Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
2.
Blood ; 137(21): 2890-2901, 2021 05 27.
Article in English | MEDLINE | ID: mdl-33512480

ABSTRACT

B-cell maturation antigen (BCMA)-specific chimeric antigen receptor (CAR) T-cell therapies have shown efficacy in relapsed/refractory multiple myeloma (RRMM). Because the non-human originated antigen-targeting domain may limit clinical efficacy, we developed a fully human BCMA-specific CAR, CT103A, and report its safety and efficacy in a phase 1 trial. Eighteen consecutive patients with RRMM, including 4 with prior murine BCMA CAR exposures, were enrolled. CT103A was administered at 1, 3, and 6 × 106 CAR-positive T cells/kg in the dose-escalation phase, and 1 × 106 CAR-positive T cells/kg in the expansion cohort. The overall response rate was 100%, with 72.2% of the patients achieving complete response or stringent complete response. For the 4 murine BCMA CAR-exposed patients, 3 achieved stringent complete response, and 1 achieved a very good partial response. At 1 year, the progression-free survival rate was 58.3% for all cohorts and 79.1% for the patients without extramedullary myeloma. Hematologic toxicities were the most common adverse events; 70.6% of the patients experienced grade 1 or 2 cytokine release syndromes. No immune effector cell-associated neurotoxicity syndrome was observed. To the cutoff date, CAR transgenes were detectable in 77.8% of the patients. The median CAR transgene persistence was 307.5 days. Only 1 patient was positive for the anti-drug antibody. Altogether, CT103A is safe and highly active in patients with RRMM and can be developed as a promising therapy for RRMM. Patients who relapsed from prior murine BCMA CAR T-cell therapy may still benefit from CT103A. This trial was registered at http://www.chictr.org.cn as #ChiCTR1800018137.


Subject(s)
B-Cell Maturation Antigen/antagonists & inhibitors , Immunotherapy, Adoptive , Multiple Myeloma/therapy , Receptors, Chimeric Antigen/therapeutic use , Single-Chain Antibodies/therapeutic use , Adult , Afibrinogenemia/etiology , Aged , Animals , Antibodies, Anti-Idiotypic/biosynthesis , Antineoplastic Agents/therapeutic use , B-Cell Maturation Antigen/immunology , Combined Modality Therapy , Drug Resistance, Neoplasm , Female , Hematologic Diseases/etiology , Humans , Immunity, Humoral , Immunotherapy, Adoptive/adverse effects , Leukemia, Plasma Cell/etiology , Leukemia, Plasma Cell/therapy , Male , Mice , Middle Aged , Multiple Myeloma/drug therapy , Receptors, Chimeric Antigen/administration & dosage , Receptors, Chimeric Antigen/immunology , Remission Induction , Single-Chain Antibodies/immunology , Transgenes
3.
Am J Hematol ; 95(12): 1531-1541, 2020 12.
Article in English | MEDLINE | ID: mdl-32864761

ABSTRACT

Plasma cell neoplasms (PCNs), comprising plasma cell myelomas (PCMs) and plasmacytomas, which occur after solid organ transplantation, represent rare subtypes of monomorphic post-transplant lymphoproliferative disorders (M-PTLDs). Data regarding the clinical and pathological features of post-transplant (PT)-PCMs are limited. To gain a better understanding of disease biology, we performed comprehensive immunophenotypic analysis, reviewed cytogenetic analysis results and evaluated clinical outcomes of PT-PCMs diagnosed and treated at our institution. Fifteen PT-PCM (M: F - 4:1) and two PT-MGUS (two males) cases were identified. The median age of PT-PCM patients was 68 years (29-79 years) and PCMs presented at a median of 9.7 years (0.5-24.7 years) after transplantation. The PT-PCMs accounted for 11.6% of all M-PTLDs and the period prevalence was 9/3108 (0.29%), 3/1071 (0.28%), 2/1345 (0.15%) and 1/878 (0.11%) post kidney, heart, liver and lung transplantation. Lytic bone disease was observed in 1/11 (9%) patients. Marrow plasma cell infiltration ranged from 10%-70% (median 20%), with 10/15 (67%) and 5/15 (33%) cases manifesting immature and plasmablastic morphology. The immunophenotype of all cases and cytogenetic abnormalities, identified in 60% of cases, were similar to multiple myeloma (MM) of immunocompetent individuals. All PT-PCMs were EBER negative. Ten of 11 (91%) patients with active MM were treated, all with proteasome inhibitor-based therapy. Treatment response and 5-year overall survival (54.5%) was comparable to MM of immunocompetent individuals. However, the survival of patients with plasmablastic PCMs was inferior to those with immature PCMs. 0ur findings indicate PT-PCMs to be predominantly late onset PTLDs that have similar clinicopathologic characteristics as conventional MM.


Subject(s)
Leukemia, Plasma Cell , Organ Transplantation , Adult , Aged , Disease-Free Survival , Female , Follow-Up Studies , Humans , Leukemia, Plasma Cell/etiology , Leukemia, Plasma Cell/mortality , Leukemia, Plasma Cell/therapy , Male , Middle Aged , Survival Rate
4.
Leuk Lymphoma ; 60(1): 118-123, 2019 01.
Article in English | MEDLINE | ID: mdl-29965787

ABSTRACT

This multicenter retrospective study included 101 patients (median age 62 years) with secondary plasma cell leukemia (sPCL). The median time from initial multiple myeloma diagnosis to sPCL was 31 months. Fifty-five out of 72 patients (75%) who received any therapy were treated with immunomodulators (IMiDs) and/or proteasome inhibitors (PIs), and 14/72 (19%) underwent salvage autologous stem cell transplantation (ASCT). The overall response rate in patients who received ASCT or PI (either alone or in combination) was higher than in those who did not (93% vs. 36% and 60% vs. 30%, respectively). The median overall survival (OS) in patients who received therapy was 4.2 months (95% CI: 1.3; 8.0) with a 1-year OS of 19%. Platelet count ≤100 × 109/L at sPCL diagnosis was the only independent predictor of a poorer OS in treated patients (HR = 3.98, p = .0001). These findings suggest that patients with sPCL may benefit from salvage ASCT- and PI-based regimens.


Subject(s)
Leukemia, Plasma Cell/therapy , Multiple Myeloma/complications , Salvage Therapy/methods , Stem Cell Transplantation , Adult , Aged , Aged, 80 and over , Female , Follow-Up Studies , Humans , Immunologic Factors/therapeutic use , Leukemia, Plasma Cell/etiology , Leukemia, Plasma Cell/mortality , Male , Middle Aged , Multiple Myeloma/mortality , Multiple Myeloma/therapy , Proteasome Inhibitors/therapeutic use , Retrospective Studies , Survival Analysis , Transplantation, Autologous , Treatment Outcome
5.
Ann Clin Lab Sci ; 47(5): 611-619, 2017 Sep.
Article in English | MEDLINE | ID: mdl-29066491

ABSTRACT

IgM multiple myeloma (MM) is a rare entity representing approximately 0.5% of all MM. It should be distinguished from malignant neoplasms of B cells with plasmacytic differentiation such as Waldenstrom macroglobulinemia (WM) and marginal zone lymphoma with plasmacytic differentiation. Plasma cell leukemia (PCL) is a rare and aggressive variant of MM characterized by the presence of circulating plasma cells. We present a case report of a patient who presented with IgM MM in primary PCL phase with high-risk cytogenetics. To our knowledge, this is the first reported case of IgM MM with primarily leukemic presentation in the era of novel drugs. We demonstrate that it is important to distinguish IgM MM from WM and review the data from clinical trials that was used to devise a treatment strategy for this high-risk patient. This case adds to the understanding of the diagnosis and management of IgM MM in leukemic phase.


Subject(s)
Leukemia, Plasma Cell/etiology , Multiple Myeloma/diagnosis , Aged , Chromosome Deletion , Chromosomes, Human, Pair 17 , Combined Modality Therapy , Diagnosis, Differential , Female , Humans , Immunoglobulin M/analysis , Immunoglobulins/analysis , Leukemia, Plasma Cell/diagnosis , Leukemia, Plasma Cell/prevention & control , Multiple Myeloma/genetics , Multiple Myeloma/physiopathology , Multiple Myeloma/therapy , Treatment Outcome , Waldenstrom Macroglobulinemia/diagnosis
6.
Leuk Lymphoma ; 58(7): 1538-1547, 2017 07.
Article in English | MEDLINE | ID: mdl-27819179

ABSTRACT

Plasma cell leukemia (PCL) is a rare, but very aggressive, plasma cell dyscrasia, representing a distinct clinicopathological entity as compared to multiple myeloma (MM), with peculiar biological and clinical features. A hundred times rarer than MM, the disease course is characterized by short remissions and poor survival. PCL is defined by an increased percentage (>20%) and absolute number (>2 × 109/l) of plasma cells in the peripheral blood. PCL is defined as 'primary' when peripheral plasmacytosis is detected at diagnosis, 'secondary' when leukemization occurs in a patient with preexisting MM. Novel agents have revolutionized the outcomes of MM patients and have been introduced also for the treatment of PCL. Here, we provide an update on biology and treatment options for PCL.


Subject(s)
Leukemia, Plasma Cell/etiology , Leukemia, Plasma Cell/therapy , Animals , Biomarkers , Combined Modality Therapy , Disease Management , Genetic Predisposition to Disease , Genomics/methods , Humans , Leukemia, Plasma Cell/diagnosis , Leukemia, Plasma Cell/metabolism , Phenotype
7.
Expert Rev Hematol ; 9(11): 1063-1073, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27759436

ABSTRACT

INTRODUCTION: Primary plasma cell leukemia (PPCL) is a rare and aggressive variant of multiple myeloma. The introduction of novel agents and modern technologies has recently partially changed the clinical and biological scenario of this malignancy, allowing limited, but not negligible, progresses. Areas covered: We will discuss: the complex landscape of genetic alterations in PPCL, derived from conventional and high-throughput technologies; the best available treatments for PPCL; the possible future therapeutic perspectives. Expert commentary: PPCL requires an immediate and intensive multi-phase treatment with short therapy-free intervals, which should include novel agents and autologous stem cell transplantation in eligible patients. Allogeneic transplantation should be considered in selected cases. In older and/or frailer individuals, personalized approaches should be applied. Integrated treatments with next generation proteasome inhibitors/IMIDs and monoclonal antibodies are currently planned or under investigation. The identification of novel genomic biomarkers may be potentially helpful for risk stratification and future personalized therapies.


Subject(s)
Leukemia, Plasma Cell/diagnosis , Leukemia, Plasma Cell/therapy , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Disease Management , Genetic Predisposition to Disease , Genetic Testing/methods , Genomics/methods , Hematopoietic Stem Cell Transplantation , Humans , Leukemia, Plasma Cell/etiology , Leukemia, Plasma Cell/mortality , Molecular Targeted Therapy , Prognosis
8.
Curr Treat Options Oncol ; 17(4): 19, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26995215

ABSTRACT

Primary plasma cell leukemia (PPCL) is an aggressive and rare variant of multiple myeloma (MM), characterized by peculiar adverse clinical and biological features. Though the poor outcome of PPCL has been slightly improved by novel treatments during the last 10 years, due to the limited number of available studies in this uncommon disease, optimal therapy remains a classic unmet clinical need. Anyway, in the real-life practice, induction with a bortezomib-based three-drug combination, including dexamethasone and, possibly, lenalidomide, or, alternatively, thalidomide, cyclophosphamide, or doxorubicin, is a reasonable first-line option. This approach may be particularly advisable for patients with adverse cytogenetics, hyperleucocytosis, and rapidly progressive disease, in whom a fast response is required, or for those with suboptimal renal function, where, however, lenalidomide should be used with caution until renal activity is restored. In younger subjects, leukemia/lymphoma-like more intensive regimens, including hyperfractionated cyclophosphamide, vincristine, doxorubicin, and dexamethasone or continue-infusion cisplatin, doxorubicin, cyclophosphamide, and etoposide, may be also combined with bortezomib +/- thalidomide. Treatment must be started immediately after a diagnosis of PPCL is made to avoid the risk of irreversible disease complications and, in such a context, the prevention of tumor lysis syndrome is mandatory. In patients eligible for autologous stem cell transplantation (AuSCT), other alkylating agents, in particular melphalan, should be initially avoided in order to allow adequate collections of CD34+ peripheral blood stem cells (PBSC). A combination of lenalidomide and dexamethasone may be a valuable alternative option to manage older or unfit patients or those with slower disease evolution or with signs of neuropathy, contraindicating the use of bortezomib. Patients not suitable for transplant procedures should continue the treatment, if a response occurs and if tolerated, considering the possibility of a prolonged maintenance therapy. AuSCT should be pursued in all eligible patients less than 65 years old who achieve a significant response after a short course of induction treatment. PBSC collection should reach a threshold of at least 5 × 10(6) CD34+ PBSC/kg using cyclophosphamide plus G-CSF and adding the mobilizing agent plerixafor, if necessary. High-dose melphalan (HDM) (200 or 140 mg/m(2), according to age and renal function) remains the preferable conditioning regimen. A second AuSCT should be always considered, even in patients achieving complete response (CR) after the first AuSCT, as the short progression-free survival (PFS) generally seen in PPCL suggests the persistence of a relevant burden of residual disease; this provides a strong rationale for the use of post-transplantation therapies in PPCL to improve depth of response, to maintain remission, and, possibly, to increase survival, though consolidation and/or maintenance strategies with novel agents, whose efficacy has been well demonstrated in MM, have not been still extensively evaluated in PPCL. The search of a suitable donor should start as soon as possible and an allogeneic stem cell transplant (AlloSCT) with a myeloablative conditioning (MAC) regimen discussed with younger patients responsive to induction therapy and with poor prognostic parameters at diagnosis. A sequence of AuSCT followed by reduced intensity conditioning (RIC) or non-myeloablative (NMA) AlloSCT may be considered in selected cases. Salvage therapies for relapsed/refractory disease, especially using new drugs not employed at diagnosis, are sometimes effective in the short term, but a rapid relapse is still generally the rule; AlloSCT in relapsed and eligible patients with sensitive disease after salvage treatments is, therefore, recommended.


Subject(s)
Leukemia, Plasma Cell/diagnosis , Leukemia, Plasma Cell/therapy , Combined Modality Therapy , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Leukemia, Plasma Cell/etiology
10.
Am J Hematol ; 89(2): 145-50, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24123068

ABSTRACT

Plasma cell leukemia (PCL) is a rare and aggressive plasma cell disorder, with poor outcome. Bortezomib-based regimens (BBR) are highly effective in myeloma, but there is limited information about their efficacy and safety in PCL. Thus, we retrospectively collected data from 42 consecutive PCL patients (25 with primary PCL-pPCL and 17 with secondary PCL-sPCL) to explore the role of BBR in this entity. BBR were administered in 29 of 42 patients, while 6 of 25 patients with pPCL underwent autologous transplantation. Objective response (≥partial response) was significantly higher in patients treated with BBR versus conventional therapies (69% vs. 30.8%, P = 0.04); 27.5% of patients treated with BBR achieved at least very good partial response (vgPR). The highest ORR was observed in pPCL patients treated with BBR (88.9%; ≥vgPR: 33.3%). In BBR-group, grade 3 of 4 hematological, neurological and renal toxicity and neutropenic infections were observed in 41.4%, 7%, 3.4%, and 31%, respectively. With a median follow-up of 51 months, median overall survival (OS) for patients treated with BBR versus conventional therapies was 13 versus 2 months (P < 0.007). Median OS of patients with pPCL and sPCL treated with BBR was 18 and 7 months, respectively (P < 0.001). In the multivariate analysis normal PLTs, treatment with BBR and high quality response were the only powerful predictors for survival. Our study carrying the longest reported median follow-up, demonstrated that treatment of PCL with BBR induces high response rates and prolongs survival over conventional therapies, regardless of additional autologous transplantation rescue or established high risk features, with manageable toxicity.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Leukemia, Plasma Cell/drug therapy , Leukemia, Plasma Cell/mortality , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Boronic Acids/administration & dosage , Bortezomib , Female , Humans , Leukemia, Plasma Cell/diagnosis , Leukemia, Plasma Cell/etiology , Male , Middle Aged , Neoplasm Staging , Prognosis , Pyrazines/administration & dosage , Retrospective Studies , Treatment Outcome
12.
Eur J Haematol ; 87(1): 20-7, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21692850

ABSTRACT

Cancer cells are characterized by having chromosomal abnormalities. The number of aberrations and the specific chromosomes affected are likely correlated with tumor progression. In this study, we analyzed the karyotype of 126 plasma cell leukemia (PCL) patients to identify the most frequently occurring imbalances and to design a model of karyotypic evolution. The Mitelman database of chromosome was searched and abnormal karyotypes were assessed. The main clones were analyzed and chromosomal gains and losses were used to design a model of genetic acquisition based on the calculation of a variable called time to occurrence. Our comprehensive study of genetic abnormalities of a large number of PCL karyotypes suggests that PCL is mainly characterized by the presence of whole chromosome losses as well as IgH rearrangements which is similar to that observed in non-hyperdiploid multiple myeloma (MM). Temporal analysis suggests that most PCL have around 10 abnormalities at diagnosis. It is possible that accumulation of abnormalities such as 17p13 (TP53) and 1p losses may trigger the extramedullary features of PCL. Our study demonstrates that cytogenetics is a valuable tool to evaluate the role of genetic imbalances on karyotypic evolution by using a mathematical model.


Subject(s)
Chromosome Aberrations , Leukemia, Plasma Cell/genetics , Chromosome Breakage , Cluster Analysis , Cytogenetic Analysis , Databases, Genetic , Gene Rearrangement, B-Lymphocyte, Heavy Chain , Humans , Karyotyping , Leukemia, Plasma Cell/drug therapy , Leukemia, Plasma Cell/etiology , Models, Genetic , Multiple Myeloma/genetics , Prognosis , Translocation, Genetic
13.
Br J Haematol ; 150(4): 418-27, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20701603

ABSTRACT

Extramedullary plasma cell cancers, such as plasma cell leukaemia (PCL) and multiple extramedullary plasmacytomas (MEP) are very aggressive malignancies. These can be primary (de-novo) or secondary due to progressive prior multiple myeloma (MM). Recent reports suggest an increase in incidence of these disorders. Compared to MM, organ invasion is common in PCL, while soft tissue tumors involving the head, neck or paraspinal area are common sites for MEP. Markers of poor prognosis are frequently observed in these extramedullary forms of plasma cell cancers, and survival is significantly inferior compared to patients with MM. Conventional chemotherapeutic and radiotherapy approaches have been employed with variable results. Even high dose chemotherapy with autologous stem cell rescue has not been able to demonstrate consistent improvement in survival outcome. Although not specifically evaluated, novel anti-plasma cell agents, such as the proteasome inhibitor bortezomib, and immunomodulatory drugs, such as lenalidomide, appear to be active against these aggressive cancers. Clinical and translational research directed at improved understanding of disease biology and development of novel therapeutics is urgently needed.


Subject(s)
Multiple Myeloma/drug therapy , Aged , Antineoplastic Agents/therapeutic use , Boronic Acids/therapeutic use , Bortezomib , Female , Humans , Lenalidomide , Leukemia, Plasma Cell/diagnosis , Leukemia, Plasma Cell/drug therapy , Leukemia, Plasma Cell/etiology , Male , Middle Aged , Multiple Myeloma/diagnosis , Multiple Myeloma/etiology , Prognosis , Pyrazines/therapeutic use , Thalidomide/analogs & derivatives , Thalidomide/therapeutic use
14.
Ann Dermatol Venereol ; 136(5): 427-30, 2009 May.
Article in French | MEDLINE | ID: mdl-19442799

ABSTRACT

BACKGROUND: Bortezomib (Velcade) is a proteasome inhibitor used in the treatment of myeloma and other blood dyscrasias. We report the cases of two patients who developed a peculiar toxic rash suggestive of Sweet's syndrome while receiving bortezomib; one patient also presented giant mucous membrane ulcerations. PATIENTS AND METHODS: Case 1: bortezomib treatment was started in a 62-year-old man for mantle cell lymphoma. Ten days after the first treatment cycle, giant, painful oral ulcerations were noted but they resolved spontaneously. One week after the second cycle, further oral ulceration appeared, this time with a papulonodular skin rash. Histology showed neutrophilic dermal infiltrates in the skin with predominantly lymphocytic inflammation of the oral mucosa. Bortezomib was stopped and all lesions resolved with colchicine treatment. Case 2: a 46-year-old woman was receiving bortezomib treatment for plasma cell leukemia. A febrile skin rash appeared two days after the first treatment cycle but resolved spontaneously. After the first bortezomib injection during the next cycle, painful papules and nodules appeared on the trunk. The skin biopsy results were consistent with Sweet's syndrome. The lesions disappeared spontaneously. Dexamethasone was administered concomitantly with bortezomib in the ensuing cycles and there was no relapse of the skin lesions. DISCUSSION: Bortezomib-induced skin lesions are common and usually do not justify treatment withdrawal. Published observations of bortezomib-induced eruption occasionally show clinical and histological features of Sweet's syndrome, but there has been no mention of oral mucosal ulcerations. In our cases, these could be related to bortezomib-induced neutrophilic dermatosis.


Subject(s)
Antineoplastic Agents/adverse effects , Boronic Acids/adverse effects , Pyrazines/adverse effects , Sweet Syndrome/chemically induced , Biopsy , Bortezomib , Colchicine/therapeutic use , Dexamethasone/therapeutic use , Female , Humans , Leukemia, Plasma Cell/etiology , Lymphoma, Mantle-Cell/drug therapy , Male , Middle Aged , Skin Ulcer/chemically induced , Skin Ulcer/pathology , Sweet Syndrome/drug therapy , Sweet Syndrome/pathology , Treatment Outcome
15.
Orv Hetil ; 149(41): 1957-9, 2008 Oct 12.
Article in Hungarian | MEDLINE | ID: mdl-18842514

ABSTRACT

The most aggressive and rare manifestation of multiple myeloma is plasma cell leukaemia (PCL). While secondary form of PCL represents those heavily pretreated cases when leukaemic transformation develops terminally after intensive chemotherapy in patients with multiple myeloma, primary cases are characterized by leukaemic symptoms present at diagnosis. The secondary form has a rapid progression. The management of PCL is still unsolved. The authors present a case of a patient with non-secretory multiple myeloma who had developed plasma cell leukaemia after peripheral stem cell transplantation. PAD (bortezomib, doxorubicin, dexamethasone) treatment resulted in complete remission and 9-month survival of the patient. Previous case reports in the literature and our experience have revealed PAD protocol to be well tolerated and effective in PCL. Combination of PAD treatment with autologous and/or allogenic stem cell transplantation might further improve patients' outcome.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Leukemia, Plasma Cell/drug therapy , Leukemia, Plasma Cell/etiology , Multiple Myeloma/complications , Peripheral Blood Stem Cell Transplantation , Aged , Antibiotics, Antineoplastic/administration & dosage , Antineoplastic Agents, Hormonal/administration & dosage , Boronic Acids/administration & dosage , Bortezomib , Dexamethasone/administration & dosage , Doxorubicin/administration & dosage , Humans , Male , Multiple Myeloma/surgery , Pyrazines/administration & dosage , Remission Induction , Transplantation, Autologous , Treatment Outcome
16.
Lab Hematol ; 12(4): 210-6, 2006.
Article in English | MEDLINE | ID: mdl-17118772

ABSTRACT

The St. Vincent's Comprehensive Cancer Center (SVCCC) has a large multiple myeloma program in downtown New York City. The laboratory at SVCCC is an integral part of the diagnosing and monitoring of its myeloma patients. Circulating plasma cells are not a common finding in multiple myeloma. Being able to detect plasma cells in peripheral blood is important because they are a prognostic indicator that correlates with disease progression. Furthermore, the peripheral blood plasma cell population can demonstrate morphologic variability. Immature plasma cells, both plasmablasts and proplasmacytes are associated with more aggressive disease and shortened survival. We encountered 3 multiple myeloma patients with circulating immature plasma cells that appeared as distinct populations on our hematology analyzer's automated white blood cell (WBC) differential. The immature plasma cells, given their unique cellular characteristics, appeared in a common place within the WBC differential scatterplot in each patient. In our laboratory, we have utilized this common graphic pattern to screen for immature plasma cells. This pattern has proven to be a useful tool in our large population of multiple myeloma patients. We have also used examination of the scatterplots in other hematologic malignancies such as chronic lymphocytic leukemia. Using this review policy, the laboratory has been able to achieve a smear review of 25% in our highly abnormal patient population.


Subject(s)
Data Display , Flow Cytometry/instrumentation , Leukemia, Plasma Cell/diagnosis , Leukocyte Count/instrumentation , Multiple Myeloma/immunology , Plasma Cells/classification , Autoanalysis/instrumentation , Equipment and Supplies/standards , Female , Humans , Laboratories, Hospital/standards , Leukemia, Plasma Cell/blood , Leukemia, Plasma Cell/etiology , Male , Middle Aged , Multiple Myeloma/complications , Plasma Cells/pathology , Reference Standards , Sensitivity and Specificity
17.
J Pak Med Assoc ; 55(10): 452-3, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16304856

ABSTRACT

Plasma cell leukemia (PCL) is a rare disease and is the least common variant of multiple myeloma accounting for 2-3% of all plasma cell dyscrasias. We report a patient who presented with history of high grade fever, weakness, palpitations, loss of appetite, bone pains and mental confusion for twenty days. Initial evaluation revealed plasmacytosis with blood plasma cell count of 5184/cumm. His hemoglobin (Hb) was 11.3 gm/dl, platelets were 75000/cumm and total leucocyte count (TLC) was 21600/cumm (24% plasma cells). Bone marrow examination revealed >60% plasmablasts. Serum LDH was high at 3117 U/L and serum calcium was also elevated at 13.9 mg/dl. A diagnosis of PCL was made and the patient was started on treatment for hypercalcaemia with Melphalan/Prednisolone regime along with supportive care. Patient deteriorated very rapidly despite treatment and died on the eighth day. A detailed report of this case and a review of PCL is presented here.


Subject(s)
Leukemia, Plasma Cell/etiology , Multiple Myeloma/complications , Antineoplastic Agents, Alkylating/therapeutic use , Drug Therapy, Combination , Fatal Outcome , Glucocorticoids/therapeutic use , Humans , Leukemia, Plasma Cell/blood , Leukemia, Plasma Cell/drug therapy , Leukocyte Count , Male , Melphalan/therapeutic use , Middle Aged , Multiple Myeloma/blood , Multiple Myeloma/drug therapy , Plasma Cells/pathology , Prednisolone/therapeutic use
18.
Tunis Med ; 83(7): 433-6, 2005 Jul.
Article in French | MEDLINE | ID: mdl-16220704

ABSTRACT

Plasma cell leukemia is considered as the leukemic variant of multiple myeloma. It is a rare entity. There are two forms: a secondary one following a known myeloma, the diagnosis of which is easy, and a primary one arising without a preceding phase of multiple myeloma. The diagnosis of the latter form is more difficult, a differential diagnosis has often to be discussed with other lymphoproliferative diseases. Prognosis is poor. We report 2 cases of secondary plasma cell leukemia diagnosed over ten years, among 59 of multiple myeloma cases. We describe the epidemiologic, clinical, biological and evolutionary characteristics.


Subject(s)
Leukemia, Plasma Cell/etiology , Multiple Myeloma/pathology , Female , Humans , Male , Middle Aged
19.
Infection ; 32(5): 296-8, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15624895

ABSTRACT

Plasmablastic leukemia (PL) as a complication of human herpes virus 8 (HHV8)-associated Castleman's disease is marked by a rapid and fatal outcome. In patients with AIDS, survival of 7 to 14 days after diagnosis has been reported. Prompt splenectomy and chemotherapy might lead to a significant survival benefit. Here we report a case of long-term survival in a patient with AIDS and multicentric Castleman's disease (MCD) complicated by PL.


Subject(s)
Acquired Immunodeficiency Syndrome/complications , Castleman Disease/etiology , Herpesviridae Infections/etiology , Leukemia, Plasma Cell/etiology , AIDS-Related Opportunistic Infections/virology , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Castleman Disease/drug therapy , Castleman Disease/surgery , Herpesvirus 8, Human , Humans , Leukemia, Plasma Cell/drug therapy , Leukemia, Plasma Cell/surgery , Male , Splenectomy
20.
J Biol Regul Homeost Agents ; 18(2): 161-5, 2004.
Article in English | MEDLINE | ID: mdl-15471221

ABSTRACT

The term "monoclonal gammopathy" (MG) includes a group of clonal plasma cell disorders, which show heterogeneous clinical behavior. While multiple myeloma (MM) and plasma cell leukemia (PCL) are incurable malignant diseases, most patients with MG of undetermined significance (MGUS) show an indolent/benign clinical course. Evidence has accumulated which supports the role of the bone marrow microenvironment in MG. Accordingly, the survival, drug-resistance and proliferation of MM cells have been shown to be largely dependent on a supportive microenvironment. Among the different environment-associated parameters, those related to the status/activity of the immune system are particularly relevant. This review focuses on the different ways clonal plasma cells (PC) interact with the immune system in different models of MG, to characterize crucial events in the development and progression of MG. These advances may support the design of novel therapeutic approaches in patients with MG.


Subject(s)
Paraproteinemias/immunology , Plasma Cells/immunology , Bone Marrow/immunology , Bone Marrow Cells/immunology , Clone Cells/immunology , Humans , Immunophenotyping , Leukemia, Plasma Cell/etiology , Leukemia, Plasma Cell/immunology , Lymphocyte Activation/immunology , Membrane Proteins/immunology , Membrane Proteins/metabolism , Multiple Myeloma/etiology , Multiple Myeloma/immunology , Paraproteinemias/etiology , T-Lymphocytes/immunology , T-Lymphocytes/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...