Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Leuk Res ; 122: 106948, 2022 11.
Article in English | MEDLINE | ID: mdl-36108425

ABSTRACT

The Phase 3 ICARIA-MM (NCT02990338) and IKEMA (NCT03275285) studies demonstrated that isatuximab (Isa) plus pomalidomide (P) and dexamethasone (d; Isa-Pd) or carfilzomib (K) and d (Isa-Kd) improved progression-free survival (PFS) versus Pd or Kd in patients with relapsed and/or refractory multiple myeloma. In this post hoc analysis of patients with soft-tissue plasmacytomas, we evaluated Isa-Pd/Isa-Kd efficacy using central radiology and central laboratory assessments. Given the low incidence of soft-tissue plasmacytomas (7.8 %, ICARIA-MM; 6.3 %, IKEMA), efficacy data were pooled across the two studies. PFS (HR, 0.47; 95 % CI, 0.21-1.08), overall response rate (50.0 % vs 17.7 %), and very good partial response or better rate (26.9 % vs 11.8 %) were improved with Isa-Pd/Isa-Kd versus Pd/Kd, with consistent improvements within individual studies. Patients with soft-tissue plasmacytomas who received Isa-Pd/Isa-Kd had similar median PFS compared with those without soft-tissue plasmacytomas and received Pd/Kd. Safety is reported individually per study. Longer median treatment duration and more Grade ≥ 3 treatment-emergent adverse events occurred in the Isa versus control arms in ICARIA-MM (36.9 vs 8.4 weeks; 85.7 % vs 70.0 %) and IKEMA (41.9 vs 29.9 weeks; 100.0 % vs 57.1 %); however, Isa did not increase the percentage of patients with fatal events or drug discontinuation. Isa-Pd or Isa-Kd is a potential new treatment option and partially overcomes the poor prognosis associated with soft-tissue plasmacytomas in relapsed and/or refractory multiple myeloma.


Subject(s)
Multiple Myeloma , Neoplasms, Plasma Cell , Plasmacytoma , Humans , Dexamethasone/therapeutic use , Multiple Myeloma/drug therapy , Neoplasms, Plasma Cell/drug therapy , Plasmacytoma/drug therapy , Clinical Trials, Phase III as Topic
2.
Indian J Pathol Microbiol ; 63(3): 485-487, 2020.
Article in English | MEDLINE | ID: mdl-32769348

ABSTRACT

Myeloma plasma cells vary from mature forms to immature, plasmablastic, and pleomorphic cells. Only a few cases of morphologic variant of plasma cell neoplasm have been reported, in which the plasma cell neoplasm presented with lymphoplasmacytic, megakaryocytic, plasmablastic, lymphocytosis-like, and variant hairy cell leukemia-like morphological features. A 66-year-old man sought medical attention with a previous 2-month history of lower back and chest pain. Magnetic resonance imaging (MRI) of the thoracic spine showed thoracic vertebral body shape and disc degeneration, and bone lesion. Blood work showed mild anemia (hemoglobin, 101 g/L; white blood cells, 6.98 × 109/L; platelets, 146 × 109/L.), hyperuricemia (UA 671 umol/L), and immunoglobulin G kappa [IgG(κ)] paraproteins. Bone marrow study revealed diffuse invasion by sheets of megakaryoblast-like cells. Flow cytometric analysis and bone marrow biopsy revealed plasma cell myeloma (PCM), and thoracic puncture biopsy indicated plasma cell neoplasms. Overall, the findings were in accordance with a PCM. To date, this is the first reported case of PCM with megakaryoblastic morphology mimicking acute leukemia. Recognizing the morphological variant of PCM is important in differentiating it from acute leukemia.


Subject(s)
Leukemia, Myeloid, Acute/diagnosis , Megakaryocyte Progenitor Cells , Neoplasms, Plasma Cell/diagnostic imaging , Plasma Cells/pathology , Spine/diagnostic imaging , Acute Disease , Aged , Antineoplastic Agents/therapeutic use , Biopsy , Bone Marrow/pathology , Diagnosis, Differential , Fatal Outcome , Humans , Leukemia, Myeloid, Acute/pathology , Magnetic Resonance Imaging , Male , Neoplasms, Plasma Cell/drug therapy , Spine/pathology
4.
BMC Ophthalmol ; 19(1): 110, 2019 May 14.
Article in English | MEDLINE | ID: mdl-31088416

ABSTRACT

BACKGROUND: The purpose of this case series was to further characterize proteasome inhibitor associated chalazia and blepharitis, to investigate outcomes of different management strategies, and to propose a treatment algorithm for eyelid complications in this patient population. METHODS: This retrospective case series included sixteen patients found to have chalazia and/or blepharitis while receiving proteasome inhibitors for plasma cell disorders at Mount Sinai Hospital in New York, NY from January 2010 through January 2017. Main outcomes were complete resolution of eyelid complications and time to resolution. Student's t-test was used to compare average values and Fisher's exact test was used to compare proportions. RESULTS: Fourteen patients had chalazia and 10 had blepharitis. Chalazia averaged 5.4 mm, and 11 patients with chalazia experienced two or more lesions. Median follow-up time was 17 months. Average time from bortezomib exposure to onset of first eyelid complication was 3.4 months. Chalazia episodes were more likely to completely resolve than blepharitis episodes (p = 0.03). Ocular therapy alone was trialed for an average of 1.8 months before proceeding to bortezomib omission. Average time to eyelid complication resolution using ocular therapy alone was 1.8 months versus 3.1 months after bortezomib omission. In this series, the combination of ocular therapy and bortezomib omission led to complete resolution of eyelid complications more often than ocular therapy alone. CONCLUSION: Proteasome inhibitor associated eyelid complications were identified in sixteen patients with plasma cell disorders. Eyelid complications may be treated with a 2-month trial of conservative ocular therapies alone, followed by continuation of ocular therapy in combination with bortezomib omission if eyelid signs persist.


Subject(s)
Blepharitis/chemically induced , Bortezomib/adverse effects , Chalazion/chemically induced , Proteasome Inhibitors/adverse effects , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Neoplasms, Plasma Cell/drug therapy , Retrospective Studies
5.
Blood Adv ; 2(15): 2012-2019, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30108109

ABSTRACT

Allogeneic hematopoietic stem cell transplantation (allo-SCT) is potentially curative for a number of hematologic conditions, both malignant and nonmalignant. However, its success can be limited by the development of acute and chronic graft-versus-host disease (GVHD). Chronic GVHD (cGVHD) is the most common long-term complication following allo-SCT, and patients who develop this condition have significantly higher morbidity and mortality and significantly lower quality of life than patients who do not. Until recently, there were no US Food and Drug Administration (FDA)-approved therapies for cGVHD treatment. In this review article, we describe how ibrutinib was identified as potential cGVHD therapy based on preclinical cGVHD models and clinical studies in B-cell malignancies and elucidation of its mechanisms of action in cGVHD. Results from a phase 2 clinical trial that was designed based on National Institutes of Health Criteria for the grading and staging of cGVHD culminated in the FDA-approval of ibrutinib as second line therapy of steroid-refractory or steroid-resistant cGVHD. Results of ibrutinib studies in phase 3 randomized studies, for cGVHD prophylaxis and as first -line testing along with steroids will be especially important in selecting the preferred indications for ibrutinib in patients at risk for or who have developed cGVHD.


Subject(s)
B-Lymphocytes , Drug Resistance/drug effects , Graft vs Host Disease/drug therapy , Neoplasms, Plasma Cell/drug therapy , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Adenine/analogs & derivatives , Animals , Chronic Disease , Drug Approval , Graft vs Host Disease/pathology , Humans , Neoplasms, Plasma Cell/pathology , Piperidines , Steroids/therapeutic use , United States , United States Food and Drug Administration
6.
Leuk Res ; 66: 73-78, 2018 03.
Article in English | MEDLINE | ID: mdl-29407586

ABSTRACT

8q24/MYC rearrangements resulting in MYC overexpression occur most frequently in lymphoid neoplasms. MYC rearrangements rarely have been described in blastic plasmacytoid dendritic cell neoplasm (BPDCN). Over an 8-year period in our hospital, 5 of 41 (12%) patients with BPDCN were shown 8q24/MYC rearrangements, including 2 with t(6;8)(p21;q24), 1 with t(8;14)(q24;q32), 1 with t(X;8)(q24;q24), and 1 with t(3;8)(p25;q24). 8q24/MYC rearrangement was present in the stemline in 4 patients and in the sideline in one; the latter was a patient with primary myelofibrosis who then developed BPDCN. MYC overexpression by immunohistochemistry was variable, but largely correlated with the percentage of blasts. Four patients were treated with acute lymphoblastic leukemia-type chemotherapy regimens and 3 had a good response; 1 patient was treated with acute myeloid leukemia-type regimens and was refractory to therapy. By the end of the follow-up, 3 patients died and 2 were alive in complete remission. We conclude that 8q24/MYC rearrangements occur in 10-15% of BPDCN, often partnered with non-immunoglobulin chromosomal loci, and may play a role in BPDCN pathogenesis. In this small patient sample, patients with BPDCN and MYC rearrangement often responded to therapy with acute lymphoblastic leukemia-type chemotherapy regimens.


Subject(s)
Chromosome Aberrations , Chromosomes, Human, Pair 8/genetics , Neoplasms, Plasma Cell/drug therapy , Neoplasms, Plasma Cell/genetics , Proto-Oncogene Proteins c-myc/genetics , Aged , Child, Preschool , Female , Humans , Male , Middle Aged , Neoplasms, Plasma Cell/metabolism , Proto-Oncogene Proteins c-myc/metabolism
9.
Article in Russian | MEDLINE | ID: mdl-25591508

ABSTRACT

OBJECTIVE: Peripheral neuropathy often occurs as a side effect of medication or as a manifestation of system disease. The purpose of this study was to evaluate the features of polyneuropathy with verified paraproteinemic hemoblastosis. MATERIAL AND METHODS: We studied 104 patients, 72 women 32 men, aged 24-79 years. Along with neurological examination, authors studied vibration sensitivity and used electromyography and several scales for assessment of pain syndrome and other clinical presentations. RESULTS AND СONCLUSION: Clinical variants of peripheral neuropathy - neuropathy associated with paraproteinemia and neuropathy during treatment of advanced multiple myeloma with bortezomib (bortezomib-induced neuropathy), were singled out. An algorithm for the examination and treatment of patients with peripheral neuropathy was proposed.


Subject(s)
Peripheral Nervous System Diseases/diagnosis , Polyneuropathies/diagnosis , Adult , Aged , Antineoplastic Agents/therapeutic use , Boronic Acids/therapeutic use , Bortezomib , Female , Humans , Male , Middle Aged , Neoplasms, Plasma Cell/complications , Neoplasms, Plasma Cell/drug therapy , Peripheral Nervous System Diseases/complications , Polyneuropathies/complications , Pyrazines/therapeutic use
12.
Blood ; 119(4): 1018-28, 2012 Jan 26.
Article in English | MEDLINE | ID: mdl-22147894

ABSTRACT

Tumor progression usually proceeds through several sequential stages, any of which could be targets for interrupting the progression process if one understood these steps at the molecular level. We extracted nascent plasma cell tumor (PCT) cells from within inflammatory oil granulomas (OG) isolated from IP pristane-injected BALB/c.iMyc(Eµ) mice at 5 different time points during tumor progression. We used laser capture microdissection to collect incipient PCT cells and analyzed their global gene expression on Affymetrix Mouse Genome 430A microarrays. Two independent studies were performed with different sets of mice. Analysis of the expression data used ANOVA and Bayesian estimation of temporal regulation. Genetic pathway analysis was performed using MetaCore (GeneGo) and IPA (Ingenuity). The gene expression profiles of PCT samples and those of undissected OG samples from adjacent sections showed that different genes and pathways were mobilized in the tumor cells during tumor progression, compared with their stroma. Our analysis implicated several genetic pathways in PCT progression, including biphasic (up- and then down-regulation) of the Spp1/osteopontin-dependent network and up-regulation of mRNA translation/protein synthesis. The latter led to a biologic validation study that showed that the AMPK-activating diabetes drug, metformin, was a potent specific PCT inhibitor in vitro.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Neoplasms, Plasma Cell/drug therapy , Stromal Cells/drug effects , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Profiling , Granuloma, Plasma Cell/drug therapy , Granuloma, Plasma Cell/metabolism , Granuloma, Plasma Cell/pathology , Metformin/pharmacology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Molecular Targeted Therapy , Neoplasms, Plasma Cell/metabolism , Neoplasms, Plasma Cell/pathology , Oligonucleotide Array Sequence Analysis , Osteopontin/genetics , Osteopontin/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/metabolism , Stromal Cells/cytology , Stromal Cells/metabolism , Tumor Cells, Cultured
13.
Clin Cancer Res ; 17(23): 7313-23, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21903769

ABSTRACT

PURPOSE: The clinical success of the first-in-class proteasome inhibitor bortezomib (VELCADE) has validated the proteasome as a therapeutic target for treating human cancers. MLN9708 is an investigational proteasome inhibitor that, compared with bortezomib, has improved pharmacokinetics, pharmacodynamics, and antitumor activity in preclinical studies. Here, we focused on evaluating the in vivo activity of MLN2238 (the biologically active form of MLN9708) in a variety of mouse models of hematologic malignancies, including tumor xenograft models derived from a human lymphoma cell line and primary human lymphoma tissue, and genetically engineered mouse (GEM) models of plasma cell malignancies (PCM). EXPERIMENTAL DESIGN: Both cell line-derived OCI-Ly10 and primary human lymphoma-derived PHTX22L xenograft models of diffuse large B-cell lymphoma were used to evaluate the pharmacodynamics and antitumor effects of MLN2238 and bortezomib. The iMyc(Cα)/Bcl-X(L) GEM model was used to assess their effects on de novo PCM and overall survival. The newly developed DP54-Luc-disseminated model of iMyc(Cα)/Bcl-X(L) was used to determine antitumor activity and effects on osteolytic bone disease. RESULTS: MLN2238 has an improved pharmacodynamic profile and antitumor activity compared with bortezomib in both OCI-Ly10 and PHTX22L models. Although both MLN2238 and bortezomib prolonged overall survival, reduced splenomegaly, and attenuated IgG2a levels in the iMyc(Cα)/Bcl-X(L) GEM model, only MLN2238 alleviated osteolytic bone disease in the DP54-Luc model. CONCLUSIONS: Our results clearly showed the antitumor activity of MLN2238 in a variety of mouse models of B-cell lymphoma and PCM, supporting its clinical development. MLN9708 is being evaluated in multiple phase I and I/II trials.


Subject(s)
Antineoplastic Agents/pharmacology , Boron Compounds/pharmacology , Glycine/analogs & derivatives , Lymphoma, B-Cell/drug therapy , Neoplasms, Plasma Cell/drug therapy , Animals , Antineoplastic Agents/pharmacokinetics , Boron Compounds/administration & dosage , Boron Compounds/pharmacokinetics , Boronic Acids/pharmacokinetics , Boronic Acids/pharmacology , Bortezomib , Cell Line, Tumor , Glycine/administration & dosage , Glycine/pharmacokinetics , Glycine/pharmacology , Humans , Lymphoma, B-Cell/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Neoplasms, Plasma Cell/metabolism , Osteolysis/drug therapy , Osteolysis/etiology , Protease Inhibitors/pharmacology , Proteasome Inhibitors , Pyrazines/pharmacokinetics , Pyrazines/pharmacology , Xenograft Model Antitumor Assays
14.
Cytometry B Clin Cytom ; 78(5): 338-47, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20533391

ABSTRACT

BACKGROUND AND AIM: The clinical impact of multiparametric flow cytometry (MFC) in multiple myeloma (MM) is still unclear and under evaluation. Further progress relies on multiparametric profiling of the neoplastic plasma cell (PC) compartment to provide an accurate image of the stage of differentiation. The primary aim of this study was to perform global analysis of CD expression on the PC compartment and subsequently to evaluate the prognostic impact. Secondary aims were to study the diagnostic and predictive impact. DESIGN AND METHODS: The design included a retrospective analysis of MFC data generated from diagnostic bone marrow (BM) samples of 109 Nordic patients included in clinical trials within NMSG. Whole marrow were analyzed by MFC for identification of end-stage CD45(-) /CD38(++) neoplastic PC and registered the relative numbers of events and mean fluorescence intensity (MFI) staining for CD19, CD20, CD27, CD28, CD38, CD44, CD45, CD56, and isotypes for cluster analysis. RESULTS: The median MFC-PC number was 15%, and the median light microscopy (LM)-PC number was 35%. However, the numbers were significant correlated and the prognostic value with an increased relative risk (95% CI) of 3.1 (1.7-5.5) and 2.9 (1.4-6.2), P < 0.0003 and P < 0.004 of MFC-PC and LM-PC counts, respectively. Unsupervised clustering based on global MFI assessment on PC revealed two clusters based on CD expression profiling. Cluster I with high intensity for CD56, CD38, CD45, right-angle light-scatter signal (SSC), forward-angle light-scatter signal (FSC), and low for CD28, CD19, and a Cluster II, with low intensity of CD56, CD38, CD45, SSC, FSC, and high for CD28, CD19 with a median survival of 39 months and 19 months, respectively (P = 0.02). CONCLUSIONS: The MFC analysis of MM BM samples produces diagnostic, prognostic, and predictive information useful in clinical practice, which will be prospectively validated within the European Myeloma Network (EMN). © 2010 International Clinical Cytometry Society.


Subject(s)
Flow Cytometry/methods , Multiple Myeloma/diagnosis , Neoplasms, Plasma Cell/diagnosis , ADP-ribosyl Cyclase 1/analysis , ADP-ribosyl Cyclase 1/immunology , Adult , Aged , Aged, 80 and over , Clinical Trials as Topic , Humans , Leukocyte Common Antigens/analysis , Leukocyte Common Antigens/immunology , Melphalan/therapeutic use , Membrane Glycoproteins/analysis , Membrane Glycoproteins/immunology , Middle Aged , Multiple Myeloma/blood , Multiple Myeloma/drug therapy , Multiple Myeloma/immunology , Neoplasms, Plasma Cell/blood , Neoplasms, Plasma Cell/drug therapy , Neoplasms, Plasma Cell/immunology , Prognosis , Retrospective Studies , Stem Cell Transplantation , Young Adult
15.
Amyloid ; 16(3): 178-80, 2009.
Article in English | MEDLINE | ID: mdl-19590993

ABSTRACT

We report a patient with AH amyloidosis associated with lymphoplasmacytic leukemia that has remained in a stable state with a nephrotic syndrome for 17 months since the commencement of cyclic rituximab therapy aimed at suppression of pathogenetic gamma heavy chains. Free light chains in serum and CD20-positive cells in peripheral blood were useful as hematological markers in the patient. Rituximab might be a potent therapeutic option for AH amyloidosis associated with a B-cell lymphoproliferative disorder.


Subject(s)
Amyloidosis/complications , Amyloidosis/drug therapy , Antibodies, Monoclonal/therapeutic use , Antirheumatic Agents/therapeutic use , Nephrotic Syndrome , Amyloidosis/etiology , Antibodies/genetics , Antibodies/immunology , Antibodies, Monoclonal, Murine-Derived , Female , Humans , Middle Aged , Neoplasms, Plasma Cell/complications , Neoplasms, Plasma Cell/drug therapy , Nephrotic Syndrome/drug therapy , Nephrotic Syndrome/etiology , Rituximab
16.
Curr Treat Options Oncol ; 10(1-2): 107-25, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19548089

ABSTRACT

OPINION STATEMENT: Primary spinal neoplasms are rare tumors that can lead to significant morbidity secondary to local bone destruction and invasion into adjacent neurological and vascular structures. These tumors represent a clinical challenge to even the most experienced physicians and require a multidisciplinary approach to ensure optimal patient outcomes. This review will discuss the most common primary bone tumors and focus on recent surgical, medical, and radiation treatment advances.


Subject(s)
Spinal Neoplasms/therapy , Adolescent , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Cysts, Aneurysmal/therapy , Child , Chordoma/surgery , Combined Modality Therapy , Disease Management , Embolization, Therapeutic , Giant Cell Tumors/radiotherapy , Giant Cell Tumors/surgery , Giant Cell Tumors/therapy , Hemangioma/therapy , Hematopoietic Stem Cell Transplantation , Humans , Interdisciplinary Communication , Middle Aged , Neoplasms, Bone Tissue/drug therapy , Neoplasms, Bone Tissue/radiotherapy , Neoplasms, Bone Tissue/surgery , Neoplasms, Plasma Cell/drug therapy , Neoplasms, Plasma Cell/surgery , Spinal Neoplasms/diagnosis , Spinal Neoplasms/drug therapy , Spinal Neoplasms/radiotherapy , Spinal Neoplasms/surgery , Young Adult
18.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 16(5): 1069-72, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18928597

ABSTRACT

This study was aimed to further explore whether brain derived neurotrophic factor (BDNF) pathway is a potential therapeutic target in multiple myeloma (MM) and whether anti-BDNF monoclonal antibody can prevent the development of this disease. The in vivo antitumor effect of anti-BDNF monoclonal antibody (McAb) on a human myeloma xenograft animal model was evaluated. The model of xenograft tumors was established in the nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice by subcutaneous injection of human myeloma cell line RPMI8226. The antibodies were injected intraperitoneally at a dose of 20 microg/mouse at day 1, 2, 3 after inoculation or at a dose of 100 microg/mouse once a week after tumors were detected. The microvascular densities in tumors were analyzed by immunohistochemistry study. The effect of anti-BDNF McAb on the proliferation of RPMI8226 cells in vitro and on endothelial cells network formation in the co-culture system were determined by using a (3)H-thymidine incorporation assay and a Matrigel network formation assay, respectively. The results showed that multiple injections of anti-BDNF McAb reduced the tumor size, decreased the microvascular density and significantly prolonged tumor-free time and survival time. Moreover, the proliferation of RPMI8226 cells was inhibited in vitro by anti-BDNF McAb, but not by the control IgG. Anti-BDNF McAb also inhibited RPMI8226-induced network formation in endothelial cells in vitro. It is concluded that anti-BDNF monoclonal antibody can inhibit cell growth and angiogenesis in subcutaneous plasmacytoma.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Brain-Derived Neurotrophic Factor/immunology , Multiple Myeloma/drug therapy , Neoplasms, Plasma Cell/drug therapy , Animals , Cell Line, Tumor , Humans , Male , Mice , Mice, SCID , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL