ArticlePDF Available

clinical profile of plasma cell leukemia at teritiary care hospital in kashmir,india

Authors:
  • SUPER-SPECIALITY HOSPITAL SRINAGAR

Abstract and Figures

INTRODUCTION: Plasma cell dyscrasias represent 1.4–2% of all malignancies and among hematologic malignancies; it constitutes 10% of the tumors. Plasma cell dyscrasias are composed of multiple myeloma (MM), and primary and secondary plasma cell leukemia. MATERIALS AND METHODS: Primary plasma cell leukemia (pPCL) is a rare and aggressive disease, representing 1–4% of plasma cell dyscrasias. RESULTS: The prognosis of this is very poor with median survival of 8–11 months in different reported series. DISCUSSION: We are reporting a study from our hospital over a period of 8 years and 9 months, in which pPCL was found in 1.8% of MM patients, with slight male predominance and earlier age than MM, and all had high disease burden with high lactate dehydrogenase (LDH), β2-microglobulin, and plasmacytosis. CONCLUSION: This disease had very aggressive behavior, especially with light chain lambda disease, and all patients succumbed within 8 months of treatment.
Content may be subject to copyright.
Clinical Profile of Plasma Cell
Leukemia at Tertiary Care Hospital in
Kashmir, India
Javvid Muzamil, Shiekh A. Aziz, Gull M. Bhat, Abdul R. Lone, Shuaeb Bhat
1
,
Firdousa Nabi
2
Abstract:
INTRODUCTION: Plasma cell dyscrasias represent 1.42% of all malignancies and among
hematologic malignancies; it constitutes 10% of the tumors. Plasma cell dyscrasias are composed
of multiple myeloma (MM), and primary and secondary plasma cell leukemia.
MATERIALS AND METHODS: Primary plasma cell leukemia (pPCL) is a rare and aggressive
disease, representing 14% of plasma cell dyscrasias.
RESULTS: The prognosis of this is very poor with median survival of 811 months in different reported
series.
DISCUSSION: We are reporting a study from our hospital over a period of 8 years and 9 months, in
which pPCL was found in 1.8% of MM patients, with slight male predominance and earlier age than
MM, and all had high disease burden with high lactate dehydrogenase (LDH), β2-microglobulin, and
plasmacytosis.
CONCLUSION: This disease had very aggressive behavior, especially with light chain lambda
disease, and all patients succumbed within 8 months of treatment.
Keywords:
Lactate dehydrogenase (LDH), multiple myeloma (MM), per iodic acid schiff (P.A.S.), peripheral
blood film (PBF), primary plasma cell leukemia (pPCL), secondary plasma cell leukemia (sPCL),
Sudan black stain (S.B.)
Introduction
Primary plasma cell leukemia (pPCL) is
arareandaggressivediseasewitha
prevalence of only 14% of all plasma cell
malignancies.
[1-3]
It is defined by the
presence of >2×10
9
/l peripheral blood
plasma cells or plasmacytosis accounting
for >20% of the white cell count.
[1]
The
prognosis of pPCL is very poor with a
median overall survival (OS) of 811
months.
[1,4]
However, the outcome of
pPCL has improved by the introduction
of autologous stem cell transplantation as
well as novel agents like bortezomib.
[1,4-7]
Compared to multiple myeloma (MM), the
presenting signs and symptoms of pPCL
have a more rapid onset with higher
tendency of hyper-metabolic symptoms
(weight loss, fever, sweating, fatigue, and
weakness), extra-medullary manifestations,
hypercalcemia, renal involvement, bone
marrow failure, and higher β2-
microglobulin but rarely osteolytic bone
lesions.
[7]
Furthermore, the presence of
poor-risk cytogenetic alterations known
from MM is markedly higher.
[1,4,8]
PCL is
classified as primary when it presents de
Access this article online
Quick Response Code:
Website:
www.jahjournal.org
DOI:
10.4103/1658-5127.192988
Departments of Medical
Oncology
1
Clinical Hematology,
Sher I Kashmir Institute of
Medical Sciences, Soura
2
Department of
Prosthodonsia, Govt.
Dental College, Srinagar,
Jammu and Kashmir,
India
Address for
correspondence:
Dr. Javvid Muzamil, MD
(Internal Medicine), DM
(Scholar), Department of
Medical Oncology, Married
Hostel, Room Number
F16, Sher I Kashmir
Institute of Medical
Sciences, Soura, Srinagar
- 190 011, Jammu and
Kashmir, India.
E-mail:
Javvidmd@gmail.com
This is an open access article distributed under the terms
of the Creative Commons Attribution-NonCommercial-
ShareAlike 3.0 License, which allows others to remix,
tweak, and build upon the work noncommercially, as
long as the author is credited and the new creations
are licensed under the identical terms.
For reprints contact: reprints@medknow.com
How to cite this article: Muzamil J, Aziz SA, Bhat
GM, Lone AR, Bhat S, Nabi F. Clinical profile of
plasma cell leukemia at Tertiary Care Hospital in
Kashmir, India. J Appl Hematol 2016;7:95-101.
©2016 Journal of Applied Hematology | Published by Wolters Kluwer - Medknow 95
Original Article
[Downloaded free from http://www.jahjournal.org on Wednesday, October 26, 2016, IP: 14.139.59.178]
novoin patients with no evidence of previous MM and
as secondary when it is observed as a leukemic
transformation of relapsed or refractory disease in
patients with previously recognized MM.
[9]
6070%
of PCL are primary, and the remaining 3040% are
secondary.
[10]
More recent data suggest that there is an
increasing incidence of secondary PCL, now
accounting for about 50% of the cases.
[4]
We are
reporting our experience about primary plasma cell
leukemia for almost 10-year period and found
different biology in our patients and very poor
prognosis.
Clinical study: It is discussed under following headings.
Materials and Methods
A study was conducted at our hospital with hemto-
oncological setup for the past 28 years. A cancer registry
is maintained for the past 15 years. Clinical survey was
conducted on plasma cell leukemia for the last 8 years
beginning from 2007 till September 2015. The first
patient was enrolled in January 2007 and the last
patient was enrolled in December 2014. All patients
of MM and de-novo plasma cell leukemia were enrolled
in the study. Patient characteristics were noted down
and all patients underwent complete hemogram with
peripheral blood film (PBF) examination. If PBF was
abnormal with presence of plasma cells with either 2000
cells/μl or 20% of total cell count, diagnosis of PCL was
established. Rest of the patients of MM were excluded
from the study with normal PBF. These patients further
underwent investigations like complete liver function
and kidney function tests, lactate dehydrogenase
(LDH), uric acid, acid base with electrolytes, β2-
microglobulin, skeletal survey, serum and urinary
electrophoresis with immunofixiation, light chain
assay in urine and serum, bone marrow aspiration
with biopsy, immunophenotyping, and cytogenetic.
Patients were treated with borteozomib-based
chemotherapy, and clinical outcome and OS were
noted down.
Study and Observation (Results)
In this study, a total of 441 patients with plasma cell
dyscrasias were enrolled, out of which 433 patients had
MM and eight patients had pPCL. The median age of
patients with MM observed was 50 years, with the
youngest patient being 35 years and the oldest being
70 years. The median age of patients with pPCL was
53 years, with the youngest patient being 45 years and
the oldest being 60 years. The male to female ratio in
MM was 4:1 and in pPCL it was 1.7:1. So the percentage
of pPCL was 1.89% among all plasma cell dyscrasias.
Further study was concentrated on patients with
pPCL.
Fifty percent of patients were hypertensive on
treatment and 25% were diabetic on oral
hypoglycemic agents. There was no other co-
morbidity observed in this series. These patients
presented with multiple symptoms and signs, which
are outlined in Table 1. The most common symptom
reported was fever followed by bone pain, which are in
contrary to MM and the most common sign observed
was pallor followed by hepato-splenomegaly and
lymphadenopathy, which are again rarely seen
in MM.
All patients were subjected to laboratory tests
in the form of complete hemogram, full chemistry,
acid base, and electrolytes, as depicted in Table 2. On
peripheral blood examination, all patients were
anemic with median HB of 9.2 g/dl, had lower total
leucocyte count (TLC) with median of 4.15 × 10
3
/μl,
and had very low platelet count with median of
20 × 10
3
/μl. On peripheral blood examination,
different degree of plasmacytosis was observed in
blood ranging from 22 to 38% and many were
having plasmablastic appearance [Figure 1], with
median plasma cell percentage of 28% and median
peripheral plasma cell count of 1162 per μl.
Skeletal survey was having multiple lytic lesions
[Figure 2] in every patient, with atypical finding in
one patient having splenic hypo-echoic lesions
[Figure 3], finding confirmed by aspiration having
plasma cell infiltration. On serum chemistry
examination, they all had normal kidney and liver
functions with lower serum proteins and
albumin, and raised LDH and gamma globulins
[Table 2].
Table 1: Symptomatology of pPCL
Symptom Percentage
Fever 100
Bone pain 40
Weight loss 30
Recurrent vomiting 25
Malaise 25
Cough 20
Dysuria 10
Sweating 10
Signs Percentages
Pallor 100
Hepatomegaly 60
Splenomegaly 50
Lymphadenopathy 45
Ascites 20
Jaundice 20
Petechie 10
Consolidation 10
Fracture 10
Muzamil, et al.: Clinical profile of plasma cell leukemia at tertiary care hospital
96 Journal of Applied Hematology | Vol 7, Issue 3, Jul-Sept 2016
[Downloaded free from http://www.jahjournal.org on Wednesday, October 26, 2016, IP: 14.139.59.178]
Then the patients were subjected to MM profile, which
is tabulated as Table 3. All patients had very high β2-
microglobulin with median value of 5.8 mg/l and
positive serum M spike with undetectable M spike in
urine on protein electrophoresis. But urine
immunofixation was positive for M spike in λregion.
On immunoglobulin assay, all immunoglobulinslike
Ig G, Ig A, and Ig M were markedly suppressed with
very high levels of lambda (λ) free light chains in serum.
The median value of lambda (λ) free light chains in
serum was 3250 mg/l. All our patients had significant
light chain disease and that too lambda chain.
Serum immune flow-cytometry of peripheral blood
revealed the plasma cell leukemia phenotype, which
is also depicted in flow diagrams [Figure 4]. It
showed high expression and positivity of CD38,
CD138, and its co-expression but it was having
very low expression and negativity of CD27, CD56,
and CD20. Different CD marker positivity intensity is
given in Table 4.
Bone marrow aspiration and biopsy revealed
plasmacytosis between 40 and 90%, with median
plasmacytosis of 70%. These bone marrow aspiration
slides revealed high degree of plasmacytosis with all
having plasmablastic appearance; rest of the cell lines
were markedly suppressed [Figure 5]. Under
immunohistochemistry, cells were per iodic acid Schiff
(P.A.S.), Sudan black stain (S.B.), and myeloperoxidase
Table 2: Laboratory investigation of pPCL patients:
Parameter Minimum
value
Median
value
Maximum
value
HB
gm/dl
8.0 09.2 10.0
TLC(10
3
/μl) 3.5 04.15 10.0
PLT(10
3
/μl) 17 20 40
Plasma cells in PBF/ μl 770 1162 3300
Percentage of plasma cells 22.0 28.0 33
Serum Calcium
mg/dl
7.5 08.29 10.5
Serum Phosphorous
mg/dl
2.4 03.40 4.5
Urea
mg/dl (Normal 15-40)
28 36.0 42
Creatinine
mg/dl (Normal 0.5 -1.5)
1.0 1.30 1.6
ALP (IU/dl)
(Normal 140-300)
190 240 300
ALT (IU/dl)
(Normal 20-40)
35 42 48
Total Bilirubin
mg/dl (Normal 0.5-1)
1.0 1.5 1.9
Serum Na
meq/dl
132 138 148
Serum K
meq/dl
3.4 4.2 4.5
PH 7.30 7.35 7.40
HCO
3 meq/dl
18 23 28
Albumin
g/dl
2.0 2.98 3.5
Protein
g/dl
4.0 4.80 6.0
Gamma globulin
g/dl
0.66 0.78 1.6
LDHU/L
(Normal 150-300)
380 760 950
Figure 1: PBF showing plasma cells with plasma blasts with prominent nucleolus
Figure 2: X-rays showing lytic lesions in both skull and femur
Figure 3: CECT abdomen showing hypoechoic lesions in spleen
Table 3: Multiple myeloma profile -
Parameter Minimum
value
Median
value
Maximum
value
β2 microglobulin
mg/l(Normal 0.7-1.8)
3.5 5.8 7.0
Serum M spike 0.20 0.26 0.75
Urine M spike Not
detected
Not
detected
Not
detected
Serum IgA
mg/dl(Normal 70-400)
55 70 220
Serum IgM
mg/dl(Normal 40-230)
28 33 100
Serum IgG
mg/dl (Normal 700-1600)
550 639 1000
Serum free λ
mg/l (Normal 5.71-26.30)
2600 3250 5500
Serum free Ќ
mg/dl (Normal 3.3-19.40)
6.5 8.5 16.5
Urine Immunofixiation Faint M
spike in λ
region
Faint M
spike in
λregion
Faint M
spike in λ
region
Muzamil, et al.: Clinical profile of plasma cell leukemia at tertiary care hospital
Journal of Applied Hematology | Vol 7, Issue 3, Jul-Sept 2016 97
[Downloaded free from http://www.jahjournal.org on Wednesday, October 26, 2016, IP: 14.139.59.178]
negative. On conventional cytogenetic karyotype, no
abnormality was noted down. Fluorescence In Situ
Hybridization (FISH) was not conducted due to
financial constraints.
Finally, based on all these parameters, the patients were
diagnosed as primary plasma cell leukemia expressing
lambda light chains exclusively with suppressed rest of
immunoglobulinsand the patients were put on weekly
intravenous borteozomib 2 mg and dexamethasone
40 mg based treatment along with other supportive
treatment in the form of tumor lysis prophylaxis,
antivirals, antimicrobials, and blood products. On
average, 15 weeks of treatment was received, with
longest treatment received being 24 weeks. Our
patients were not transplant candidates on the basis
of general performance. Interim assessment of disease
was done after 8 weeks of treatment with complete
hemogram, PBF, and myeloma profile. There was no
plasma cell seen on PBF examination and serum light
chains were reduced by 80%.
Most of our patients dropped general performance
from ECOG PS II to PS IV. They developed renal
failure and respiratory tract infection, which were
treated with broad-spectrum antibiotics, dialysis
support, and other intensive care support. All our
patients succumbed to different kinds of infection.
The median survival seen was 5 months and the
longest survival was 8 months. There was no
patient available after 8 months for disease
assessment.
So, we conclude that pPCL is a very rare disease in
plasma cell dyscrasias contributing to 1.89% of all
plasma cell dyscrasias, with slight male
predominance and earlier age than MM, and all had
highdiseaseburdenwithhighLDH,β2-microglobulin,
and plasmacytosis. This disease had very aggressive
behavior, especially with light chain lambda disease,
and all patients succumbed within 8 months of
treatment.
Discussion
pPCL is the most aggressive form of the plasma cell
dyscrasias. It is defined by the presence of >2×10
9
/l
peripheral blood plasma cells or plasmacytosis
accounting for >20% of the differential white cell count,
and it does not arise from preexisting MM.
[3,9]
We also used
the same criteria for diagnosis and all our patients had low
median TLC, with median plasma cell of 1160/μl(28%).
pPCL is rare, with only 14% of patients with MM
presenting as pPCL.
[1,2,4,11-14]
In addition, <1% of patients
presenting with extreme leukocytosis (>50 × 10
9
/l) are
diagnosed with PCL.
[15]
In our study as well, the total
patients with pPCL were 1.89% among 441plasma cell
dyscrasias which corresponds to other international
studies. None of our patients has TLC more than
10 × 10
3
/dl.
In PCL, tumor cells accumulate in the bone marrow
(BM) but also have an increased capacity to recirculate
in blood, with subsequent egression and formation of
extra-medullary disease. The dissemination of tumor
cells out of the BM is not only related to changes in
expression of adhesion molecules and chemokine
receptors but also to the presence of several
molecular aberrations, which contribute to BM
microenvironment-independent tumor growth,
inhibition of apoptosis, and escape from immune
surveillance. Interestingly, gene expression profiling
identifies pPCL as a distinct molecular entity among
myeloma samples.
[14]
Figure 4: Flow diagram showing plasma cell immunophenotype
Table 4: Immunoflow cytometery in pPCL-
Parameter Minimum
expression
Median
expression
Maximum
expression
CD 38 85.3 % 90.2 % 95.0 %
CD 138 82.5 % 86.2 % 89.9 %
Co expression
CD38/138
80.0 % 83.3 % 86.0 %
CD 45 0.00 % 0.00 % 0.00 %
CD 27 02.0 % 04.0 % 5.80 %
CD 56 0.03 % 0.70 % 0.90%
CD 20 5.00 % 6.80 % 10.0 %
Figure 5: Bone marrow showing immature plasma cells with plasma blasts
Muzamil, et al.: Clinical profile of plasma cell leukemia at tertiary care hospital
98 Journal of Applied Hematology | Vol 7, Issue 3, Jul-Sept 2016
[Downloaded free from http://www.jahjournal.org on Wednesday, October 26, 2016, IP: 14.139.59.178]
Compared to MM, tumor cells from patients with pPCL
and secondary plasma cell leukemia (sPCL) have
reduced expression of the adhesion molecules
NCAM (neural cell adhesion molecule/CD56) and
LFA-1 (leukocyte function-associated antigen-1),
which may contribute to the extra-medullary
accumulation of tumor cells in PCL.
[1,16-18]
The absence
of CD56 or LFA-1 is associated with reduced binding of
tumor cells to BM stromal cells.
[19,20]
Furthermore, loss of
CD56 results in increased production of matrix
metalloproteinase-9, which leads to destruction of the
basal membrane and extracellular matrix.
[21]
In our
study, all patients were negative for CD56 and CD27.
Patients with pPCL have a younger age at presentation
compared to patients with MM or sPCL.
[4,11,12,22,23]
However, their performance status at diagnosis is
usually worse,
[1]
which may be related to the more
advanced stage of disease (Durie-Salmon stage III:
8096%; International Staging System stage III:
6380%).
[1,6,7,11,24-26]
Extra-medullary involvement, such
as hepatomegaly, splenomegaly, lymphadenopathy,
leptomeningeal infiltration, or extra-medullary
plasmacytomas, is more frequent in pPCL,
[1,2,4,7,12-
14,23,25-27]
with extensive bone disease being more
common in patients with MM.
[1,4,25]
All our patients
were younger than patients with MM, with median
age being 53 years. Performance of all our patients was
also worse at presentation. However, the stage as per
Durie-Salmon staging system was II, but as per ISS, it
was III. Extra-medullary involvement was very high,
with hepatomegaly in 60%, splenomegaly in 50%, and
lymphadenopathy in 45% and none had central
nervous system (CNS) involvement. Bone
involvement was seen in less than half of the patients.
There are variousparameters which detect tumor burden
in PCL, for exampleplasma cell percentage, LDH, andβ2-
microglobulin. The median percentage of BM plasma
cells is significantly higher in pPCL than in MM
[1,4,26]
as
was seen in our patients as well. We documented bone
marrowplasmacytosisbetween 40and90%, with median
plasmacytosis of 70% and mostly plasmablastic
appearance. In addition, renal failure is more common
in pPCL, which can be partly explained by the higher
incidence of light-chain disease.
[1,4,14,23]
Furthermore,
hypercalcemia, anemia, thrombocytopenia, elevated
plasma cell labeling index, increased LDH, and gene
expression profile (GEP)-defined high-risk disease are
more frequent at presentation in pPCL compared to
MM.
[1,2,4,6,7,14,25,27]
In our series, none of our patients had
renal failure at the beginning but it developed in all
of them over the course of the disease. All our patients
had lambda light chain disease only, with high degree
of thrombocytopenia, moderate anemia and high
LDH.
Peripheral blood examination in pPCL shows circulating
tumor cells and typically a leuko-erythroblastic blood
picture in up to 67% of patients.
[4,25]
BM biopsy typically
demonstrates extensive BM involvement, disrupting
normal hematopoiesis. In some cases, tumor resembles
normal plasma cells, whereas in others, lympho-
plasmacytoid or immature plasma cells predominate.
[28-
30]
Our series also highlighted the presence of plasma
blastic or immature plasma cells both in blood and bone
marrow.
The most striking immune-phenotypic feature is
increased expression of CD20 and CD23, and down-
regulation of CD56 may be related to the high incidence
of t(11;14) in pPCL.
[1,31-33]
Tumor cells are positive for
CD38 and CD138 in both PCL and MM.
[1,16,28]
We also
demonstrated similar results in our series with high
expression of CD38/138 and loss of CD56.
There is a paucity of literature on the treatment of pPCL,
and no randomized trials have been reported exclusively
for patients with pPCL. The prognosis of pPCL after
conventional chemotherapy without novel agents is
poor, with median OS of 7months.
[1,4,6,12,23,30,34]
The
introduction of immune-modulatory drugs and
proteasome inhibitors has significantly improved the
survival of patients with MM.
[35,36]
Increasing evidence
suggests that these agents also improve outcome of
pPCL, but the benefit may be less pronounced
compared to classic MM. A retrospective analysis
performed by the Inter-groupe Francophone du
Myélome showed that patients with pPCL treated
with novel agents had a survival of 15 months
compared to 8 months for patients who did not receive
novel agents as part of their treatment.
[37]
Several case
reports and small case series suggest that bortezomib,
alone or in combination with other agents, is effective in
newly diagnosed pPCL and may also be active in
refractory pPCL or sPCL.
[5,34,38-43]
In pPCL, the efficacy of
combinations of novel agents, such as lenalidomide,
bortezomib, and dexamethasone (RVD),
[44]
bortezomib,
thalidomide, and dexamethasone (VTD),
[5,34,37]
or
melphalan, prednisone, bortezomib, and thalidomide
(VMPT),
[5]
appears very promising. In our series, we
treated all patients with borteozomib and
dexamethasone, and all were transplant-ineligible
based on their performance status. For initial 8 weeks
of therapy, all our patients responded evidence by
interim assessment, but subsequently all progressed
fast with deterioration in renal functions and
superadded infections. The median survival we
documented was 5 months and the longest survival of
8months.
There were some peculiarities in our patients. All were
young, and had moderate anemia with critical
Muzamil, et al.: Clinical profile of plasma cell leukemia at tertiary care hospital
Journal of Applied Hematology | Vol 7, Issue 3, Jul-Sept 2016 99
[Downloaded free from http://www.jahjournal.org on Wednesday, October 26, 2016, IP: 14.139.59.178]
thrombocytopenia. All had high LDH and β2-
microglobulin with high degree of plasmacytosis in
bone marrow and blood. All our patients had
lambda light chain disease with suppressed other
immunoglobulins. Response to treatment was very
short-lived and the median survival was lower than
evidenced by modern treatment strategies, partly due
to different genetics in our patients.
Conclusion
Primary plasma cell leukemia is a very aggressive
disease and should be picked up early and treated
aggressively. The authors have recommended that at
least 20% of plasma cells be seen in PBF, so as to
diagnose this condition. As aggressiveness of this
disease is concerned, these criteria need to be revised
and less threshold of monoclonal plasma cells
percentage be seen in PBF for its diagnosis. Further
work needs to be done for classifying this leukemia,
refining its staging and refining its treatment.
Financial support and sponsorship
Nil.
Conflicts of interest
There are no conflicts of interest.
References
1. García-Sanz R, Orfão A, González M, Tabernero MD, Bladé J,
Moro MJ, et al. Primary plasma cell leukemia: Clinical,
immunophenotypic, DNA ploidy, and cytogenetic
characteristics. Blood 1999;93:1032-7.
2. Dimopoulos MA, Palumbo A, Delasalle KB, Alexanian R.
Primary plasma cell leukaemia. Br J Haematol 1994;88:754-9.
3. Kyle RA, Maldonado JE, Bayrd ED. Plasma cell leukemia. Report
on 17 cases. Arch Intern Med 1974;133:813-8.
4. Tiedemann RE, Gonzalez-Paz N, Kyle RA, Santana-Davila R,
Price-Troska T, Van Wier SA, et al. Genetic aberrations and
survival in plasma cell leukemia. Leukemia 2008;22:1044-52.
5. DArena G, Valentini CG, Pietrantuono G, Guariglia R, Martorelli
MC, Mansueto G, et al. Frontline chemotherapy with bortezomib-
containing combinations improves response rate and survival in
primary plasma cell leukemia: A retrospective study from
GIMEMA multiple Myeloma Working Party. Ann Oncol
2012;23:1499-502.
6. Drake MB, Iacobelli S, van Biezen A, Morris C, Apperley JF,
Niederwieser D, et al. Primary plasma cell leukemia and
autologous stem cell transplantation. Haematologica 2010;95:
804-9.
7. Pagano L, Valentini CG, De Stefano V, Venditti A, Visani G,
Petrucci MT, et al. Primary plasma cell leukemia: A
retrospective multicenter study of 73 patients. Ann Oncol
2011;22:1628-35.
8. Avet-Loiseau H, Daviet A, Brigaudeau C, Callet-Bauchu E, Terré
C, Lafage-Pochitaloff M, et al. Cytogenetic, interphase, and
multicolor fluorescence in situ hybridization analyses in
primary plasma cell leukemia: A study of 40 patients at
diagnosis, on behalf of the Intergroupe Francophone du
Myélome and the Groupe Français de Cytogénétique
Hématologique. Blood 2001;97:822-5.
9. International Myeloma Working Group. Criteria for the
classification of monoclonal gammopathies, multiple myeloma
and related disorders: A report of the International Myeloma
Working Group. Br J Haemat 2003;121:749-57.
10. Bladé J, Kyle RA. Nonsecretory myeloma, immunoglobulin D
myeloma, and plasma cell leukemia. Hematol Oncol Clin North
Am 1999;13:1259-72.
11. Avet-Loiseau H, Roussel M, Campion L, Leleu X, Marit G, Jardel
H, et al. Cytogenetic and therapeutic characterization of
primary plasma cell leukemia: The IFM experience. Leukemia
2012;26:158-9.
12. Noel P, Kyle RA. Plasma cell leukemia: An evaluation of response
to therapy. Am J Med 1987;83:1062-8.
13. Vela-Ojeda J, García-Ruiz Esparza MA, Rosas-Cabral A, Padilla-
González Y, García-Chávez J, Tripp-Villanueva F, et al.
Intermediate doses of melphalan and dexamethasone are
better than vincristine, adriamycin, and dexamethasone (VAD)
and polychemotherapy for the treatment of primary plasma cell
leukemia. Ann Hematol 2002;81:362-7.
14. Usmani SZ, Nair B, Qu P, Hansen E, Zhang Q, Petty N, et al.
Primary plasma cell leukemia: Clinical and laboratory
presentation, gene-expression profiling, and clinical outcome
with Total Therapy protocols. Leukemia 2012;26:2398-405.
15. Hugo ZP, Perry MC, Steensma DP. Extreme leukocytosis in the
21st Century is often iatrogenic [abstract]. Blood (ASH Annual
Meeting Abstracts) 2008;112:5401.
16. Kraj M, Kopeć-Szlęzak J, Pogłód R, Kruk B. Flow cytometric
immunophenotypic characteristics of 36 cases of plasma cell
leukemia. Leuk Res 2011;35:169-76.
17. Pellat-Deceunynck C, Barillé S, Jego G, Puthier D, Robillard N,
Pineau D, et al. The absence of CD56 (NCAM) on malignant
plasma cells is a hallmark of plasma cell leukemia and of a special
subset of multiple myeloma. Leukemia 1998;12:1977-82.
18. Pellat-Deceunynck C, Barille S, Puthier D, Rapp MJ, Harousseau
JL, Bataille R, et al. Adhesion molecules on human myeloma
cells: Significant changes in expression related to malignancy,
tumor spreading, and immortalization. Cancer Res
1995;55:3647-53.
19. Barillé S, Collette M, Bataille R, Amiot M. Myeloma cells
upregulate interleukin-6 secretion in osteoblastic cells through
cell-to-cell contact but downregulate osteocalcin. Blood
1995;86:3151-9.
20. Schmidmaier R, Mandl-Weber S, Gaul L, Baumann P, Bumeder I,
Straka C, et al. Inhibition of lymphocyte function associated
antigen 1 by LFA878 induces apoptosis in multiple myeloma cells
and is associated with downregulation of the focal adhesion
kinase/phosphatidylinositol 3 kinase/Akt pathway. Int J
Oncol 2007;31:969-76.
21. Edvardsen K, Chen W, Rucklidge G, Walsh FS, Obrink B, Bock E.
Transmembrane neural cell-adhesion molecule (NCAM), but not
glycosyl-phosphatidylinositol-anchored NCAM, down-regulates
secretion of matrix metalloproteinases. Proc Natl Acad Sci U S A
1993;90:11463-7.
22. Ramsingh G, Mehan P, Luo J, Vij R, Morgensztern D. Primary
plasma cell leukemia: A Surveillance, Epidemiology, and End
Results database analysis between 1973 and 2004. Cancer
2009;115:5734-9.
23. Costello R, Sainty D, Bouabdallah R, Fermand JP, Delmer A,
Diviné M, et al. Primary plasma cell leukaemia: A report of 18
cases. Leuk Res 2001;25:103-7.
24. Kraj M, Kopeć-Szlęzak J, Pogłód R, Kruk B. Flow cytometric
immunophenotypic characteristics of plasma cell leukemia. Folia
Histochem Cytobiol 2011;49:168-82.
25. Woodruff RK, Malpas JS, Paxton AM, Lister TA. Plasma cell
leukemia (PCL): A report on 15 patients. Blood 1978;52:839-45.
26. ColovićM, JankovićG, SuvajdzićN, MilićN, DordevićV,
JankovićS. Thirty patients with primary plasma cell leukemia:
A single center experience. Med Oncol 2008;25:154-60.
27. Jiménez-Zepeda VH, Domínguez VJ. Plasma cell leukemia: A
rare condition. Ann Hematol 2006;85:263-7.
Muzamil, et al.: Clinical profile of plasma cell leukemia at tertiary care hospital
100 Journal of Applied Hematology | Vol 7, Issue 3, Jul-Sept 2016
[Downloaded free from http://www.jahjournal.org on Wednesday, October 26, 2016, IP: 14.139.59.178]
28. Johnson MR, Del Carpio-Jayo D, Lin P, Giralt S, Anderlini P,
Champlin RE, et al. Primary plasma cell leukemia: Morphologic,
immunophenotypic, and cytogenetic features of 4 cases treated
with chemotherapy and stem cell transplantation. Ann Diagn
Pathol 2006;10:263-8.
29. Saccaro S, Fonseca R, Veillon DM, Cotelingam J, Nordberg ML,
Bredeson C, et al. Primary plasma cell leukemia: Report of 17 new
cases treated with autologous or allogeneic stem-cell
transplantation and review of the literature. Am J Hematol
2005;78:288-94.
30. Bernasconi C, Castelli G, Pagnucco G, Brusamolino E. Plasma cell
leukemia: A report on 15 patients. Eur J Haematol Suppl
1989;51:76-83.
31. Walters M, Olteanu H, Van Tuinen P, Kroft SH. CD23 expression
in plasma cell myeloma is specific for abnormalities of
chromosome 11, and is associated with primary plasma cell
leukaemia in this cytogenetic sub-group. Br J Haematol
2010;149:292-3.
32. Buda G, Carulli G, Orciuolo E, Cannizzo E, Pelosini M, Galimberti
S, et al. CD23 expression in plasma cell leukaemia. Br J Haematol
2010;150:724-5.
33. Hundemer M, Klein U, Hose D, Raab MS, Cremer FW, Jauch A,
et al. Lack of CD56 expression on myeloma cells is not a marker
for poor prognosis in patients treated by high-dose
chemotherapy and is associated with translocation t(11;14).
Bone Marrow Transplant 2007;40:1033-7.
34. Musto P, Rossini F, Gay F, Pitini V, Guglielmelli T, DArena G,
et al. Efficacy and safety of bortezomib in patients with plasma
cell leukemia. Cancer 2007;109:2285-90.
35. Kumar SK, Rajkumar SV, Dispenzieri A, Lacy MQ, Hayman SR,
Buadi FK, et al. Improved survival in multiple myeloma and the
impact of novel therapies. Blood 2008;111:2516-20.
36. Richardson PG, Mitsiades C, Schlossman R, Munshi N, Anderson
K.New drugs for myeloma. Oncologist 2007;12:664-89.
37. Chaoui D, Leleu X, Roussel M, Royer B, Rubio MT, Ducastelle S,
et al. Has the prognostic of primary plasma cell leukemia
improved with new drugs? [abstract]. Blood (ASH Annual
Meeting Abstracts) 2009;114:3869.
38. Esparís-Ogando A, Alegre A, Aguado B, Mateo G, Gutiérrez N,
Bladé J, et al. Bortezomib is an efficient agent in plasma cell
leukemias. Int J Cancer 2005;114:665-7.
39. Katodritou E, Verrou E, Gastari V, Hadjiaggelidou C, Terpos E,
Zervas K. Response of primary plasma cell leukemia to the
combination of bortezomib and dexamethasone: Do specific
cytogenetic and immunophenotypic characteristics influence
treatment outcome? Leuk Res 2008;32:1153-6.
40. Grassinger J, Südhoff T, Andreesen R, Hennemann B. Complete
remission and successful stem cell mobilization after treatment of
refractory plasma cell leukemia with bortezomib. Ann Hematol
2006;85:132-3.
41. Libby E, Candelaria-Quintana D, Moualla H, Abdul-Jaleel M,
Rabinowitz I. Durable complete remission of primary plasma cell
leukemia with the bortezomib plus melphalan and prednisone
(VMP) regimen. Am J Hematol 2010;85:733-4.
42. Al-Nawakil C, Tamburini J, Bardet V, Chapuis N, Bourry E, Roux
C, et al. Bortezomib, doxorubicin and dexamethasone association
is an effective option for plasma cell leukemia induction therapy.
Leuk Lymphoma 2008;49:2012-4.
43. Lebon D, Gruson B, Garidi R, Dutel JL, Thevenot C, Choufi B,
et al. Bortezomib, doxorubicin and dexamethasone (PAD) prior to
high-dose melphalan/autologous stem cell transplantation for
newly diagnosed plasma cell dycrasias [abstract]. Blood (ASH
Annual Meeting Abstracts) 2009;114:4960.
44. Goyal M, Mohammad N, Palanki SD, Vaniawala SN. Primary
plasma cell leukemia with light chain secretion and multiple
chromosomal abnormalities: How successfully treated? A
case report with review of literature. Indian J Med Paediatr
Oncol 2010;31:96-100.
Muzamil, et al.: Clinical profile of plasma cell leukemia at tertiary care hospital
Journal of Applied Hematology | Vol 7, Issue 3, Jul-Sept 2016 101
[Downloaded free from http://www.jahjournal.org on Wednesday, October 26, 2016, IP: 14.139.59.178]
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Leukemia is one of the leading journals in hematology and oncology. It is published monthly and covers all aspects of the research and treatment of leukemia and allied diseases. Studies of normal hemopoiesis are covered because of their comparative relevance.
Article
Full-text available
To determine whether primary plasma cell leukemia (PPCL) remains a high-risk multiple myeloma feature in the context of contemporary therapy and gene-expression profiling (GEP), we reviewed records of 1474 patients with myeloma, who were enrolled in Total Therapy protocols or treated identically off protocol. A total of 27 patients (1.8%) were classified as having PPCL. As a group, these patients more often had low hemoglobin, high beta-2-microglobulin, high lactate dehydrogenase, low albumin and cytogenetic abnormalities. Among 866 patients with GEP results, the PPCL group more often had disease that was classified as high risk, and in CD-1 and MF molecular subgroups. Regardless of the therapeutic protocol, patients with PPCL had shorter median overall survival (OS; 1.8 years), progression-free survival (PFS; 0.8 years) and complete response duration (CRD; 1.3 years) than the remainder, whose clinical outcomes had improved markedly with successive protocols. Multivariate analyses of pretreatment parameters showed that PPCL was a highly significant independent adverse feature linked to OS, PFS and CRD. In GEP analyses, 203 gene probes distinguished PPCL from non-PPCL; the identified genes were involved in the LXR/RXR activation, inositol metabolism, hepatic fibrosis/hepatic stellate-cell activation and lipopolysaccharide/interleukin-1-mediated inhibition of RXR function pathways. Different treatment approaches building on these genomic differences may improve the grave outcome of patients with PPCL.Leukemia advance online publication, 18 May 2012; doi:10.1038/leu.2012.107.
Article
Background: Most reports of the causes of extreme leukocytosis (i.e., total white blood count (WBC) >50 × 109/L) have included relatively small numbers of cases or data from the era prior to FDA approval of filgrastim (recombinant G-CSF; February 1991), sargramostim (GM-CSF; March 1991), and pegfilgrastim (G-CSF conjugated to polyethylene glycol; January 2002). The contribution of myeloid growth factors to extreme leukocytosis in contemporary medical practice is unclear. Methods: We evaluated the medical records of all patients with extreme leukocytosis evaluated in the clinical laboratory of the University of Missouri-Columbia (UMC) Medical Center during the years 2000 to 2007. We collected complete blood count data, information on the etiology of the leukocytosis, demographic factors, and clinical complications. Results: We identified 371 unique patients who had at least one WBC >50 × 109/L during this 8-year period (43.7% females; median age 56 (range 0–92)). Review of the primary cause of the leukocytosis in each case revealed that, while the various subtypes of leukemia (n=165, 44.5% of cases) and leukemoid reactions to general medical disorders (n=155, 41.8%) still represent the most common etiologies of extreme WBC elevation, growth factor-associated leukocytosis (GFL) is common, accounting for 13.7% (n=51) of patients. Among the subset of patients with GFL, the median peak WBC count was 60.8 × 109/L (range, 50–114.7); 51% were female, and the median age was 52 years (range 0–85). The median absolute WBC differential was as follows: neutrophils 40.8 × 109/L, granulocytes 55.7, bands 7.1, lymphocytes 2.4, monocytes 1.3, eosinophils 0, and basophils 0. Most (84%) of the patients receiving growth factors were being treated for cancer, the most common being non-Hodgkin lymphoma (NHL, 19.6%), Hodgkin lymphoma (HL, 11.8%), breast cancer (9.8%), ovarian cancer (5.9%), melanoma (5.9%), and endometrial cancer (5.9%). The non-malignant diagnoses (n = 8) included autoimmune neutropenia, bruton’s agammagloblinemia with pneumonia, leukopenia of undefined etiology, preterm neutropenia/sepsis (n = 4), and neutropenia associated with HIV. 70.6% of patients with GFL were receiving filgrastim, 29.4% pegfilgrastim, and 0% sargramostim. 0% of patients were undergoing conditioning for stem cell transplant. While myeloid growth factors are generally well tolerated, extreme leukocytosis is concerning because it may be associated with serious adverse events such as spontaneous splenic rupture, leukostasis/hyperviscosity, and hemorrhage. Other diagnoses continue to be important contributors to extreme leukocytosis. Of the 165 patients with leukemia, 43% were diagnosed with CLL, 15.8% CML, 15.2% AML, 13.3% ALL, 3.6% CMML, 3.6% BCR/ABL-negative myeloproliferative disorders, and 3.6% NHL. Other diagnoses included plasma cell leukemia, undifferentiated leukemia, and an undefined lymphoproliferative disease with macroglobulinemia variant. The causes of non-clonal leukocytosis (leukemoid reactions) could be divided into four broad categories: infection (n=109, 70.3%), reaction to hemorrhage (n=15, 9.7%), paraneoplastic (n=11, 7.1%), and other miscellaneous causes (20, 12.9%). The most common infections causing extreme leukocytosis include sepsis (59, 54.1%), pneumonia (19, 17.4%), and Clostridium difficile-associated colitis (5, 4.6%). Paraneoplastic causes of leukocytosis were diverse and associated neoplasms included cancer of unknown primary (n = 2), cholangiocarcinoma, extranodal marginal zone B-cell lymphoma, HL, SCLC, NSCLC, lymphoplasmacytic lymphoma, melanoma, peripheral T-cell lymphoma, and squamous cell carcinoma of the penis. Conclusion: In the current era, extreme leukocytosis is often iatrogenic and associated with filgrastim or pegfilgrastim usage. WBC counts should be routinely monitored in patients receiving pegfilgrastim or filgrastim and the medication discontinued with evolving leukocytosis.
Article
3869 Poster Board III-805 Introduction The outcome of patients with plasma-cell leukemia (PCL) is poor. Avet-Loiseau reported on behalf the IFM, our first experience in PCL patients and showed that the median overall survival (OS) was 8 months (Avet-Loiseau, Blood, 2001). Since 1999, novel agents such as Thalidomide, Bortezomib (Velcade) or Lenalidomide (Revlimid) have been widely used in the treatment of multiple myeloma, both at the time of relapse or part of upfront therapy. Patients and methods In this retrospective analysis, we have looked at the outcome of PCL patients treated within the IFM since 1999 in order to study the impact of novel agents on survival. Results 31 cases, 20 males, 11 females, median age 55 years (34-78) were analyzed. Twenty one patients less than 65 years received high-dose therapy as part of frontline treatment : 19 autologous haematopoietic stem cell transplantation (HSCT) and 5 allogeneic transplantation. Novel agents were used part of induction therapy in 6 cases, at the time of relapse for 9 patients, for both induction and relapse in 16 cases. Thirteen patients received 1 novel agent, 11 received 2 and 7 patients received the 3 novel agents. The median number of lines of therapy was 2 (1 to 4). Bortezomib was used as up front treatment in 15 patients and at relapse for 9 patients. Overall response rate according the IMWG criterias was 70% (17/24) including 11 CR or VGPR (45%). PAD (Bortezomib, Adriamycin and Dexamethasone) and VTD (Bortezomib, Thalidomide, Dexamethasone) regimens provided the best response rates. Lenalidomide was used in 13 patients mostly at relapse. A response was obtained in 53% of patients including 2CR and 2 VGPR (30%). Nineteen patients were treated with Thalidomide-based regimens. Overall response rate was 52% (10/19) including 2 CR and 6 VGPR (31%). Overall, for the whole group of patients, the median progression-free survival was 8 months (0-26) and the median OS was 15 months (6-108). When comparing this survival with that described in our previous experience reported before 1999, we clearly showed that the use of novel agents improved the survival of patients with PCL. Conclusion In this retrospective study, novel agents improved the prognosis of P-PCL. Prospective IFM phase II studies are ongoing to confirm these results. Disclosures No relevant conflicts of interest to declare.
Article
The aim of this prospective study was to define the flow cytometric characteristics of simultaneously investigated bone marrow and peripheral blood plasma cells antigens expression in 36 plasma cell leukemia (PCL) patients. The immunophenotypic profile of plasma cells was determined with a panel of monoclonal antibodies. The antigen expression intensity was calculated as relative fluorescence intensity (RFI). Bone marrow plasma cells showed expression of particular antigens in the following proportion of cases: CD49d 100%, CD29 94%, CD54 93%, CD44 83%, CD56 60%, CD18 26%, CD11b 29%, CD11a 19%, CD117 27%, CD71 30%, CD126 100% and CD19 0%, while the expression of those antigens on peripheral blood plasma cells was present in the following percentage of patients: CD49d 100%, CD29 96%, CD54 93%, CD44 95%, CD56 56%, CD18 50%, CD11b 53%, CD11a 29%, CD117 26%, CD71 28%, CD126 100% and CD19 0%. The expression of CD54 was significantly higher than that of adhesion molecules belonging to the integrin b2 family: CD11a, CD18 and CD11b, on both bone marrow and peripheral blood cells (p < 0.01). Expression of CD18, CD11a and CD11b was differential between two cell compartments: lower on bone marrow and higher on peripheral blood cells. We found that plasma cells in the bone marrow of patients with plasma cell leukaemia showed significantly greater granularity and size than those in the peripheral blood (p = 0.0001 and p = 0.04, respectively). However, no differences in cell size or granularity were revealed between bone marrow plasma cells from patients with PCL and multiple myeloma. In conclusion, impaired expression of adhesion molecules such as CD11a/CD18 (LFA-1) or CD56 may explain hematogenic dissemination characterizing PCL. The following pattern of adhesion molecule expression according to the proportion of plasma cells expressing a given antigen in peripheral blood and bone marrow and arranged in diminishing order may be established: CD49d > CD44 > CD54 > CD29 > CD56 > CD18 > CD11b > CD11a. Immuno-phenotyping of plasma cells in PCL, as in multiple myeloma, might be useful in detecting minimal residual disease in cases with aberrant antigen expression and for selecting therapeutic agents towards specific membrane targets. (Folia Histochemica et Cytobiologica 2011; Vol. 49, No. 1, pp. 168–182)
Article
Proposals and rationale for revision of the World Health Organization diagnostic criteria for polycythemia vera, essential throm-bocythemia, and primary myelofibrosis: recommendations from an ad hoc international expert panel. Blood, 110, 1092–1097.