Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 101
Filter
1.
Virchows Arch ; 481(4): 621-646, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35819517

ABSTRACT

The first section of the bone marrow workshop of the European Association of Haematopathology (EAHP) 2020 Virtual Meeting was dedicated to pediatric myeloid neoplasms. The section covered the whole spectrum of myeloid neoplasms, including myelodysplastic syndromes (MDS), myeloproliferative neoplasms (MPN), myelodysplastic/myeloproliferative neoplasms (MDS/MPN), and acute myeloid leukemia (AML). The workshop cases are hereby presented, preceded by an introduction on these overall rare diseases in this age group. Very rare entities such as primary myelofibrosis, pediatric MDS with fibrosis, and MDS/MPN with JMML-like features and t(4;17)(q12;q21); FIP1L1::RARA fusion, are described in more detail.


Subject(s)
Myelodysplastic Syndromes , Myelodysplastic-Myeloproliferative Diseases , Myeloproliferative Disorders , Neoplasms , Bone Marrow/pathology , Child , Humans , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Myeloproliferative Disorders/diagnosis , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Neoplasms/pathology
2.
Best Pract Res Clin Haematol ; 33(2): 101139, 2020 06.
Article in English | MEDLINE | ID: mdl-32460987

ABSTRACT

The term myelodysplastic/myeloproliferative neoplasm (MDS/MPN) refers to a group of clonal hematopoietic neoplasms with overlapping clinical, morphologic and genetic myelodysplastic and myeloproliferative features observed at the time of first presentation. Impaired hematopoiesis morphologically associated with evidence of myelodysplasia manifests clinically with cytopenia/s. Simultaneously, myeloproliferation is seen within the bone marrow and leads to cytosis in the peripheral blood. The diagnostic category of MDS/MPN encompasses a heterogeneous group of diseases which share similarities among them, but at the same time have distinct clinical and pathologic features and eventually diverse prognosis; such differences justify their separation in a classification scheme. In the era of genetic and genomic tests, their distinction from conventional myelodysplastic syndromes or myeloproliferative neoplasms still relies on close clinocopathological correlation, with evaluation of both peripheral blood and bone marrow samples being essential in this sense. A multiparametric integration of clinicopathologic data and cytogenetics and molecular genetics results is the preferred diagnostic approach.


Subject(s)
Immunophenotyping , Myelodysplastic Syndromes , Myeloproliferative Disorders , Humans , Myelodysplastic Syndromes/classification , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/metabolism , Myeloproliferative Disorders/classification , Myeloproliferative Disorders/diagnosis , Myeloproliferative Disorders/metabolism
3.
Leukemia ; 31(3): 625-636, 2017 03.
Article in English | MEDLINE | ID: mdl-27568520

ABSTRACT

PRDM1/BLIMP-1, a master regulator of plasma-cell differentiation, is frequently inactivated in activated B-cell-like (ABC) diffuse large B-cell lymphoma (DLBCL) patients. Little is known about its genetic aberrations and relevant clinical implications. A large series of patients with de novo DLBCL was effectively evaluated for PRDM1/BLIMP-1 deletion, mutation, and protein expression. BLIMP-1 expression was frequently associated with the ABC phenotype and plasmablastic morphologic subtype of DLBCL, yet 63% of the ABC-DLBCL patients were negative for BLIMP-1 protein expression. In these patients, loss of BLIMP-1 was associated with Myc overexpression and decreased expression of p53 pathway molecules. In addition, homozygous PRDM1 deletions and PRDM1 mutations within exons 1 and 2, which encode for domains crucial for transcriptional repression, were found to show a poor prognostic impact in patients with ABC-DLBCL but not in those with germinal center B-cell-like DLBCL (GCB-DLBCL). Gene expression profiling revealed that loss of PRDM1/BLIMP-1 expression correlated with a decreased plasma-cell differentiation signature and upregulation of genes involved in B-cell receptor signaling and tumor-cell proliferation. In conclusion, these results provide novel clinical and biological insight into the tumor-suppressive role of PRDM1/BLIMP-1 in ABC-DLBCL patients and suggest that loss of PRDM1/BLIMP-1 function contributes to the overall poor prognosis of ABC-DLBCL patients.


Subject(s)
Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/mortality , Mutation , Repressor Proteins/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor , Biopsy , Female , Follow-Up Studies , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Lymphoma, Large B-Cell, Diffuse/diagnosis , Lymphoma, Large B-Cell, Diffuse/drug therapy , Male , Middle Aged , Neoplasm Staging , Positive Regulatory Domain I-Binding Factor 1 , Prognosis , Repressor Proteins/metabolism , Sequence Deletion , Transcriptome , Treatment Outcome , Young Adult
4.
Int J Lab Hematol ; 38 Suppl 1: 12-9, 2016 May.
Article in English | MEDLINE | ID: mdl-27161873

ABSTRACT

Since the publication of the latest World Health Organization (WHO) classification in 2008, there has been a significant effort for clarification of unresolved questions, especially with the help of the rapidly developing field of molecular genetic studies, next-generation sequencing in particular. Numerous entities within the WHO categories of myeloproliferative neoplasms (MPNs) and myelodysplastic (MDS)/MPNs have been extensively studied, with large published series attempting to characterize and better define their morphologic and molecular genetic features. This emerging genetic landscape maintains a robust correlation with the various disease entities recognized by the WHO classification scheme based on a careful integration of detailed clinical information, bone marrow and peripheral blood morphology, immunohistology, and genomics. This brief review summarizes the current guidelines as they apply to diagnosing both the classical BCR-ABL1 negative MPN (polycythemia vera, essential thrombocythemia, and primary myelofibrosis) and the more common subtypes of MDS/MPN overlap syndromes. The more important recent molecular updates as well as the upcoming changes to the current WHO classification, expected to be published in late 2016, will also be briefly reviewed.


Subject(s)
Myelodysplastic-Myeloproliferative Diseases/diagnosis , Myeloproliferative Disorders/diagnosis , Guidelines as Topic , Humans , Myelodysplastic-Myeloproliferative Diseases/classification , Myelodysplastic-Myeloproliferative Diseases/genetics , Myeloproliferative Disorders/classification , Myeloproliferative Disorders/genetics , Neoplasms/classification , Neoplasms/genetics , World Health Organization
5.
Leukemia ; 29(1): 66-75, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24935723

ABSTRACT

The World Health Organization classification of myelodysplastic syndromes (MDS) is based on morphological evaluation of marrow dysplasia. We performed a systematic review of cytological and histological data from 1150 patients with peripheral blood cytopenia. We analyzed the frequency and discriminant power of single morphological abnormalities. A score to define minimal morphological criteria associated to the presence of marrow dysplasia was developed. This score showed high sensitivity/specificity (>90%), acceptable reproducibility and was independently validated. The severity of granulocytic and megakaryocytic dysplasia significantly affected survival. A close association was found between ring sideroblasts and SF3B1 mutations, and between severe granulocytic dysplasia and mutation of ASXL1, RUNX1, TP53 and SRSF2 genes. In myeloid neoplasms with fibrosis, multilineage dysplasia, hypolobulated/multinucleated megakaryocytes and increased CD34+ progenitors in the absence of JAK2, MPL and CALR gene mutations were significantly associated with a myelodysplastic phenotype. In myeloid disorders with marrow hypoplasia, granulocytic and/or megakaryocytic dysplasia, increased CD34+ progenitors and chromosomal abnormalities are consistent with a diagnosis of MDS. The proposed morphological score may be useful to evaluate the presence of dysplasia in cases without a clearly objective myelodysplastic phenotype. The integration of cytological and histological parameters improves the identification of MDS cases among myeloid disorders with fibrosis and hypocellularity.


Subject(s)
Bone Marrow/pathology , Myelodysplastic Syndromes/classification , Adult , Aged , Female , Humans , Male , Middle Aged , Myelodysplastic Syndromes/pathology , Severity of Illness Index , World Health Organization
6.
Blood Cancer J ; 4: e261, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25382611

ABSTRACT

PRDM1/Blimp-1 is a tumor suppressor gene in the activated B-cell subtype of diffuse large B-cell lymphomas. Its inactivation contributes to pathogenesis in this setting by impairing terminal B-cell differentiation induced by constitutive nuclear factor-κB activation. The role of PRDM1 in Burkitt lymphoma (BL) lymphomagenesis is not known. Here we identified hypermethylation of the promoter region and exon 1 of PRDM1 in all six Epstein-Barr virus (EBV)-positive BL cell lines and 12 of 23 (52%) primary EBV-positive BL or BL-related cases examined, but in none of the EBV-negative BL cell lines or primary tumors that we assessed, implying a tumor suppressor role for PRDM1 specifically in EBV-associated BL. A direct induction of PRDM1 hypermethylation by EBV is unlikely, as PRDM1 hypermethylation was not observed in EBV-immortalized B lymphoblastoid cell lines. Treatment of EBV-positive BL cells with 5' azacytidine resulted in PRDM1 induction associated with PRDM1 demethylation, consistent with transcriptional silencing of PRDM1 as a result of DNA methylation. Overexpression of PRDM1 in EBV-positive BL cell lines resulted in cell cycle arrest. Our results expand the spectrum of lymphoid malignancies in which PRDM1 may have a tumor suppressor role and identify an epigenetic event that likely contributes to the pathogenesis of BL.


Subject(s)
Burkitt Lymphoma/metabolism , DNA Methylation , Herpesvirus 4, Human , Repressor Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Burkitt Lymphoma/genetics , Burkitt Lymphoma/pathology , Burkitt Lymphoma/virology , Cell Line, Tumor , Female , Humans , Male , Positive Regulatory Domain I-Binding Factor 1 , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics
8.
J Clin Pathol ; 66(11): 956-61, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23775435

ABSTRACT

BACKGROUND: High levels of cyclin E (CCNE) are accompanied by shorter survival in cyclophosphamide, hydroxydaunorubicin, oncovin and prednisone (CHOP)-treated diffuse large B-cell lymphomas (DLBCL), independent of the international prognostic index (IPI). Data on the prognostic role of CCNE in the 'rituximab (R)-era' are lacking. METHODS: To test reproducibility and applicability of observations from the 'pre-R era' to the 'R era', we examined the prognostic role of CCNE expression by immunohistochemistry in 1579 DLBCL on tissue microarrays (TMA); 339 patients were treated by CHOP and 635 by R-CHOP. RESULTS: 1209 samples (77%) were evaluable; failures were due to missing TMA punches and fixation artefacts. Mean expression of CCNE was 13% (0-85%); applying a cut-off of >16%, 382 DLBCL (31%) were positive. CCNE did not correlate with any of the known variables (IPI, primary site, cell of origin, proliferation, and BCL2- or C-MYC rearrangements). We were able to reproduce data suggesting an IPI- and response to therapy independent, negative prognostic impact of CCNE in CHOP-treated DLBCL patients: CCNE-positive cases had a median survival of 16 months compared with 57 months in negative ones (p=0.012). In R-CHOP-treated patients the prognostic impact of CCNE was abrogated and only IPI, cell of origin and response to therapy had a prognostic significance. CONCLUSIONS: Addition of R to CHOP overcomes the negative prognostic impact of CCNE in DLBCL. Thus, R not only prolongs survival in DLBCL but also serves a cautionary note that prognostic factors should not be transferred into the 'R era' without proper scientific studies.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/administration & dosage , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Cyclin E/metabolism , Lymphoma, Large B-Cell, Diffuse/drug therapy , Oncogene Proteins/metabolism , Aged , Cohort Studies , Cyclophosphamide/administration & dosage , Doxorubicin/administration & dosage , Female , Follow-Up Studies , Humans , Immunohistochemistry , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Male , Middle Aged , Prednisone/administration & dosage , Prognosis , Reproducibility of Results , Rituximab , Tissue Array Analysis , Treatment Outcome , Vincristine/administration & dosage
9.
Leukemia ; 26(9): 2103-13, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22437443

ABSTRACT

Gene expression profiling (GEP) has stratified diffuse large B-cell lymphoma (DLBCL) into molecular subgroups that correspond to different stages of lymphocyte development-namely germinal center B-cell like and activated B-cell like. This classification has prognostic significance, but GEP is expensive and not readily applicable into daily practice, which has lead to immunohistochemical algorithms proposed as a surrogate for GEP analysis. We assembled tissue microarrays from 475 de novo DLBCL patients who were treated with rituximab-CHOP chemotherapy. All cases were successfully profiled by GEP on formalin-fixed, paraffin-embedded tissue samples. Sections were stained with antibodies reactive with CD10, GCET1, FOXP1, MUM1 and BCL6 and cases were classified following a rationale of sequential steps of differentiation of B cells. Cutoffs for each marker were obtained using receiver-operating characteristic curves, obviating the need for any arbitrary method. An algorithm based on the expression of CD10, FOXP1 and BCL6 was developed that had a simpler structure than other recently proposed algorithms and 92.6% concordance with GEP. In multivariate analysis, both the International Prognostic Index and our proposed algorithm were significant independent predictors of progression-free and overall survival. In conclusion, this algorithm effectively predicts prognosis of DLBCL patients matching GEP subgroups in the era of rituximab therapy.


Subject(s)
Algorithms , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Gene Expression Profiling , Lymphoma, Large B-Cell, Diffuse/classification , Lymphoma, Large B-Cell, Diffuse/drug therapy , Antibodies, Monoclonal, Murine-Derived/administration & dosage , Biomarkers, Tumor/metabolism , Cyclophosphamide/administration & dosage , Doxorubicin/administration & dosage , Female , Humans , Immunoenzyme Techniques , Immunophenotyping , Lymphoma, Large B-Cell, Diffuse/mortality , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Prednisone/administration & dosage , Prognosis , Rituximab , Survival Rate , Tissue Array Analysis , Vincristine/administration & dosage
10.
Mod Pathol ; 24(1): 108-16, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20852592

ABSTRACT

Splenic hamartoma is a rare tumor-like lesion composed of structurally disorganized red pulp elements. It has been hypothesized that two other splenic lesions, cord capillary hemangioma and myoid angioendothelioma, may fall within the spectrum of splenic hamartoma, simply representing morphological variants. In this study, we compared the vascular and stromal composition of cord capillary hemangioma and myoid angioendothelioma with those of classical hamartoma. In addition, we assessed the clonal vs polyclonal nature of the lesions in nine female cases by performing clonality analysis for X-chromosome inactivation at the human androgen receptor locus (HUMARA) on laser-assisted microdissected samples. In 15 of 17 cases, increased reticulin and/or collagen content was observed. The classical hamartoma cases showed a vasculature predominantly composed of CD8+ CD31+ CD34- splenic sinuses, whereas cases of cord capillary hemangioma and myoid angioendothelioma contained many CD8- CD31+ CD34+ cord capillaries, but very little CD8+ vasculature. All cases lacked expression of D2-40 and Epstein Barr virus-encoded RNA. All cases showed a proliferation index of ≤5% by Ki-67. Cases of classical hamartoma lacked significant perisinusoidal expression of collagen IV and low-affinity nerve growth factor receptor. Both markers were variably expressed in the other lesions. Increased CD163-positive histiocytes were found in four cases (three cord capillary hemangiomas and one myoid angioendothelioma). HUMARA analysis was informative in all nine tested cases, of which three cases showed a non-random X-chromosome inactivation pattern, indicating clonality. All three clonal cases were cord capillary hemangiomas. Our study has shown that in spite of considerable morphologic heterogeneity and overlapping features, classical hamartoma and cord capillary hemangioma and myoid angioendothelioma are different in terms of their vascular and stromal composition. Clonality analysis supports a true neoplastic origin for the cord capillary hemangioma. A larger study using additional immunohistochemical and molecular studies is necessary to further evaluate the biological significance of the current findings.


Subject(s)
Chromosomes, Human, X , Hamartoma/genetics , Hemangioma, Capillary/genetics , Splenic Neoplasms/genetics , X Chromosome Inactivation/genetics , Adolescent , Adult , Aged , Child , Clone Cells , Diagnosis, Differential , Female , Hamartoma/pathology , Hemangioendothelioma/genetics , Hemangioendothelioma/pathology , Hemangioma, Capillary/pathology , Humans , Male , Middle Aged , Splenic Neoplasms/pathology , Young Adult
11.
Gene Ther ; 16(12): 1452-64, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19657370

ABSTRACT

X-linked chronic granulomatous disease (X-CGD) is an inherited immunodeficiency with absent phagocyte NADPH-oxidase activity caused by defects in the gene-encoding gp91(phox). Here, we evaluated strategies for less intensive conditioning for gene therapy of genetic blood disorders without selective advantage for gene correction, such as might be used in a human X-CGD protocol. We compared submyeloablative with ablative irradiation as conditioning in murine X-CGD, examining engraftment, oxidase activity and vector integration in mice transplanted with marrow transduced with a gamma-retroviral vector for gp91(phox) expression. The frequency of oxidase-positive neutrophils in the donor population was unexpectedly higher in many 300 cGy-conditioned mice compared with lethally irradiated recipients, as was the fraction of vector-marked donor secondary CFU-S12. Vector integration sites in marrow, spleen and secondary CFU-S12 DNA from primary recipients were enriched for cancer-associated genes, including Evi1, and integrations in or near cancer-associated genes were more frequent in marrow and secondary CFU-S12 from 300 cGy-conditioned mice compared with fully ablated mice. These findings support the concept that vector integration can confer a selection bias, and suggest that the intensity of the conditioning regimen may further influence the effects of vector integration on clonal selection in post-transplant engraftment and hematopoiesis.


Subject(s)
Bone Marrow/radiation effects , Gene Transfer Techniques , Genetic Vectors , Granulomatous Disease, Chronic/therapy , Hematopoiesis , Retroviridae/genetics , Transplantation Conditioning/methods , Animals , Female , Granulomatous Disease, Chronic/genetics , Hematopoietic Stem Cell Transplantation , Male , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , NADPH Oxidase 2 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Neoplasms/genetics , Neutrophils/metabolism , Stem Cells , Transduction, Genetic , Virus Integration
12.
Leukemia ; 22(7): 1308-19, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18480833

ABSTRACT

The 2001 World Health Organization (WHO)-sponsored classification of hematopoietic tumors has, for the first time, clearly defined a group of rare myeloid neoplasms termed myelodysplastic/myeloproliferative diseases (MDS/MPDs). This group includes three main entities, chronic myelomonocytic leukemia, atypical chronic myeloid leukemia and juvenile myelomonocytic leukemia, and also several less well defined, 'unclassifiable' disorders with MDS/MPN-like features. In the upcoming fourth edition of the WHO fascicle, due out later this year, the term 'MPD' is replaced by 'myeloproliferative neoplasm (MPN)'. Accordingly, the term MDS/MPD is being replaced by 'MDS/MPN' that will be used in this review. Although much progress has been made in understanding the molecular pathogenesis of myeloid neoplasms, most of the diseases included in the group of MDS/MPN still remain 'clinicopathologically assigned'. In other words, they can only be accurately categorized by a careful multiparametric approach that is based on the integration of bone marrow and peripheral blood morphology with other laboratory and clinical findings. The current 'spotlight' review provides practical guidelines, which should allow for a reproducible classification of these uncommon neoplasms when encountered in clinical practice.


Subject(s)
Myelodysplastic-Myeloproliferative Diseases/classification , Myeloproliferative Disorders/classification , Anemia, Refractory/classification , Chromatin/chemistry , Diagnosis, Differential , Humans , Janus Kinase 2/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/classification , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelomonocytic, Chronic/classification , Leukemia, Myelomonocytic, Chronic/diagnosis , Leukemia, Myelomonocytic, Chronic/genetics , Mutation , Prognosis
15.
Acta Myol ; 23(3): 151-3, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15938572

ABSTRACT

Neosporosis was diagnosed in a 2-month-old dog by indirect immunofluorescence antibody test (IFAT) and confirmed by means of histopathology and immunohistochemistry. The associated myositis was characterized by Major Histocompatibility Complex expression on some muscle fibres. This finding indicates an immunological activation, of the muscle cells that, acquiring Major Histocompatibility Complex expression, may, in some way, contribute to antigen presentation. A possible role of these glycoproteins in the pathogenesis of Neospora-associated myositis is discussed.


Subject(s)
Coccidiosis/veterinary , Dog Diseases/metabolism , Histocompatibility Antigens Class II/metabolism , Histocompatibility Antigens Class I/metabolism , Myositis/veterinary , Neospora , Animals , Coccidiosis/metabolism , Coccidiosis/pathology , Dog Diseases/parasitology , Dog Diseases/pathology , Dogs , Male , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myositis/metabolism , Myositis/parasitology
16.
Am J Kidney Dis ; 37(2): 388-99, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11157382

ABSTRACT

Most forms of interstitial nephritis are cell mediated and lack tubulointerstitial immune deposits. These forms include allergic, infectious, and idiopathic interstitial nephritis. Immune complex deposits in the tubular basement membranes and interstitium most commonly are encountered in conjunction with glomerular diseases. Predominantly tubulointerstitial immune deposits without significant glomerular involvement can occur in Sjögren's syndrome and in a small subset of lupus nephritis. We report eight unusual cases of tubulointerstitial nephritis with massive tubulointerstitial immune deposits occurring in adults with hypocomplementemia and no evidence of systemic lupus erythematosus or Sjögren's disease. Most patients were older men. The renal biopsy specimens manifested a spectrum of changes ranging from tubulointerstitial nephritis to atypical lymphoid hyperplasia to changes suggestive of marginal zone B-cell lymphoma. Chronic local antigenic stimulation may predispose to lymphoma in these cases, analogous to what is postulated to occur in cases of mucosa-associated lymphoid tissue (MALT) lymphomas in extranodal sites, such as salivary gland, stomach, and thyroid. The preferential tubulointerstitial immune deposition and significant interstitial plasma cell component suggest pathomechanisms that involve local immune complex formation.


Subject(s)
Glomerulonephritis, Membranoproliferative/pathology , Kidney Tubules/immunology , Kidney Tubules/pathology , Nephritis, Interstitial/pathology , Adult , Aged , Biopsy , Female , Fluorescent Antibody Technique , Gene Rearrangement, gamma-Chain T-Cell Antigen Receptor , Glomerulonephritis, Membranoproliferative/drug therapy , Glomerulonephritis, Membranoproliferative/immunology , Humans , Immunosuppression Therapy , Kidney/pathology , Kidney Tubules/ultrastructure , Lymphocytes , Lymphoma, B-Cell/pathology , Male , Middle Aged , Nephritis, Interstitial/drug therapy
18.
Neurosurgery ; 49(5): 1039-44; discussion 1044-5, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11846895

ABSTRACT

OBJECTIVE: Intracranial plasmacytomas are rare lesions that can arise from the calvarium, dura, or cranial base and exhibit a benign course unless associated with myeloma. Attention has recently been focused on the role of the cell adhesion molecules CD56 and CD31 in the pathogenesis of myeloma. No such information is available for intracranial plasmacytomas and myeloma-associated lesions. METHODS: We investigated the relationship between CD56 and CD31 expression, intracranial location, and progression to myeloma for a series of nine intracranial plasmacytomas (three dural, one calvarial, and five cranial base lesions). These parameters were also correlated with proliferation indices, as assessed by MIB-1 immunostaining of the histological sections. A single pathologist (AO) performed immunohistochemical analyses and reviewed all slides. RESULTS: Intracranial plasmacytomas presented more commonly in female patients (89%). The three dural lesions were CD56- and CD31-negative and exhibited MIB-1 staining of less than 10%; no patient developed myeloma or recurrence. Of the five cranial base lesions, three were CD56-positive, none was CD31-positive, and two exhibited MIB-1 labeling of more than 45%, with plasmablastic morphological features. Compared with other intracranial plasmacytomas, five of five patients with cranial base lesions developed bone marrow biopsy-proven myeloma (P < 0.05) within 8 months. The calvarial lesion was CD56- and CD31-positive, and the patient developed myeloma soon after diagnosis. Both of the two highly proliferative plasmablastic lesions recurred, one after gross total resection without radiotherapy and the other after a biopsy and 2000-cGy radiotherapy. CONCLUSION: Among intracranial plasmacytomas, cranial base location was the strongest predictor of the development of multiple myeloma. Expression of the cell adhesion molecules CD31 and CD56 was not predictive of outcome. Extramedullary dural-based lesions were CD56-negative and were not associated with myeloma. A high proliferation index and plasmablastic morphological features were predictive of a short time to recurrence and aggressive behavior. We recommend 4050- to 5040-cGy fractionated radiotherapy for all intracranial plasma cell neoplasms and gross total resection for non-cranial base lesions.


Subject(s)
Brain Neoplasms/pathology , Multiple Myeloma/pathology , Plasmacytoma/pathology , Adult , Aged , Aged, 80 and over , CD56 Antigen/analysis , Dura Mater/pathology , Female , Humans , Immunoenzyme Techniques , Magnetic Resonance Imaging , Male , Middle Aged , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Prognosis , Skull Base Neoplasms/pathology , Skull Neoplasms/pathology
19.
Leuk Lymphoma ; 39(1-2): 1-18, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10975379

ABSTRACT

Acute hyperleukocytic leukemias (AHL) are associated with a very high early mortality rate mostly due to respiratory failure or intracranial bleeding. The pathophysiological process leading to these complications is called leukostasis but the biological mechanisms underlying its development and progression remain unclear. Although traditionally related to "over-crowding" of leukemic blasts in the capillaries of the microcirculation, leukostasis is likely to result from direct endothelial cell damage. This damage is probably mediated by soluble cytokines released during the interaction between leukemic cells and vascular endothelium and by the subsequent migration of leukemic blasts in the perivascular space. Leukemic cell's ability to respond to chemotactic cytokines and their expression of specific adhesion molecules are probably more important in determining whether leukostasis will develop than the number of circulating blasts. This could explain why leukostasis does not develop in all patients with AHL. The identification of the adhesion molecules, cytokines and receptors mediating endothelial cell damage in AHL should become a priority if therapeutic improvements are desired. Leukapheresis is widely used but it is unclear whether it provides additional benefit to a simpler and less invasive intervention with allopurinol, hydroxyurea and intravenous fluids. Cranial irradiation is not generally recommended. Induction chemotherapy should be started without delay. It is hoped that specific pharmacological inhibitors of the interaction between leukemic cells and vascular endothelium will result in an improved outcome for this very high-risk population.


Subject(s)
Leukemia, Myeloid/blood , Leukostasis/etiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/blood , Acute Disease , Adolescent , Adult , Child , Child, Preschool , Cytogenetics , Cytokines/adverse effects , Cytokines/blood , Endothelium, Vascular/injuries , Endothelium, Vascular/pathology , Female , Humans , Infant , Leukemia, Myeloid/complications , Leukemia, Myeloid/pathology , Leukostasis/blood , Leukostasis/therapy , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
20.
Leuk Lymphoma ; 38(5-6): 563-76, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10953978

ABSTRACT

Posttransplant lymphoproliferative disorders (PTLDs), which are highly associated with Epstein-Barr virus infection, have a low frequency of molecular genetic abnormalities. Recently it has been suggested certain EBV substrains may be associated with specific lymphoma subtypes. The goals of our study were two fold: 1) to determine the prevalence of EBNA-1 substrains and prognostic utility in PTLD and 2) to determine the incidence of p53 gene mutations and p53 protein overexpression in 32 EBV-positive PTLD cases. Tumor DNA was sequenced to identify EBNA-1 substrains at codon 487 and p53 gene mutations in exons 5-8. The PTLD samples contained the following EBNA-1 substrains: P-thr in 17/32 (53%), P-ala in 11/32 (34%), and V-leu in 4/32 (13%). More heterogeneity within major subtypes was seen in the PTLD cases than in the referral group. A second group of 25 referral (non-PTLD) samples including infectious mononucleosis (6) and sequential EBV positive virology samples (19) contained P-thr in 17/25 (68%); P-ala in 2/25 (8%); and V-leu in 6/25 (24%). In the 29 B-cell PTLD the time to presentation was an average of 13.3 months in the P-ala group, 16.6 months in the P-thr group, and 40.6 months in the V-leu group: (p>0.05). There was no difference in survival in patients (median overall--60 months) between the three different substrains of EBNA-1 (Log rank test, p=0.39). One of 31 (4.1%) cases (a diffuse large cell B-cell) had a p53 mutation. Seven of 31 (23%) cases (all B-cell), including the p53 mutated case, had over-expression of p53 protein. We conclude EBNA-1 substrains vary in PTLD and suggest the pattern reflects the geographical incidence of substrains in the region. We also conclude p53 mutations are not a significant molecular genetic abnormality in PTLD.


Subject(s)
Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/virology , Genes, p53 , Herpesvirus 4, Human/isolation & purification , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/virology , Epstein-Barr Virus Infections/etiology , Herpesvirus 4, Human/genetics , Humans , Immunosuppression Therapy/adverse effects , Lymphoproliferative Disorders/etiology , Lymphoproliferative Disorders/mortality , Mutation , Organ Transplantation/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...