendations of a consor-
bons PL, Francis G, Goldstein NS, Hayes M, Butany J. Canadian Association of tium of academic and pharmaceutical
M, Hicks DG, Lester S, Love R, Mangu Pathologists-Association canadienne des based histopathology researchers. Meth-
PB, McShane L, Miller K, Osborne CK, pathologists National Standards Commit- ods 2014;70:34–8.
Paik S, Perlmutter J, Rhodes A, Sasano tee/Immunohistochemistry: best practice 6. Bussolati G, Leonardo E. Technical pit-
H, Schwartz JN, Sweep FC, Taube S, Tor- recommendations for standardization of falls potentially affecting diagnoses in
lakovic EE, Valenstein P, Viale G, Vis- immunohistochemistry tests. Am J Clin immunohistochemistry. J Clin Pathol
scher D, Wheeler T, Williams RB, Wittliff Pathol 2010;133:354–65. 2008;61:1184–92.
JL, Wolff AC. American Society of Clini- 4. Fitzgibbons PL, Bradley LA, Fatheree LA, 7. US Department of Health and Human Ser-
cal Oncology/College of American Alsabeh R, Fulton RS, Goldsmith JD, vices, Food and Drug Administration Cen-
Pathologists guideline recommendations Haas TS, Karabakhtsian RG, Loykasek ter for Devices and Radiological Health,
for immunohistochemical testing of PA, Marolt MJ, Shen SS, Smith AT, Immunology Branch, Division of Clinical
estrogen and progesterone receptors in Swanson PE. Principles of analytic valida- Laboratory Devices, Office of Device Eval-
breast cancer. J Clin Oncol 2010; tion of immunohistochemical assays: uation. Guidance for Submission of
28:2784–95. Guideline from the College of American Immunohistochemistry Applications to

© 2015 John Wiley & Sons Ltd, Int. Jnl. Lab. Hem. 2015, 37, 431–449
E. E. TORLAKOVIC ET AL. | STANDARDIZATION OF BONE MARROW IMMUNOHISTOCHEMISTRY 445

the FDA: Final Guidance for Industry. S, Petcu EB, Swanson PE, Taylor CR, Vy- 27. Wang J, Weiss LM, Chang KL, Slovak
June 3, 1998. http://www.fda.gov/Medi- berg M. Standardization of negative con- ML, Gaal K, Forman SJ, Arber DA. Diag-
calDevices/DeviceRegulationandGuidance/ trols in diagnostic immunohistochemistry: nostic utility of bilateral bone marrow
GuidanceDocuments/ucm094002.htm. recommendations from the international examination: significance of morphologic
Accessed April 21, 2014. ad hoc expert panel. Appl Immunohisto- and ancillary technique study in malig-
8. Torlakovic EE, Naresh K, Kremer M, van chem Mol Morphol 2014;22:241–52. nancy. Cancer 2002;94:1522–31.
der Walt J, Hyjek E, Porwit A. Call for a 17. Conklin CM, Craddock KJ, Have C, Laskin 28. Haddy TB, Parker RI, Magrath IT. Bone
European programme in external quality J, Couture C, Ionescu DN. Immunohisto- marrow involvement in young patients
assurance for bone marrow immunohis- chemistry is a reliable screening tool for with non-Hodgkin’s lymphoma: the
tochemistry; report of a European Bone identification of ALK rearrangement in importance of multiple bone marrow
Marrow Working Group pilot study. J non-small-cell lung carcinoma and is anti- samples for accurate staging. Med Pediatr
Clin Pathol 2009;62:547–51. body dependent. J Thorac Oncol Oncol 1989;17:418–23.
9. Valent P, Orazi A, B€ usche G, Schmitt- 2013;8:45–51. 29. Brunning RD, Bloomfield CD, McKenna
Gr€aff A, George TI, Sotlar K, Streubel B, 18. Goteri G, Zizzi A, Sabato S, Costagliola A, RW, Peterson LA. Bilateral trephine bone
Beham-Schmid C, Cerny-Reiterer S, Stramazzotti D, Rubini C, Discepoli G, Cap- marrow biopsies in lymphoma and other
Krieger O, van de Loosdrecht A, Kern W, elli D, Leoni P. Immunostaining for nucleo- neoplastic diseases. Ann Intern Med
Ogata K, Wimazal F, Varkonyi J, Sperr phosmin in bone marrow trephine biopsy 1975;82:365–6.
WR, Werner M, Kreipe H, Horny HP. specimens in acute myeloid leukemias. 30. Jatoi A, Dallal GE, Nguyen PL. False-neg-
Standards and impact of hematopatholo- Anal Quant Cytol Histol 2010;32:201– ative rates of tumor metastases in the
gy in myelodysplastic syndromes (MDS). 6. histologic examination of bone marrow.
Oncotarget 2010;1:483–96. 19. Lee SH, Erber WN, Porwit A, Tomonaga Mod Pathol 1999;12:29–32.
10. Beiske K, Burchill SA, Cheung IY, Hiyama M, Peterson LC; International Council for 31. Peterson LC, Brunning R. Bone marrow
E, Seeger RC, Cohn SL, Pearson AD, Matt- Standardization in Hematology. ICSH specimen processing. In: Neoplastic
hay KK; International neuroblastoma Risk guidelines for the standardization of bone Hematopathology, 2nd edn. Knowles DM
Group Task Force. Consensus criteria for marrow specimens and reports. Int J Lab (ed.). Philadelphia, PA: Lippincott, Wil-
sensitive detection of minimal neuroblas- Hematol 2008;30:349–64. liams & Wilkins; 2001: 1391–405.
toma cells in bone marrow, blood and 20. Dimenstein IB. Bone grossing techniques: 32. Bain BJ, Bailey K. Pitfalls in obtaining
stem cell preparations by immunocytology helpful hints and procedures. Ann Diagn and interpreting bone marrow aspirates:
and QRT-PCR: recommendations by the Pathol 2008;12:191–8. to err is human. J Clin Pathol 2011;64:
International Neuroblastoma Risk Group 21. Dimenstein IB. Decalcification in surgical 373–9.
Task Force. Br J Cancer 2009;100:1627– pathology. Grossing Technology in Surgi- 33. Wilkins BS. Pitfalls in bone marrow
37. cal Pathology. http://grossingtechnolo- pathology: avoiding errors in bone mar-
11. Bennett JM, Orazi A. Diagnostic criteria gy.com/newsite/home/grossing-tech- row trephine biopsy diagnosis. J Clin
to distinguish hypocellular acute myeloid niques/decalcification-in-surgical-pathol- Pathol 2011;64:380–6.
leukemia from hypocellular myelodys- ogy. Accessed May 03, 2014. 34. Torlakovic EE, Nielsen S, Francis G, Garr-
plastic syndromes and aplastic anemia: 22. Bain BJ, Clark DM, Wilkins BS. Techni- att J, Gilks B, Goldsmith JD, Hornick JL,
recommendations for a standardized cal methods applicable to trephine biopsy Hyjek E, Ibrahim M, Miller K, Petcu E,
approach. Haematologica 2009;94:264–8. specimens, in Appendix. Bone Marrow Swanson PE, Zhou X, Taylor CR, Vyberg
12. Cotelingam JD. Bone marrow biopsy: inter- Pathology, 4th edn. West Sussex, UK: M. Standardization of positive controls in
pretive guidelines for the surgical patholo- Wiley-Blackwell; 2010: 1–5 diagnostic immunohistochemistry: rec-
gist. Adv Anat Pathol 2003;10:8–26. 23. Bain BJ. Bone marrow trephine biopsy. J ommendations from the International Ad
13. Borgen E, Pantel K, Schlimok G, M€ uller Clin Pathol 2001;54:737–742. Hoc Expert Committee. Appl Immuno-
P, Otte M, Renolen A, Ehnle S, Coith C, 24. Wilkins BS, Clark DM. Making the most histochem Mol Morphol 2015;23:1–18.
Nesland JM, Naume B. A European in- of bone marrow trephine biopsy. Histo- 35. Buesche G, Georgii A, Kreipe HH. Diag-
terlaboratory testing of three well-known pathology 2009;55:631–40. nosis and quantification of bone mar-
procedures for immunocytochemical 25. Cheung CC, Banerjee D, Barnes PJ, Ber- row fibrosis are significantly biased by
detection of epithelial cells in bone mar- endt RC, Butany J, Canil S, Clarke BA, El- the pre-staining processing of bone
row. Results from analysis of normal Zimaity H, Garratt J, Geldenhuys L, Gilks marrow biopsies. Histopathology 2006;
bone marrow. Cytometry B Clin Cytom CB, Manning L, Mengel M, Perez-Ordo- 48:133–48.
2006;70:400–9. nez B, Pilavdzic D, Riddell R, Swanson PE, 36. Erber WN, Willis JI, Hoffman GJ. An
14. Buhr T, L€ anger F, Schlue J, von Wasie- Torlakovic EE. Canadian Association of enhanced immunocytochemical method
lewski R, Lehmann U, Braumann D, Kre- Pathologists-Association canadienne des for staining bone marrow trephine sec-
ipe H. Reliability of lymphoma pathologistes National Standards Commit- tions. J Clin Pathol 1997;50:389–93.
classification in bone marrow trephines. tee for High Complexity Testing/Immuno- 37. Hodges A, Koury MJ. Needle aspiration
Br J Haematol 2002;118:470–6. histochemistry: guidelines for the and biopsy in the diagnosis and monitor-
15. Ng AP, Wei A, Bhurani D, Chapple P, preparation, release, and storage of ing of bone marrow diseases. Clin Lab
Feleppa F, Juneja S. The sensitivity of unstained archived diagnostic tissue sec- Sci 1996;9:349–53.
CD138 immunostaining of bone marrow tions for immunohistochemistry. Am J 38. Korac P, Jones M, Dominis M, Kusec R,
trephine specimens for quantifying mar- Clin Pathol 2014;142:629–33. Mason DY, Banham AH, Ventura RA.
row involvement in MGUS and mye- 26. Talaulikar D, Shadbolt B, Dahlstrom JE, Application of the FICTION technique for
loma, including samples with a low McDonald A. Routine use of ancillary the simultaneous detection of immuno-
percentage of plasma cells. Haematolog- investigations in staging diffuse large B- phenotype and chromosomal abnormali-
ica 2006;91:972–5. cell lymphoma improves the Interna- ties in routinely fixed, paraffin wax
16. Torlakovic EE, Francis G, Garratt J, Gilks tional Prognostic Index (IPI). J Hematol embedded bone marrow trephines. J Clin
B, Hyjek E, Ibrahim M, Miller R, Nielsen Oncol 2009;2:49. Pathol 2005;58:1336–1338.

© 2015 John Wiley & Sons Ltd, Int. Jnl. Lab. Hem. 2015, 37, 431–449
446 E. E. TORLAKOVIC ET AL. | STANDARDIZATION OF BONE MARROW IMMUNOHISTOCHEMISTRY

39. Bai Y, Tolles J, Cheng H, Siddiqui S, in diagnostic pathology. Eur J Histochem base pairs using DNA extracted from
Gopinath A, Pectasides E, Camp RL, 2012;56:e12. decalcified, paraffin wax embedded bone
Rimm DL, Molinaro AM. Quantitative 51. Kubic VL, Brunning RD. Immunohisto- marrow trephine biopsies. Mol Pathol
assessment shows loss of antigenic epi- chemical evaluation of neoplasms in 2000;53:19–23.
topes as a function of pre-analytic vari- bone marrow biopsies using monoclonal 63. Singh VM, Salunga RC, Huang VJ, Tran
ables. Lab Invest 2011;91:1253–61. antibodies reactive in paraffin-embedded Y, Erlander M, Plumlee P, Peterson MR.
40. Siddiqui S, Rimm DL. Pre-analytic vari- tissue. Mod Pathol 1989;2:618–29. Analysis of the effect of various decalcifi-
ables and phospho-specific antibodies: 52. Wolf BC, Brady K, O’Murchadha MT, cation agents on the quantity and quality
the Achilles heel of immunohistochemis- Neiman RS. An evaluation of immuno- of nucleic acid (DNA and RNA) recov-
try. Breast Cancer Res 2010;12:113. histologic stains for immunoglobulin light ered from bone biopsies. Ann Diagn
41. Portier BP, Wang Z, Downs-Kelly E, chains in bone marrow biopsies in Pathol 2013;17:322–6.
Rowe JJ, Patil D, Lanigan C, Budd GT, benign and malignant plasma cell prolif- 64. Shao YY, Wang L, Hicks DG, Ballock RT.
Hicks DG, Rimm DL, Tubbs RR. Delay to erations. Am J Clin Pathol 1990;94:742– Analysis of gene expression in mineral-
formalin fixation ‘cold ischemia time’: 6. ized skeletal tissues by laser capture
effect on ERBB2 detection by in-situ 53. Tabilio A, Falini B, Aversa F, Zuccaccia microdissection and RT-PCR. Lab Invest
hybridization and immunohistochemis- M, Cernetti C, Gerli R, Rutili D, Grignani 2006;86:1089–95.
try. Mod Pathol 2013;26:1–9. F, Martelli MF. Intracytoplasmic lyso- 65. Verdenius HH, Alma L. A quantitative
42. Yildiz-Aktas IZ, Dabbs DJ, Bhargava R. zyme in malignant hematologic disorders: study of decalcification methods in his-
The effect of cold ischemic time on the an immunoperoxidase study. Tumori tology. J Clin Pathol 1958;11:229–36.
immunohistochemical evaluation of estro- 1982;68:417–25. 66. Madden VJ, Henson MM. Rapid decalcifi-
gen receptor, progesterone receptor, and 54. Munshi NC, Wilson C. Increased bone cation of temporal bones with preserva-
HER2 expression in invasive breast carci- marrow microvessel density in newly tion of ultrastructure. Hear Res 1997;111:
noma. Mod Pathol 2012;25:1098–105. diagnosed multiple myeloma carries a 76–84.
43. Bonnas C, Specht K, Spleiss O, Froehner poor prognosis. Semin Oncol 2001;28: 67. Keithley EM, Truong T, Chandronait B,
S, Dietmann G, Kr€ uger JM, Arbogast S, 565–9. Billings PB. Immunohistochemistry and
Feuerhake F. Effects of cold ischemia and 55. Pinkus GS, Pinkus JL. Myeloperoxidase: a microwave decalcification of human tem-
inflammatory tumor microenvironment specific marker for myeloid cells in paraf- poral bones. Hear Res 2000;148:192–6.
on detection of PI3K/AKT and MAPK fin sections. Mod Pathol 1991;4:733–41. 68. Cunningham CD 3rd, Schulte BA,
pathway activation patterns in clinical 56. Forni M, Meyer PR, Levy NB, Lukes RJ, Bianchi LM, Weber PC, Schmiedt BN.
cancer samples. Int J Cancer 2012;131: Taylor CR. An immunohistochemical Microwave decalcification of human
1621–32. study of hemoglobin A, hemoglobin F, temporal bones. Laryngoscope 2001;111:
44. Naresh KN, Lapert I, Hasserjian R, Lyki- muramidase, and transferrin in erythroid 278–82.
dis D, Elderfield K, Horncastle D, Smith hyperplasia and neoplasia. Am J Clin 69. Gonzalez-Chavez SA, Pacheco-Tena C,
N, Murray-Brown W, Stamp GW. Opti- Pathol 1983;80:145–51. Macıas-Vazquez CE, Luevano-Flores E.
mal processing of bones marrow trephine 57. Pinkus GS, Said JW. Specific identifica- Assessment of different decalcifying pro-
biopsy: the Hammersmith Protocol. J Clin tion of intracellular immunoglobulin in tocols on Osteopontin and Osteocalcin
Pathol 2006;59:903–911. paraffin sections of multiple myeloma immunostaining in whole bone speci-
45. Leong AS-Y, Gilham PN. The effect of and macroglobulinemia using an immun- mens of arthritis rat model by confocal
progressive formaldehyde fixation on the operoxidase technique. Am J Pathol immunofluorescence. Int J Clin Exp
preservation of tissue antigens. Pathology 1977;87:47–57. Pathol 2013;6:1972–83.
1989;21:81–9. 58. Athanasou NA, Quinn J, Heryet A, 70. Milan L, Trachtenberg MC. Ultrasonic
46. Bonds LA, Barnes P, Foucar K, Sever CE. Woods CG, McGee JO. Effect of decalcifi- decalcification of bone. Am J Surg Pathol
Acetic acid-zinc-formalin: a safe alterna- cation agents on immunoreactivity of cel- 1981;5:573–579.
tive to B-5 fixative. Am J Clin Pathol lular antigens. J Clin Pathol 1987;40: 71. Reineke T, Jenni B, Abdou MT, Frigerio
2005;124:205–11. 874–8. S, Zubler P, Moch H, Tinguely M. Ultra-
47. Werner M, Chott A, Fabiano A, Battifora 59. Alers JC, Krijtenburg PJ, Vissers KJ, van sonic decalcification offers new perspec-
H. Effect of formalin tissue fixation and Dekken H. Effect of bone decalcification tives for rapid FISH, DNA, and RT-PCR
processing on immunohistochemistry. procedures on DNA in situ hybridization analysis in bone marrow trephines. Am J
Am J Surg Pathol 2000;24:1016–9. and comparative genomic hybridization: Surg Pathol 2006;30:892–6.
48. Goldstein NS, Ferkowicz M, Odish E, EDTA is highly preferable to a routinely 72. Callis G, Sterchi D. Decalcification of
Mani A, Hastah F. Minimum formalin used acid decalcifier. J Histochem Cyto- bone: literature review and practical
fixation time for consistent estrogen chem 1999;47:703–10. study of various decalcifying agents,
receptor immunohistochemical staining 60. Quintanilla-Martinez L, Tinguely M, methods, and their effects on bone his-
of invasive breast carcinoma. Am J Clin Bonzheim I, Fend F. Bone marrow tology. J Histotechnol 1998;21:49–58.
Pathol 2003;120:86–92. biopsy: processing and use of molecular 73. Brown RSD, Edwards J, Bartlett JW,
49. Gala JL, Chenut F, Hong KB, Rodhain J, techniques. Pathologe 2012;33:481–9. Jones C, Dogan A. Routine acid decalcifi-
Camby P, Philippe M, Scheiff JM. A 61. Kremer M, Quintanilla-Martınez L, cation of bone marrow samples can pre-
panel of antibodies for the immunostain- N€ahrig J, von Schilling C, Fend F. Immu- serve for FISH and CGH studies in
ing of Bouin’s fixed bone marrow tre- nohistochemistry in bone marrow metastatic prostate cancer. J Histochem
phine biopsies. J Clin Pathol 1997;50: pathology: a useful adjunct for morpho- Cytochem 2002;50:113–115.
521–4. logic diagnosis. Virchows Arch 2005;447: 74. Miranda RN, Mark Hon Fong L, Medei-
50. Benerini Gatta L, Cadei M, Balzarini P, 920–37. ros LJ. Fluorescent in situ hybridization
Castriciano S, Paroni R, Verzeletti A, Cor- 62. Wickham CL, Boyce M, Joyner MV, in routinely processed bone marrow aspi-
tellini V, De Ferrari F, Grigolato P. Appli- Wilkins BS, Jones DB, Ellard S. Amplifi- rate clot and core biopsy sections. Am J
cation of alternative fixatives to formalin cation of PCR products in excess of 600 Pathology 1994;145:1309–14.

© 2015 John Wiley & Sons Ltd, Int. Jnl. Lab. Hem. 2015, 37, 431–449
E. E. TORLAKOVIC ET AL. | STANDARDIZATION OF BONE MARROW IMMUNOHISTOCHEMISTRY 447

75. WHO. 2005 Mercury in Health Care: biopsy specimens after microwave heat- 99. Thiele J, Kvasnicka HM, Diehl V. Bone
Policy Paper, http://www.who.int/water_ ing. J Clin Pathol 1995;48:840–4. marrow CD34+ progenitor cells in Phila-
sanitation_health/medicalwaste/mercury/ 89. Zhang Q, Wang J, Wu H, Zhang L, Zhou delphia chromosome-negative chronic
en/index.html, Accessed July 01, 2014. J, Ye Q, Shao X, Guan C, Xu J, Yang Y, myeloproliferative disorders—a clinico-
76. Nagasaka T, Lai R, Chen YY, Chen W, Zhou R, Ouyang J. Low-temperature gly- pathological study on 575 patients. Leuk
Arber DA, Chang KL, Weiss LM. The use col methacrylate resin embedding Lymphoma 2005;46:709–15.
of archival bone marrow specimens in method: a protocol suitable for bone 100. Douds JJ, Long DJ, Kim AS, Li S. Diag-
detecting B-cell non-Hodgkin’s lympho- marrow immunohistochemistry, PCR, nostic and prognostic significance of
mas using polymerase chain reaction and fish analysis. Microsc Res Tech CD200 expression and its stability in
methods. Leuk Lymphoma 2000;36:347– 2010;73:1067–71. plasma cell myeloma. J Clin Pathol
52. 90. Kunze E, Middel P, Fayyazi A, Schweyer 2014;67:792–6.
77. Page KM. Bone. In: Theory and Practice S. Immunohistochemical staining of 101. Cogbill CH, Swerdlow SH, Gibson SE.
of Histological Techniques. Bancroft JD, plastic (methyl-methacrylate)-embedded Utility of CD279/PD-1 immunohisto-
Stevens A (eds). New York: Churchill bone marrow biopsies applying the chemistry in the evaluation of benign
Livingstone; 1996: 309–39. biotin-free tyramide signal amplification and neoplastic T-Cell-rich bone marrow
78. Rolls GO. Difficult Blocks and Reprocess- system. Appl Immunohistochem Mol infiltrates. Am J Clin Pathol 2014;142:
ing. Wetzlar: Leica Microsystems; 2011. Morphol 2008;16:76–82. 88–98.
79. Krenacs T, Bagdi E, Stelkovics E, Ber- 91. Wolf E, Roser K, Hahn M, Welkerling H, 102. L’Abbate A, Lo Cunsolo C, Macrı E,
eczki L, Krenacs L. How we process tre- Delling G. Enzyme and immunohisto- Iuzzolino P, Mecucci C, Doglioni C, Coco
phine biopsy specimens: epoxy resin chemistry on undecalcified bone marrow M, Muscarella LA, Salati S, Tagliafico E,
embedded bone marrow biopsies. J Clin biopsies after embedding in plastic: a Minoia C, De Tullio G, Guarini A, Testoni
Pathol 2005;58:897–903. new embedding method for routine N, Agostinelli C, Storlazzi CT. FOXP1 and
80. Brown D, Gatter K, Natkunam Y, Warnke application. Virchows Arch (Pathol Anat) TP63 involvement in the progression of
R. Bone Marrow Diagnosis: An Illustrated 1992;420:17–24. myelodysplastic syndrome with 5q- and
Guide. Oxford: Blackwell Publishing, 92. Erber WN, Gibbs TA, Ivey JG. Antigen additional cytogenetic abnormalities.
2006: p1. retrieval by microwave oven heating for BMC Cancer 2014;14:396.
81. Burkhardt R. Bone marrow and bone tis- immunohistochemical analysis of bone 103. Park J, Moon S, Lee J, Park J, Lee D, Jung
sue. Color Atlas of Clinical Histopathol- marrow trephine biopsies. Pathology K, Song Y, Lee E. Bone marrow analysis of
ogy. New York: Springer-Verlag, 1971: 1996;28:45–50. immune cells and apoptosis in patients with
8–11. 93. True LD. Methodological requirements systemic lupus erythematosus. Lupus.
82. Frisch B, Bartl R. Appendix. In: Atlas of for valid tissue-based biomarker studies 2014;23:975–85.
bone marrow pathology. Frisch B, Bartl that can be used in clinical practice. Vir- 104. Zhao S, Xing Y, Natkunam Y. Use of CD137
R (eds). Dordrecht: Kluwer Academic chows Arch 2014;464:257–63. ligand expression in the detection of small
Publishers; 1990: p. 32. 94. Johnsen IK, Hahner S, Briere JJ, Ozimek B-cell lymphomas involving the bone mar-
83. Gatter KC, Heryet A, Brown DC, Mason A, Gimenez-Roqueplo AP, Hantel C, row. Hum Pathol 2014;45:1024–30.
DY. Is it necessary to embed bone mar- Adam P, Bertherat J, Beuschlein F. Eval- 105. Xiao R, Cerny J, Devitt K, Dresser K, Nath
row biopsies in plastic for haematological uation of a standardized protocol for pro- R, Ramanathan M, Rodig SJ, Chen BJ,
diagnosis? Histopathology 1987;11:1–7. cessing adrenal tumor samples: Woda BA, Yu H. MYC protein expression
84. Toth Z, Czoma V. Immunohistochemistry preparation for a European adrenal is detected in plasma cell myeloma but not
on epoxy resin-embedded bone marrow tumor bank. Horm Metab Res 2010;42: in monoclonal gammopathy of undeter-
biopsy experience with 936 cases. Appl 93–101. mined significance (MGUS). Am J Surg
Immunohistochem Mol Morphol 95. Neumeister VM, Anagnostou V, Siddiqui Pathol 2014;38:776–83.
2011;19:15–20. S, England AM, Zarrella ER, Vassilako- 106. Tian WL, He F, Fu X, Lin JT, Tang P, Hu-
85. Kunze E, Middel P, Fayyazi A, Schweyer poulou M, Parisi F, Kluger Y, Hicks DG, ang YM, Guo R, Sun L. High expression
S. Immunohistochemical staining of plas- Rimm DL. Quantitative assessment of of heat shock protein 90 alpha and its
tic (methyl-methacrylate)-embedded effect of preanalytic cold ischemic time on significance in human acute leukemia
bone marrow biopsies applying the biotin- protein expression in breast cancer tissues. cells. Gene 2014;542:122–8.
free tyramide signal amplification system. J Natl Cancer Inst 2012;104:1815–24. 107. Padhi S, Varghese RG, Ramdas A. Cyclin
Appl Immunohistochem Mol Morphol 96. Orazi A. Histopathology in the diagnosis D1 expression in multiple myeloma by
2008;16:76–82. and classification of acute myeloid leuke- immunohistochemistry: case series of 14
86. Gaiser T, Bernhards J. Tyramide signal mia, myelodysplastic syndromes, and patients and literature review. Indian J
amplification: an enhanced method for myelodysplastic/myeloproliferative dis- Med Paediatr Oncol 2013;34:283–91.
immunohistochemistry on methyl-meth- eases. Pathobiology 2007;74:97–114. 108. Kreft A, Holtmann H, Schad A, Kirkpa-
acrylate-embedded bone marrow tre- 97. Joshi R, Horncastle D, Elderfield K, Lam- trick CJ. Detection of residual leukemic
phine sections. Acta Haematol 2007;117: pert I, Rahemtulla A, Naresh KN. Bone blasts in adult patients with acute T-lym-
122–7. marrow trephine combined with immu- phoblastic leukemia using bone marrow
87. Blythe D, Hand NM, Jackson P, Barrans nohistochemistry is superior to bone mar- trephine biopsies: comparison of fluores-
SL, Bradbury RD, Jack AS. Use of methyl row aspirate in follow-up of myeloma cent immunohistochemistry with conven-
methacrylate resin for embedding bone patients. J Clin Pathol 2008;61:213–6. tional cytologic and flow-cytometric
marrow trephine biopsy specimens. J 98. Das R, Hayer J, Dey P, Garewal G. Com- analysis. Pathol Res Pract 2010;206:560–4.
Clin Pathol 1997;50:45–9. parative study of myelodysplastic syn- 109. Merli M, Arcaini L, Boveri E, Rattotti S,
88. McCluggage WG, Roddy S, Whiteside C, dromes and normal bone marrow Picone C, Passamonti F, Tenore A, Sozza-
Burton J, McBride H, Maxwell P, Bharu- biopsies with conventional staining and ni L, Lucioni M, Varettoni M, Rizzi S,
cha H. Immunohistochemical staining of immunocytochemistry. Anal Quant Cytol Morello L, Ferretti V, Pascutto C, Paulli
plastic embedded bone marrow trephine Histol 2005;27:152–6. M, Lazzarino M. Assessment of bone

© 2015 John Wiley & Sons Ltd, Int. Jnl. Lab. Hem. 2015, 37, 431–449
448 E. E. TORLAKOVIC ET AL. | STANDARDIZATION OF BONE MARROW IMMUNOHISTOCHEMISTRY

marrow involvement in non-Hodgkin’s 111. Schmidt B, Kremer M, G€ otze K, John K, interactions? Leuk Lymphoma 2006;47:
lymphomas: comparison between histol- Peschel C, H€ofler H, Fend F. Bone mar- 1730–1.
ogy and flow cytometry. Eur J Haematol row involvement in follicular lymphoma: 113. Manaloor EJ, Neiman RS, Heilman DK,
2010;85:405–15. comparison of histology and flow cytom- Albitar M, Casey T, Vattuone T, Kotylo P,
110. Iancu D, Hao S, Lin P, Anderson SK, Jor- etry as staging procedures. Leuk Lym- Orazi A. Immunohistochemistry can be
gensen JL, McLaughlin P, Medeiros LJ. phoma 2006;47:1857–62. used to subtype acute myeloid leukemia
Follicular lymphoma in staging bone 112. Katz BZ, Polliack A. Discrepancies in in routinely processed bone marrow
marrow specimens: correlation of histo- quantitative assessment of bone marrow biopsy specimens. Comparison with flow
logic findings with the results of flow involvement in lymphoma: do they cytometry. Am J Clin Pathol
cytometry immunophenotypic analysis. reflect specialized micro-environmental 2000;113:814–22.
Arch Pathol Lab Med 2007;131:282–7. cellular niches and cell-stromal

APPENDIX and decalcification. Spleen, tonsil, and appendix sam-


ples were obtained and TMAs created. Tissues were
either prospectively collected or were retrieved from
BAC K G R O U N D the archives of pathology departments. They included
ICSH BM IHC Standardization Working Party tested
clinically relevant fixation and decalcification condi- Figure A1. CD34 immunoassay using clone QBEnd/
tions for the most frequently used IHC markers. 10. Effect of different methods of decalcification and
Herein, the results summarize the most important different IHC protocols. Tissue samples were fixed
according to individual laboratory methods for
conclusions for CD34 IHC staining.
clinical samples, except for modification in method
B, in which decalcification time was extended for
15 min longer than recommended by the supplier.
METHODS
(A) CD34 staining can be demonstrated to some
Members of the working party submitted tissues pro- degree in most tested samples after insufficient
fixation (2 h in 10% buffered formalin) and are
cessed according to the BM protocol used in their insti-
overall clinically acceptable (score ≥ 3). However,
tution as well as tissues processed specifically to test even after 15 min of extended decalcification (B),
alternative conditions of fixation and decalcification. the results become poor for most samples and most
The primary intent was to evaluate the results of epi- protocols. Optimized protocol with Karnofsky fixative
tope preservation in the clinically relevant range for the (C) is also overall suboptimal for demonstration of
10 IHC test that are some of the most used on BM tissue CD34. Formic acid after 2 h of decalcification shows
very good to excellent results for most protocols, but
biopsy. The secondary intent was to evaluate if differ-
samples used in this protocol did not contain bone so
ence in the protocols that are used in different laborato- this time is not applicable to clinical practice
ries can ameliorate unfavorable conditions of fixation although the results are significantly better
(P = 0.001 for 2 h vs. 6 h or 16 h) (D). Longer
decalcification in formic acid between 6 and 16 h (E
and F) does not show any differences that appear
relevant to clinical practice (the difference between 6
and 16 h are not significant), and most samples will
show epitope loss to some degree. Samples that were
decalcified in EDTA show excellent epitope
demonstration even with suboptimal protocols (G).
Another commercial decalcifying agent after formalin
fixation (H) as well as one method that employed
simultaneous formalin fixation and rapid
decalcification (I) showed significant epitope loss in
most protocols. The most rapid protocol with 2 h
zinc formalin and 30 min TBD1 decalcifying solution
showed poor and very poor results with suboptimal
protocols, but very good to excellent results with
optimal protocols. Note: Different IHC protocols are
illustrated by different colors.

© 2015 John Wiley & Sons Ltd, Int. Jnl. Lab. Hem. 2015, 37, 431–449
E. E. TORLAKOVIC ET AL. | STANDARDIZATION OF BONE MARROW IMMUNOHISTOCHEMISTRY 449

spleen, lymph node, gut, and bone marrow samples.


TMAs were constructed and unstained sections sent to
five laboratories including laboratories in the USA,
Canada, and Europe. Pre-analytical methods were set
up as follows: (i) 10% buffered formalin fixation for 2,
4, 18, or 72 h followed by rapid decalcification or 18 h
of fixation followed by 36 h EDTA decalcification; (ii)
modified Karnofsky fixative 1% glutaraldehyde/4%
formaldehyde fixation for 4, 18, or 72 h followed by
formic acid decalcification for 6 or 18 h or EDTA for 36
or 72 h; (iii) B5 fixation for 4, 18, or 72 h of fixation
followed by 6 or 18 h of formic acid decalcification or
rapid decalcification, and (iv) buffered zinc formalin
fixation for 4, 18, or 72 h followed by rapid decalcifica-
tion. Interpretation by an expert panel was done by
Figure A2. Immunoassay using clone QBEnd/10 evaluating the results on the multihead microscope. If
(CD34). Effect of different method of decalcification any discrepancies in scoring were present, the final
and different IHC protocols. Tissue samples were fixed score was obtained by majority vote. The semiquantita-
and decalcified as follows: formalin fixation for 2 and tive scoring was used (0–4+); 4+ was designated for
1 h of rapid decalcification (A), formalin fixation for optimal results, 3+ for suboptimal but clinically useful
2 h and 10 30″ rapid decalcification (B) showing
significant difference between recommended and
result, while 0–2+ scores were considered unacceptable
extended decalcification (P = 0.004). Formalin fixation for clinical practice.
for 24 and 1 h of rapid decalcification (C), and formalin
fixation for 24 h and 10 30″ rapid decalcification (D);
only minimal differences in the results were obtained. R E S U LT S
The results of this experiment show that proper
The results of CD34 testing are illustrated in Fig-
fixation is essential to stabilize epitopes against
potentially harmful decalcification protocols and that ures A1 and A2. Proper fixation is important if decal-
optimal protocols may partly compensate for cification is not strictly monitored. Also, some
suboptimal tissue processing. It also illustrates that methods were overall more robust than others (proper
careful timing of decalcification is important if short fixation in formalin followed by standard decalcifica-
fixation protocols with formalin are used. Note: tion in EDTA), which were successful in demonstra-
Different IHC protocols are illustrated by different
colors.
tion of the epitope at clinically significant levels only
by highly sensitive protocols.

© 2015 John Wiley & Sons Ltd, Int. Jnl. Lab. Hem. 2015, 37, 431–449

You might also like