Chapter 10
CLINICAL FLOW CYTOMETRY
A Transition in Utilization
Charles L. Goolsby, Mary Paniagua, Laura Marszalek
Department of Pathology (CLG), Robert H. Lurie Comprehensive Cancer Center (CLG and
MP), Northwestern University Feinberg School of Medicine, and Northwestern Memorial
Hospital (LM), Chicago, Illinois
1. INTRODUCTION
Over the last 25 years, flow cytometry analysis of hematopoietic
malignancies has evolved dramatically. In the early days, single color
analysis, or one antigen at a time, on ficoll-hypaque separated cells was the
standard approach. Thus, co-expression of antigens on cells, a hallmark of
current practice, could only be inferred by comparing numbers of cells
positive for each antigen. Density gradient preparation of the cells carried
with it the potential for either loss or enrichment of specific cell populations
that was variable from sample to sample. These technical limitations limited
the sensitivity and specificity of leukemia/lymphoma immunophenotyping in
those early days. Certainly, even with these limitations, however, these data
were still very useful as an adjunct to diagnosis, although at the time, seldom
were they integrated into the primary diagnostic package. Most frequently,
the immunophenotypic results were issued as a separate document,
frequently days after the primary diagnostic pathology report.
In the intervening years since those early beginnings, there has been
steady development of reagents and instrumentation. We have seen not only
dramatic increases in the numbers of highly specific monoclonal antibodies
but, particularly recently, in the numbers of fluorescent dyes available as
well. The standard of practice is now routine four color immunophenotyping
with many laboratories moving to five, six, seven or more colors. For the
most part, density gradient preparation of cells for these analyses is a thing
240 Clinical Flow Cytometry
of the past. These changes have dramatically increased the sensitivity and
specificity of these assays. The analyses have become the preferred method
of lineage determination in acute leukemias when morphology or special
stains are unable to make that assessment.1,2 Sub-classification of the B cell
chronic lymphoproliferative disorders now depends heavily on
immunophenotypic data,2 and cytometric analyses provide a sensitive
method of detection of clonal B cell populations.2 Routinely, laboratories
can detect malignant cell populations at significantly less than 1% of the
total cells (frequently less than 0.1%) and in some cases can approach RT-
PCR levels of sensitivity. In addition, it is now accepted that careful and
thorough correlation of these data with other pathology findings,
cytogenetics, and molecular analyses is critical for correct interpretation and
maximizing the appropriate utilization of these data.1,2 As a result, in most
institutions, immunphenotyping has become part of the primary diagnostic
work-up, ideally incorporated into the primary diagnostic reports. In
addition, to continuing to serve an increasingly important adjunctive role in
diagnosis, in some instances, immunophenotyping provides important
prognostic information as well, for example CD38 expression in HIV,3
CD38 expression in B-CLL,4-6 or Zap70 expression levels in B-CLL.7
Although not the topic of this chapter, the reader is referred to any of a
number of excellent reviews on the diagnostic and prognostic uses of these
data for more details.1,2 It is worth noting that the role as an adjunct to
diagnosis continues to evolve with new AML sub-classifications based on
signal pathway responses to growth factors being proposed.8 This is a very
attractive concept in that these measurements will most likely reflect at the
functional level abnormalities that are the result of disease characteristic
translocations and genetic abnormalities. Perhaps most exciting is that they
may then directly tie to new, and emerging, “targeted” therapies directed at
these abnormal signaling events that in many instances have lead to the
dysregulated proliferation and/or apoptosis that drive the disease process.
2. HISTORICAL PERSPECTIVE
In addition to its utility as an adjunct to diagnosis, exciting new horizons
for clinical cytometry are rapidly emerging, and developing, as tools in the
therapeutic management of leukemia and lymphoma patients. Increasingly
flow cytometric analyses are becoming key in therapeutic decisions and in
therapeutic monitoring of patients. The explosion of antigen and ligand
directed therapies for a range of hematopoietic malignancies, including anti-
CD20,9-12 anti-CD22,13 anti-CD52,11,12,14 anti-CD30,15 anti-CD33,11,12,16-18
anti-CD45,11,17,18 and IL-2,19 are having a significant impact on most clinical
Clinical Flow Cytometry 241
flow cytometry laboratories. Determining whether the malignant cells
express the relevant antigen or ligand receptor is key in determining patient
eligibility for a given therapy. Further, monitoring to see if the therapeutic
reagent has bound and whether that has resulted in cell death of both the
relevant malignant cells as well as normal cells is key in therapeutic
monitoring.9 The first major foray of flow cytometry into the arena of
therapeutic monitoring of an antibody treatment was in the setting of OKT3
immunosuppressive therapy in solid organ transplant patients.20-24 OKT3, an
anti-CD3 directed antibody, bound to the T cells leading to death of the T
cells and reductions or loss of cell mediated immune responses in these
transplant patients. Flow cytometric analyses employing a panel of
antibodies reacting with T cell antigens were used to determine if the drug
(OKT3 antibody) had bound to the T cells. Detection of T cells using
fluorescently labeled antibodies directed against either CD3 epitopes
independent of the OKT3 binding site, TCR, or other T cell associated
antigens such as CD2, CD5, or CD7 were employed along with a
fluorescently labeled OKT3 antibody. This allowed one to first, determine
on the T cells present (detected based on TCR, CD2, CD5, or CD7 staining)
whether the unlabeled OKT3 drug had bound since it would block binding of
the labeled OKT3 reagent being used. Further, it facilitated determination of
therapeutic efficacy by enumerating either, or both, the relative or absolute
numbers of T cells indicating whether the drug had successfully eliminated
the T cells. Flow cytometric bead based assays were also used to monitor
circulating levels of the OKT3 drug 21 and to determine if the patient was
producing antibodies directed against the treatment OKT3 antibody23,24
which might potentially be responsible for immunosuppressive treatment
failure. In addition, examination of T cells and T cell subsets was useful in
assessing T cell immune status following reduction or modulation of
immunosuppressive therapy.
3. CURRENT ROLE IN PATIENT THERAPEUTIC
MANAGEMENT
As noted above, the use of flow cytometric immunophenotyping as a tool
in therapeutic monitoring is growing, at least initially, primarily due to the
rapidly expanding numbers of antigen, or receptor, directed therapies that are
now options for treatment of a number of hematopoietic malignancies.9-19
The first impact of immunophenotyping is in determining whether a patient
is eligible for a specific targeted therapy through assessment of the relevant
therapeutic target (antigen(s) or receptor(s)) expression specifically on the
malignant cells. Many clinical flow cytometry laboratories now routinely
242 Clinical Flow Cytometiy
catalogue the expression of a number of potential therapeutically relevant
antigens on the malignant cells (CD20, CD22, CD25, CD52, CD30 for
example) for every relevant diagnostic specimen. In many instances, this
permits alternative treatment decisions to be made at a later time, potentially
without the need to re-biopsy the patient. In addition, as with OKT3
immunosuppressive therapy, flow cytometric immunphenotyping can be
useful in monitoring therapy by determining if the drug has bound and if that
has lead to reductions or loss of the malignant cells. Importantly, the effect
of drug on the patient’s normal cells expressing the same therapeutic target
(antigen or receptor) can also be assessed.
As an example, monitoring of rituximab (anti-CD20) therapy in patients
with a B cell malignancy is now routine and can play an important role in
monitoring of these patients.9 In a like manner to monitoring of OKT3
therapy described above, a panel of antibodies directed against B cell related
antigens including CD20, CD19, CD79b, kappa, and lambda, allows one to
determine if the anti-CD20 therapeutic has bound and whether the B cells,
more specifically the clonal B cells, have been eliminated in that patient.
With initiation of rituximab therapy, typically, a loss of peripheral B cells is
seen over 1-3 weeks.9,25,26 However, in some patients, one can see binding
of the drug but no loss of the clonal B cells.9 An example of this
demonstrating successful binding of the anti-CD20 therapeutic without loss
of the clonal B cells in a rituximab treated B cell lymphoma patient is shown
in Figure 1.
Flow cytometric analyses can also provide a sensitive tool for monitoring
of recurrence in these patients, a not always clear picture as morphologic
assessment of post-treatment bone marrows following rituximab therapy can
be complex. Immunophenotypic analyses can be a significant adjunct in
differentiating reactive responses from residual or recurrent disease in these
patients.9 In addition, it is worth noting that, although rare, recurrence of true
CD20 negative B cells of the same clone as the pre-treatment malignancy
have also been reported. Further, flow cytometric analyses provide a highly
sensitive detection of residual disease cells in the setting of antibody
mediated therapies.27 Although at present it is not always clear how to
interpret the finding of a small (<0.1%) clonal population, the potential for
detecting, and understanding, emergence of resistance in this setting
remains. This sensitivity may also prove useful in selecting and assessing the
effectiveness of additional, or co-, therapies. Again, although the potential
impact of small B cell clones may not be clear, screening of patients prior to
stem cell collection, or of collected stem cell products, during periods of
remission following antibody therapy is also frequently done. Lastly, again
in parallel with the OKT3 therapeutic monitoring model, flow cytometric
immunophenotyping can be useful in monitoring regeneration of the B cell
Clinical Flow Cytometry 243
compartment following cessation of therapy, as interestingly, return of
normal B cell levels in the peripheral blood can take many months. 9,25,26
Of note are reports of differential cell killing in different tissue
compartments following anti-CD52 therapy (Campath).28-30 In B cell non-
Hodgkin’s lymphoma, poor responses have been reported in lymph nodes as
compared to peripheral blood and bone marrow.29,30 Also, a case report of a
patient with a t(14;18)+ lymphoma in which the peripheral blood and bone
marrow remained PCR positive following chemotherapy showed eradication
of the B cell clone in the peripheral blood at the PCR level but not in the
bone marrow with Campath treatment.28 It is important to note in the latter
case that the bone marrow remained positive for lymphoma by morphology
and immunohistochemistry following Campath as well. This differential
response in different tissues to therapy has lead to clinical trials assessing the
244 Clinical Flow Cytometry
combination of rituximab and Campath in treatment of B cell
malignancies.31 Although significant clinical responses were seen, in initial
trials of rituximab addition to Campath treatment of B-CLL patients,
eradication of the clonal B cells was still not seen in the bone marrows of
these patients.32 Thus, in the immunophenotypic monitoring of patients
treated with Campath, and likely other antibody mediated treatments,
examination of not only peripheral blood, but in many patients, simultaneous
examination of bone marrow and tissue samples may be equally as
important.
Not unexpectedly, a similar testing scenario as that described above for
anti-CD20 therapy is being seen in the monitoring of other antigen directed
therapies. There is every reason to believe that the numbers of antigen and/or
ligand targeted therapies for the treatment of hematopoietic malignancies is
only going to grow over the next several years.10,33 In addition, an
increasing utilization of these therapeutic strategies is being seen in a variety
of other diseases and treatment settings.34-44 Thus, an increasing demand for
assessment of a large number of therapy related markers can be anticipated
in most clinical flow cytometry laboratories. These assays will probably
become more complex involving minimally four color analyses, and in many
cases more than four colors. This will allow more efficient analysis of the
large number of antigens and ligands that will be of interest to the clinician.
However, there are more important, and fundamental, reasons for an
extension to greater than three or four color techniques. In many instances,
assessment of two, three, or even four antigens is necessary to simply
specifically identify the aberrant cell in a sample. This may leave only one,
or in some cases no, fluorescence color to assess the presence or expression
of a potential therapeutic target on the malignant cells or to assess the effect
of the therapeutic on those same malignant cells. Thus, effective assessment
of therapeutic binding and effects is going to require an extension into more
complex analyses. Specifically assessing the effect of the drug on apoptotic
and proliferation regulatory pathways will lead to new understanding of the
mechanisms of drug action,45 and as importantly, perhaps mechanisms of
resistance. An additional complexity may arise from the need to quantify
antigen, or therapeutic target, levels. Golay et al,46 have shown that at least
in vitro the cytotoxic killing of B cells by rituximab correlates with CD20
staining intensity and, although not yet proven, it has been suggested that
this may be the case in vivo as well.47 Further, flow cytometric monitoring
may play an important role in assessing cytokine or other treatments
designed to modulate the expression level of a therapeutic target, potentially
increasing the effectiveness of a directed therapy.48,49 Thus, the need to
quantitatively measure target molecule levels is almost certainly going to be
important.
Clinical Flow Cytometry 245
4. FUTURE POTENTIAL ROLES
Perhaps even more exciting is the potential role that cytometric analyses
may play in assessing and monitoring of the new generation of therapies
such as imatinib mesylate (STI571, Gleevec)50,51 that more specifically target
abnormal or dysregulated proteins in the malignant or abnormal cells in a
patient. Driven by the Human Genome and Proteomics initiatives, the pace
at which genes and molecules associated with specific diseases are being
identified has greatly accelerated as has the understanding of these
abnormalities in the context of cell regulation and the biology of disease.
This knowledge will foster increasingly rapid development of new
therapeutics that, like imatinib mesylate, more specifically target
abnormalities or dysregulated functions in the malignant or abnormal cells.
Clearly, in treatment of human malignancies, it is believed that development
of rational therapies targeted at known abnormalities in tumor cells versus
normal cells will produce significant reductions in therapy-induced toxicity
compared to conventional chemotherapy. This hope was realized in the
treatment of chronic myelogenous leukemia patients with imatinib mesylate,
where extremely low toxicity was seen. With this in mind, especially for
cancer modalities, previously reliable laboratory endpoints often may not
provide adequate information about the effects of treatment during therapy.52
Not surprisingly, many of the new rationally designed, directed therapies
target molecules involved in signal transduction pathways that regulate cell
proliferation, apoptosis, differentiation, or angiogenesis.52-54 The
development of new, and novel, flow cytometry cell based assays to measure
the impact of therapy on these regulatory pathways, both in the malignant, or
abnormal, cells as well as in normal cells, will play a vital role in the
monitoring of these therapies as well as development of new therapies. Most
regulatory pathways controlling cellular proliferation and apoptosis involve
a series, or cascade, of alterations of the state (such as phosphorylation) of a
number of molecules, ultimately changing the expression of key proteins
such as cyclins, cyclin dependent kinases (CDKs), or apoptosis regulatory
proteins that regulate cell growth and cell death. Sophisticated multi-
parametric analyses assessing in specific cell sub-populations the targeted
protein(s) and the therapeutic as well as the levels and molecular states, such
as phosphorylation, of key intermediate molecules in these pathways that are
regulated by, and downstream of, the targeted proteins will be required.
Further, since multiple regulatory pathways control growth and death,
cellular response to drug and emergence of resistance may not be restricted
to a single pathway. Thus, the cellular proliferative and apoptotic responses
regulated by these disrupted pathways along with the key endpoint
246 Clinical Flow Cytometry
regulatory molecules such as cyclins or CDKs will also need to be monitored
in the same cell sub-populations as part of these analyses.
Imatinib mesylate therapy50,51 for chronic myelogenous leukemia (CML)
patients represents one of the first successful examples of a paradigm in
which a genetic abnormality associated with a disease is identified followed
by identification of the gene(s) involved, the characterization of the protein,
and development of a new drug specifically targeting this aberrant protein as
a therapy. As such it may serve as a model in many respects for the
development of not only new drug therapies but for the tools that will be
needed to monitor these types of therapies. CML is a chronic leukemia
characterized by the accumulation of abnormal myeloid cells harboring the
Philadelphia chromosome.55,56 The Philadelphia chromosome represents a
translocation between chromosomes 9 and 22 leading to a fusion between
the BCR and c-ABL genes. This leads to the production of a fusion protein,
BCR-ABL kinase, having abnormal kinase activity, which plays a key role
in the development and progression of the disease.55-57 Imatinib mesylate
represents a kinase inhibitor developed as a therapeutic for the treatment of
CML.50,51 It has high specificity for inhibition of the BCR-ABL kinase,
although at higher concentrations it does show inhibition of other kinases
such as c-kit and PDGF receptor. Initial response to imatinib mesylate has
been spectacular with most patients rapidly achieving a complete clinical
remission.50,51 However, a very high percentage of these patients relapse, or
develop resistance to imatinib therapy.50,51,58
Several roles for cytometric (cell based) monitoring of these patients, and
as a model other kinase inhibitor treated patients as well, may arise.
Interestingly, during escalation of dose in imatinib mesylate clinical trials,
only limited toxicity was seen. Thus, the paradigm of determining toxic drug
levels during clinical trials and to use that information in assessing relevant
treatment dose is less applicable with imatinib mesylate. As mentioned
above, this may be a common theme as more rational targeted therapies
become available. An alternative that may provide an adjunct to determining
effective doses in vivo will be monitoring the phosphorylation of key
molecules that are regulated through phosphorylation by kinases targeted by
the drug used,59 in the case of imatinib, the BCR-ABL kinase. The
development of antibody reagents highly specific for phosphorylated
epitopes on signal transduction protein intermediates will permit one, in
single cell measurements, to functionally assess whether a kinase upstream
of the protein intermediate being measured is active or not. Immediate
downstream phosphorylation targets of the BCR-ABL kinase which may be
suitable monitors of BCR-ABL kinase activity include Crkl as well as
members of the Jak-Stat pathway, 60-65 specifically Stat1 and Stat5-- targets
which BCR-ABL kinase is able to directly phosphorylate independent of Jak
Clinical Flow Cytometry 247
activation.60,64 Additionally, the autophosphorylation site on BCR-ABL
could also be included. An example of the cytometric application of these
reagents in a model system highlighting their specificity for detection of
phosphorylated Stat5 (p-Stat5) is shown in Figure 10-2. These data
supporting the specificity of these reagents comes from stimulation of
peripheral T cells66 with the cytokines, IL-2 and IL-4. IL-2 activates the Jak-
Stat pathway leading to phosphorylation of a number of Stat molecules
including Stat5,67 and T cells stimulated with IL-2 show significant
increased staining for p-Stat566 as opposed to unstimulated controls or cells
stimulated with the cytokine IL-4, which also stimulates the Jak-Stat
pathway but primarily other stat molecules and not Stat5.67,68 In the specific
case of monitoring BCR-ABL kinase activity, we, in collaboration with
several other laboratories (Clinical Cytometry Consortium; Drs. Hedley,
Jacobberger, and Shankey), have developed multi-parametric assays to
measure p-Stat5 in clinical specimens. As a model for this application, K562
cells, a CML cell line constitutively expressing BCR-ABL kinase and
having high level of expression of p-Stat5,60 were spiked into normal
peripheral blood mononuclear cells.
248 Clinical Flow Cytometry
Figure 10-3 illustrates the ability to detect the high levels of p-Stat5 in
the spiked K562 cells based on their expression of CD15 and high level p-
Stat5, and to modulate that expression in vitro with imatinib mesylate
treatment. In general, coupling the measurement of P-Stat5 with
immunophenotypic markers to identify or enrich for the abnormal malignant
cells allows rapid determination in vivo of whether imatinib treatment has
inhibited BCR-ABL kinase activity specifically in the malignant cells
through assessment of p-Stat5 levels. The potential for this approach looks
extremely promising and this assay is currently being implemented in
several imatinib mesylate clinical trial centers. Recent articles by Hedley et
al59 and Nolan et al69 demonstrates the applicability of these types of assays
in other signaling pathways as well. Specifically, in the work of Hedley et al,
monitoring of p-Mek and p-Erk, signaling intermediates in the MAP kinase
pathway, in the presence or absence of a raf kinase inhibitor, points to the
potential for the application of similar approaches in monitoring of patients
during clinical trials of this inhibitor as well.
The importance of single cell measurements, as opposed to bulk
measurements on cell lysates, in assessing phosphorylation, or activity, of
Clinical Flow Cytometry 249
key molecules in signaling cascades was also noted. It was clear that in this
experimental setting the MAPK pathway was either on or off,59 a
determination that would have been difficult using methods that assess
whole cell lysates to measure the same endpoints. Whether this on/off
signaling at the single cell level is also true in more physiological settings
will be of great interest and these techniques provide the tools to directly
address that question. Thus, single cell measurements of signaling
mechanisms critical to clinical monitoring of patients (as discussed in more
detail below) may also be crucial to understanding the more basic biology of
signaling processes as well.
Although potentially important to understanding the biology of signaling
processes and the effects of the kinase inhibitors used, how important is the
determination of these phosphorylation events on a single cell basis versus
more traditional assays employing cell lysates in a clinical setting? Some
argument can be made that during overt disease, when the malignant
population is the predominant population present, bulk methods employing
lysis of all cells and measurement of phosphorylated targets such as p-Stat5
in Western analyses or other techniques may be adequate. However, as
patients respond, and the malignant population decreases, utilization of bulk
assessment techniques will less reflect the therapeutic impact on the
malignant cells and more reflect the impact on the normal cells in the
patient. In fact, when morphologic and clinical remission is reached, bulk
methods will predominantly be a reflection of only the normal cell response.
Bulk methods will rarely be effective in identification of rare malignant cells
that are resistant to drug therapy or in the sensitive, early detection of
resistant cells before overt relapse occurs. These are two arenas in which
flow cytometry based single cell measurements will be exquisitely sensitive
in assessing phosphorylation, or activation state, of multiple proteins in rare
cells. These measurements could also play a key role in selection of the most
appropriate co-therapy when resistance to an initial kinase inhibitor drug
arises.
Further, single cell measurement techniques will almost certainly be
important in understanding the mechanisms of drug resistance in individual
patients. These techniques afford a sensitive assessment of re-activation of a
signaling pathway despite presence of drug. For example, in development of
resistance to imatinib mesylate therapy, in general, one sees continued, or
recurrent, high level expression of p-Stat5 in the presence of imatinib
mesylate58 due to either amplification of BCR-ABL or mutation of that locus.
During remission, flow cytometry techniques to measure rare, resistant cells
emerging early in development of resistance will almost certainly be a more
sensitive technique for monitoring development of imatinib resistance.
However, whether this will be useful in management decisions of CML
250 Clinical Flow Cytometry
patients where all patients develop resistance and combination therapies are
becoming commonplace remains to be seen. Nonetheless, this paradigm
applies more generally and undoubtedly will play a role in this arena at some
level in not only imatinib but other targeted therapies as well.
With appropriate reagents, single cell measurements may be a useful
adjunct in acquiring an understanding of the development of resistance to
targeted therapies. Was resistance due to inhibition of drug uptake, or
alternatively, to other mechanisms of drug inhibition? Development of
resistance may in some instances be more complex than simply altering drug
uptake or development of mechanisms to inhibit drug action with subsequent
re-activation of the relevant pathway. Regulation of cell proliferation and
apoptosis occurs through a number of alternative, and in some cases
redundant, pathways. Thus, in some instances, development of resistance
will arise through cells utilizing alternative signaling pathways to the one
which is blocked. Multi-parametric approaches coupling measurement of
specific inhibition of one pathway as described above along with
measurement of other relevant regulatory pathways and functional endpoint
measurements of cell proliferation, cell regulatory molecules (cyclins, cyclin
dependant kinase, etc), apoptosis, or apoptosis regulatory molecules may be
useful in identifying emergence of resistance even when the therapeutic drug
is still effectively blocking its target molecule. The ability to utilize these
techniques to simultaneously measure multiple pathways has clearly been
demonstrated.69 In addition, knowing what these responses are and
expression of which molecules have been altered may point to the
mechanism of resistance, to what other pathways may need to be monitored,
and to potentially appropriate additional therapies.
In addition, particularly when one moves outside of the realm of
hematopoietic malignancies, only rarely are single kinase abnormalities
going to be the primary, or only, abnormality responsible for disease.52 Even
in the setting of CML, these techniques may be key in assessing the effects
of combination therapies used with imatinib mesylate and helping to
determine on a patient specific basis what the most effective combination of
treatments may be. Although most CML patients in chronic phase respond to
imatinib, response in accelerated and blast crisis phase is more
heterogeneous. Before initiation of therapy, assessment of in vitro response
to imatinib may be useful in determining which of these patients are likely to
respond to imatinib in vivo.
Another exciting arena over the last several years has been development
of gene therapy options.70-73 Insertion and expression of specific genes into
abnormal cells designed to functionally correct the defect in these cells is an
exciting prospect. Initial forays into this arena have meet with some well
publicized problems, nonetheless, this approach remains promising and is
Clinical Flow Cytometry 251
rapidly evolving. The potential heterogeneity in expression of an inserted
foreign gene into the abnormal cells and its effectiveness in restoring normal
function or overcoming the effects of an abnormal protein lends itself to
cytometric techniques facilitating evaluation of transfected gene expression
and its effects on specific regulatory pathways and in specific cell sub-
populations that would be difficult by other technologies. Flow cytometric
approaches will play a role in the process of generation of transfected cells
for these therapeutic approaches. For some time, flow cytometry approaches
have been employed to measure the expression level of a transfected or
transduced gene into a cell.74-80 Sorting of cells can also be used in lieu of
drug selection to establish stable transfected cell lines as well as for selection
of cells having a specific expression level of a transfected gene (protein)
from the heterogeneous expression typically seen to generate cell lines
having homogeneous, typically high, expression levels.81
Far more exciting, however, is an elegant example of the use of flow
cytometry to monitor the effectiveness in correcting a regulatory defect by
induction of a normal transfected gene, in this case wild-type p53, into a
background with abnormal expression using an adenovirus delivery system
recently shown by Jaccobberger et al.82 Not only did these approaches prove
to be a sensitive way of confirming successful, high expression of the wild-
type p53, but more importantly, the sophisticated multi-parametric
approaches were able to clearly show that this protein was functionally
interacting with other cellular proteins to restore normal p53 function in
these cells. It is likely that cytometric approaches will play a significant role
not only in the laboratory aspects of generation and characterization of these
therapeutic agents but also in the monitoring of successful function and
maintenance of these cells in the patient.
5. SUMMATION
Over the last twenty years, cytometric evaluation of hematopoietic
malignancies has evolved from an adjunctive test which was secondary in
the diagnostic work-up, typically following by several days the primary
diagnostic work-up, to one which although still adjunctive to morphology is
now a key aspect of the primary diagnostic package. The maturing of the
technology in this setting is satisfying; however, more exciting are the
glimpses of the next evolution of this technology in a clinical setting,
perhaps first to be seen in the realm of hematopoietic malignancies. Rapid
movement of cytometric approaches into the arena of therapeutic
management and monitoring of patients is being seen. This movement
involves more complex multi-parametric approaches than previously have
252 Clinical Flow Cytometry
been employed in most clinical cytometry laboratories with five, six, seven,
or even more color analyses finding their way into the clinical setting. These
developments and needs are going to require cytometry technologists with
extensive experience and training along with laboratory directors,
pathologists, and clinicians who are knowledgeable about the analyses along
with interpretation and application of the results. Interpretation of these
experimental results will require sophisticated cytometry skills as well as a
detailed knowledge of the signaling and regulatory pathways in which the
measured molecules function. This is a very exciting time in clinical
cytometry, perhaps the most exciting time yet, but a challenge remains in
providing the appropriate training mechanisms and tools to ensure that these
analyses can be adequately performed in a wide range of clinical
laboratories.
REFERENCES
1. Jennings CD, Foon KA. Recent advances in flow cytometry: Application to the diagnosis
of hematologic malignancy. Blood 1997; 90:2863-2892.
2. Peterson LC, Goolsby C. Flow cytometric immunophenotyping of hematological
malignancies involving the blood and bone marrow. A Review. Curr Diag Path
1997;4:187-195.
3. Liu Z. Hultin LE. Cumberland WG. Hultin P. Schmid I. Matud JL. Detels R. Giorgi JV.
Elevated relative fluorescence intensity of CD38 antigen expression on CD8+ T cells is a
marker of poor prognosis in HIV infection: results of 6 years of follow-up. Cytometry.
1996;26:1-7.
4. Del Poeta G, Maurillo L, Venditti A, Buccisano F, Epiceno AM, Capelli G, Tamburini A,
Suppo G, Battaglia A, Del Principe MI, Del Moro B, Masi M, Amadori S. Clinical
significance of CD38 expression in chronic lymphocytic leukemia. Blood 2001;98:2633-
2639.
5. Damle RN, Wasil T, Fais F, Ghiotto F, Valetto A, Allen SL, Buchbinder A, Budman D,
Dittmar K, Kolitz J, Lichtman SM, Schulman P, Vinciguerra VP, Rai KR, Ferrarini M,
Chiorazzi N. Ig V gene mutation status and CD38 expression as novel prognostic
indicators in chronic lymphocytic leukemia. Blood 1999;94:1840-1847.
6. Hamblin TJ, Davis Z, Gardiner A, Oscier DG, Stevenson FK. Unmutated Ig genes are
associated with a more aggressive form of chronic lymphocytic leukemia. Blood
1999;94:1848.
7. Crespo M, Bosch F, Villamor N, Bellosillo B, Colomer D, Rozman M, Marce S, Lopez-
Guillermo A, Campo E, Montserrat E. ZAP-70 expression as a surrogate for
immunoglobulin-variable-region mutations in chronic lymphocytic leukemia. N Eng J
Med 2003;348:1764-1775.
8. Nolan GP. Multiparameter Kinase Profiling in Primary and Cancer Cells. Presented at the
AACR-NCI-EORTC International Conference On Molecular Targets and Cancer
Therapeutics. Discovery, Biology and Clinical Applications. Boston, MA November,
2003.
Clinical Flow Cytometry 253
9. Douglas VK, Gordon LI, Goolsby CL, White CA, Peterson LC. Lymphoid aggregates in
bone marrow mimic residual lymphoma after Rituximab therapy for non-Hodgkin
lymphoma. Am J Clin Pathol 1999;112:844-853.
10.Illidge TM, Bayne MC. Antibody therapy of lymphoma. Exp Opin Pharmacother
2001;2:953-961.
11.Syrigos KN, Pliarchopoulou K, Harrington KJ. The development of monoclonal antibody
therapy in leukemias. Hybridoma 2001;20:145-148.
12. White CA, Weaver RL, Grillo-Lopez AJ. Antibody-targeted immunotherapy for treatment
of malignancy. Annu Rev Med 2001;52;125-145.
13.Siegel AB, Goldenberg DM, Cesano A, Coleman M, Leonard JP. CD22-directed
monoclonal antibody therapy for lymphoma. Sem in Oncology 2003;30:457-64.
14.Flynn JM, Byrd JC. Campath-1H monoclonal antibody therapy. Curr Opin Oncol
2000;12:574-581.
15.Koon HB, Junghans RP. Anti-CD30 antibody-based therapy. Curr Opin Oncol
2000;12:588-593.
16.Radich J, Sievers E. New developments in the treatment of acute myeloid leukemia.
Oncology 2000;14:125-131.
17.Ruffner KL, Matthews DC. Current uses of monoclonal antibodies in the treatment of
acute leukemia. Sem Oncol 2000;27:531-539.
18.Sievers EL. Targeted therapy of acute myeloid leukemia with monoclonal antibodies and
immunoconjugates. Can Chemotherapy & Pharmacology 2000;46:S18-22.
19.Foss FM. Interleukin-2 fusion toxin: Targeted therapy for cutaneous T cell lymphoma.
Ann New York Acad Sci 2001;941:166-176.
20.Cinti P, Cociolo P, Evangelista B, Orlandini AM, Bruzzone P, Renna Molajoni E,
Cortesini R. OKT3 prophylaxis in kidney transplant recipients: Drug monitoring by flow
cytometry. Transplantation Proc 1996;28:3214-3216.
21.Hammond EA, Yowell RL, Greenwood J, Hartung L, Renlund D, Wittwer C. Prevention
of adverse clinical outcome by monitoring of cardiac transplant patients for murine
monoclonal CD3 antibody (OKT3) sensitization. Transplantation 1993;55:1061-1063.
22.Harford AM, Shopp GM, Ashmore LM, Seppelt JE, Mosdell DM, Gibel LJ, Sterling WA.
OKT3-treated kidney transplant patients: Monitoring of effects on peripheral blood
mononuclear cells by using two-color flow cytometry. Transplantation Proc 1988;20:245-
248.
23.Lim VL, Gumbert M, Garovoy MR. A flow cytometric method for the detection of the
development of antibody to Orthoclone OKT3. J Immunol Meth 1989;121:197-201.
24.McIntyre JA, Kincade M, Higgins NG. Detection of IGA anti-OKT3 antibodies in OKT3-
treated transplant recipients. Transplantation 1996;61:1465-1469.
25.McLaughlin P, Grillo-Lopez AJ, Link BK, Levy R, Czuczman MS, Williams ME,
Heyman MR, Bence-Bruckler I, White CA, Cabanillas F, Jain V, Ho AD, Lister J, Wey K,
Shen D, Dallaire BK. Rituximab chimeric anti-CD20 monoclonal antibody therapy for
relapsed indolent lymphoma: Half of patients respond to a four-dose treatment program. J
Clin Oncol 1998;16:2825-2833.
26.Reff ME, Carner K, Chambers KS, Chinn PC, Leonard JE, Raab R, Newman RA, Hanna
N, Anderson DR. Depletion of B-cells in vivo by a chimeric mouse human monoclonal
antibody to CD20. Blood 1994;83:435-445.
27.Rawstron AC, Kennedy B, Evans PA, Davies FE, Richards SJ, Haynes AP, Russell NH,
Hale G, Morgan GJ, Jack AS, Hillmen P. Quantitation of minimal disease levels in
chronic lymphocytic leukemia using a sensitive flow cytometric assay improves the
prediction of outcome and can be used to optimize therapy. Blood 2001;98:29-35.
254 Clinical Flow Cytometry
28.DelleKarth G, Laczika K, Scholten C, Lechner K, Jaeger U. Clearance of PCR-detectable
lymphoma cells from the peripheral blood, but no bone marrow after therapy with
Campath-1H. Am J Hematol 1995;50:146-152.
29.Lim SH, Davey G, Marcus R. Differential response in a patient treated with Campath-1H
monoclonal antibody for refractory non-Hodgkin lymphoma. Lancet 1993;341:432-433.
30.Poynton CH, Mort D, Maughan TS. Adverse reactions to Campath-1H monoclonal
antibody. Lancet 1993;341;1037.
31.Nabhan C, Tallman MS, Riley MB, Fitzpatrick J, Gordon LI, Gartenhaus R, Kuzel T,
Siegel R, Rosen ST. Phase I study of Rituximab and Campath-1H in patients with
relapsed or refractory chronic lymphocytic leukemia. Abst. Blood 2001;98:365a.
32. Lal A, Nabhan C, Rosen S, Goolsby C, Peterson L. Peripheral blood (PB), bone marrow
(BM) and flow cytometric (FC) changes in patients with chronic lymphocytic leukemia
(CLL) treated with Campath-1H and Rituximab. Presented at USCAP meeting, Chicago,
February 2002.
33.Davis AT. Monoclonal antibody-based therapy of lymphoid neoplasms: What’s on the
horizon? Sem Hematol 2000;37:34-42.
34.Bukowski RM. Cytokine therapy for metastatic renal cell carcinoma. Sem Urol Oncol
2001;19:148-154.
35.Graca L, Honey K, Adams E, Cobbold SP, Waldmann H. Cutting edge: Anti-CD 154
therapeutic antibodies induce infectious transplantation tolerance. J Immunol
2000; 165:4783-4786.
36.Hale G, Jacobs P, Wood L, Fibbe WE, Barge R, Novitzky N, Toit C, Abrahams L,
Thomas V, Bunjes D, Duncker C, Wiesneth M, Selleslag D, Hidajat M, Starobinski M,
Bird P, Waldmann H. CD52 antibodies for prevention of graft-versus-host disease and
graft rejection following transplantation of allogeneic peripheral blood stem cells. Bone
Marrow Transplantion 2000;26(1):69-76.
37.Issacs JD, Greer S, Sharma S, Symmons D, Smith M, Johnston J, Waldmann H, Hale G,
Hazleman BL. Morbidity and mortality in rheumatoid arthritis patients with prolonged
and profound therapy-induced lymphopenia. Arth Rheum 2001;44:1998-2008.
38.Kottaridis PD, Milligan DW, Chopra R, Chakraverty RK, Chakrabarti S, Robinson S,
Peggs K, Verfuerth S, Pettengell R, Marsh JC, Schey S, Mahendra P, Morgan GJ, Hale G,
Waldmann H, de Elvira MC, Williams CD, Devereux S, Linch DC, Goldstone AH,
Mackinnon S. In vivo Campath-1H prevents graft-versus-host disease following non-
myeloablative stem cell transplantation. Blood 2000;96:2419-2425.
39.Mitsuyasu RT. The potential role of interleukin-2 in HIV. AIDS 2001;15:S22-27.
40.Philip PA, Flaherty LE. Biochemotherapy for melanoma. Curr Oncol Rep 2000;2:314-
321.
41.Ratanatharathorn V, Ayash L, Lazarus HM, Fu J, Uberti JP. Chronic graft-versus-host
disease: clinical manifestation and therapy. Bone Marrow Trans 2001;28:121-129.
42.Simpson D. New developments in the prophylaxis and treatment of graft versus host
disease. Exp Opin Pharmacother 2001;2:1109-1117.
43.Smith KA. Low-dose daily interleukin-2 immunotherapy: Accelerating immune
restoration and expanding HIV-specific T-cell immunity without toxicity. AIDS
2001;15:S28-35.
44.Zecca M, DeStefano P, Nobili B, Locatelli F. Anti-CD20 monoclonal antibody for the
treatment of severe, immune-mediated, pure red cell aplasia and hemolytic anemia. Blood
2001;97:3995-3997.
Clinical Flow Cytometry 255
45.Mathas S, Rickers A, Bommert K, Dorken B, Mapara MY. Anti-CD20- and B-cell
receptor-mediated apoptosis: Evidence for shared intracellular signaling pathways.
Cancer Res 2000;60:7170-7176.
46.Golay J, Lazzari M, Facchinetti V, Bernasconi S, Borleri G, Barbui T, Rambaldi A,
Introna M. CD20 levels determine the in vitro susceptibility to rituximab and complement
of B-cell chronic lymphocytic leukemia: further regulation by CD55 and CD59. Blood
2001;98:3383-3389.
47. Huh YO, Keating MJ, Saffer HL, Jilani I, Lerner S, Albitar M. Higher levels of surface
CD20 expression on circulating lymphocytes compared with bone marrow and lymph
nodes in B-cell chronic lymphocytic leukemia. Amer J Clin Path 2001;116:437-43.
48.Treon SP, Shima Y, Preffer FI, Doss DS, Ellman L, Schlossman RL, Grossbard ML,
Belch AR, Pilarski LM, Anderson KC. Treatment of plasma cell dyscrasias by antibody-
mediated immunotherapy. Sem Oncol 1999;26:97-106.
49.Venugopal P, Sivaraman S, Huang XK, Nayini J, Gregory SA, Preisler HD. Effects of
cytokines on CD20 antigen expression on tumor cells from patients with chronic
lymphocytic leukemia. Leuk Res 2001;25:99-100.
50. Brunstein CG, McGlave PB. The biology and treatment of chronic myelogenous
leukemia. Oncology 2001; 15:23-31.
51. Maslak P, Scheinberg D. Targeted therapies for the myeloid leukaemias. Exp Opin In vest
Drugs 2000;9:1197-1205.
52.Druker BJ, Lydon NB. Lessons learned from the development of an Abl tyrosine kinase
inhibitor for chronic myelogenous leukemia. J Clin Invest 2000;105:3-7.
53.Al-Obeidi FA, Lam KS. Development of inhibitors for protein tyrosine kinases.
Oncogene 2000; 19:5690-5701.
54.Seidel HM, Lamb P, Rosen J. Pharmaceutical intervention in the JAK/STAT signaling
pathway. Oncogene 2000; 19:2645-2656.
55.Blennerhasset GT, Furth M, Anderson A, Burns JP, Chaganti RS, Blick M, Talpaz M, Dev
VG, Chan LC, Wiedemann LM. Clinical evaluation of a DNA probe assay for the
Philadelphia (Ph1) translocation in chronic myelogenous leukemia. Leukemia
1988:2:648-657.
56.Sawyers CL. The bcr-abl gene in chronic myelogenous leukaemia. In: Cancer Surveys,
Vol 15, Oncogenes in the Development of Leukaemia, Imperial Cancer Research Fund, pp
37-51, 1992.
57.Van Etten RA. Malignant transformation by abl and bcr/abl. In: Oncogenes and tumor
suppressor genes in human malignancies, pp 167-192, 1993.
58.Gorre ME, Mohammed M, Ellwood K, Hsu N, Paquette R, Rao PN, Sawyers CL. Clinical
resistance to STI-571 cancer therapy caused by BCR-ABL gene mutation or amplification.
Science 2001 ;293:876-880.
59.Chow S, Patel H, Hedley DW. Measurement of MAP kinase activation by flow cytometry
using phospho-specific antibodies to MEK and ERK: Potential for pharmacodynamic
monitoring of signal transduction inhibitors. Comm Clin Cytometry 2001 ;46:72-78.
60.Carlesso N, Frank DA, Griffin JD. Tyrosyl phosphorylation and DNA binding activity of
signal transducers and activators of transcript (STAT) proteins in hematopoietic cell lines
transformed by Bcr/Abl. J Exper Med 1996;183:811-820.
61.Chai SK, Nichols GL, Rothman P. Constitutive activation of JAKs and STATs in BCR-
Abl-expressing cell lines and peripheral blood cells derived from leukemic patients. J
Immunol 1997;159:4720-4728.
62. Coffer PJ, Koenderman L, de Groot RP. The role of STATs in myeloid differentiation and
leukemia. Oncogene 2000; 19:2511 -2522.
256 Clinical Flow Cytometry
63. Frank DA. STAT signaling in the pathogenesis and treatment of cancer. Molec Med
1999;5;432-456.
64.Ilaria RL Jr, Van Etten RA. P210 and induce the tyrosine phosphorylation
and DNA binding activity of multiple specific STAT family members. J Biol Chem
1996:271:31704-31710.
65.Nieborowska-Skorska M, Wasik MA, Slupianek A, Salomoni P, Kitamura T, Calabretta
B, Skorski T. Signal transducer and activator of transcription (STAT)5 activation by
BCR/ABL is dependent on intact Src homology (SH)3 and SH2 domains of BCR/ABL
and is required for leukemogenesis. J Exp Med 1999;189:1229-1242.
66.Jacobberger J, Sramkowski R, Feisa P, Ye P, Gottlieb M, Hedley D, Shankey T, Smith B,
Paniagua M, Goolsby C. Immunoreactivity of stat5 phosphorylated on tyrosine 694 as a
cell-based measure of Bcr/Abl kinase activity. Cytometry 54A, 75-88 2003.
67.Lin J-X, Migone T-S, Tsang M, Friedmann M, Weatherbee JA, Zhou L, Yamauchi A,
Bloom ET, Mietz J, John S, Leonard WJ. The role of shared receptor motifs and common
stat proteins in the generation of cytokine pleiotropy and redundancy by IL-2, IL-4, IL-7,
1L-13 and IL-15. Immunity 1995;2:331-339.
68.Jiang H, Marris MB, Rothman P. IL-4/IL-13 signaling beyond JAK/STAT. J All Clin
Immunol 2000;105:1063-1070.
69. Perez OD, Nolan GP. Simultaneous measurement of multiple active kinase states using
polychromatic flow cytometry. Nature Biotechnology 2002 ;20:155-62.
70. Brenner MK. Gene transfer and the treatment of haematological malignancy. J Int Med
2001:249:345-358.
71.Cusack JC Jr, Tanabe KK. Cancer gene therapy. Surg Oncol Clin North Am 1998;7:421-
469.
72.Pandha HS, Martin LA, Rigg AS, Ross P, Dalgleish AG. Oncological applications of
gene therapy. Curr Opin Invest Drugs 2000:1:122-134.
73.Roskrow MA, Gansbacher B. Recent developments in gene therapy for oncology and
hematology. Crit Rev Oncol-Hematol 1998:28:139-151.
74.Fiering SN, Roederer M, Nolan GP, Micklem DR, Parks DR, Herzenberg LA. Improved
FACS-Gal: Flow cytometric analysis and sorting of viable eukaryotic cells expressing
reporter gene constructs. Cytometry 1991;12:291-301.
75.Jasin M, Zalamea P. Analysis of Escherichia coli beta-galactosidase expression in
transgenic mice by flow cytometry of sperm. Proc Natl Acad Sci USA 1992:89:10681-
10685.
76.Klein D, Indraccolo S, von Rombs K, Amadori A, Salmons B, Gunzburg WH. Rapid
identification of viable retrovirus-transduced cells using the green fluorescent protein as a
marker. Gene Ther 1997;4:1256-1260.
77.Mazurier F, Moreau-Gaudry F, Maguer-Satta V, Salesse S, Pigeonnier-Lagarde V, Ged C,
Belloc F, Lacombe F, Mahon FX, Reiffers J, de Verneuil H. Rapid analysis and efficient
selection of human transduced primitive hematopoietic cells using the humanized S65T
green fluorescent protein. Gene Ther 1998:5:556-562.
78.Puchalski RB, Manoharan TH, Lathrop AL, Fahl WE. Recombinant glutathione S-
transferase (GST) expressing cells purified by flow cytometry on the basis of a GST-
catalyzed intracellular conjugation of glutathione to monochlorobimane. Cytometry
1991:12:651-665.
79.Wiechen K, Zimmer C, Dietel M. Selection of a high activity c-erbB-2 riboxyme using a
fusion gene of c-erbB-2 and the enhanced green fluorescent protein. Cancer Gene Ther
1998;5:45-51.
Clinical Flow Cytometry 257
80.Wittrup KD, Bailey JE. A single-cell assay of beta-galactosidase activity in
Saccharomyces cerevisiae. Cytometry 1988;9:394-404.
81. Van Tendeloo VF, Ponsaerts P, Van Broeckhoven C, Berneman ZN, Van Bockstaele DR.
Efficient generation of stably electrotransfected human hematopoietic cell lines without
drug selection by consecutive FAC sorting. Cytometry 2000;41:31-35.
82. Jacobberger JW, Sramkoski RM, Zhang D, Zumstein LA, Doerksen LD, Merritt JA,
Wright SA, Shults KE. Bivariate analysis of the p53 pathway to evaluate Ad-p53 gene
therapy efficacy. Comm Clin Cytometry 1999;38:201-213.