throbber
Editorials and Perspectives
`
`ers in myeloproliferative diseases: relationships with JAK2
`V617 F status, clonality, and antiphospholipid antibodies. J
`Thromb Haemost 2007;5:1679-85.
`17. Falanga A, Marchetti M, Vignoli A, Balducci D, Russo L,
`Guerini V, et al. V617F JAK-2 mutation in patients with
`essential thrombocythemia: relation to platelet, granulo-
`cyte, and plasma hemostatic and inflammatory molecules.
`Exp Hematol 2007;35:702-11.
`18. Arellano-Rodrigo E, Alvarez-Larran A, Reverter JC,
`Colomer D, Villamor N, Bellosillo B, et al. Platelet turnover,
`coagulation factors, and soluble markers of platelet and
`endothelial activation in essential thrombocythemia: rela-
`tionship with thrombosis occurrence and JAK2 V617F allele
`burden. Am J Hematol 2009;84:102-8.
`19. Trappenburg MC, van Schilfgaarde M, Marchetti M, Spronk
`HM, ten Cate H, Leyte A, et al. Elevated procoagulant
`microparticles expressing endothelial and platelet markers
`in essential thrombocythemia. Haematologica 2009;
`94:911-8.
`20. Marchetti M, Castoldi E, Spronk HM, van OR, Balducci D,
`Barbui T, et al. Thrombin generation and activated protein
`C resistance in patients with essential thrombocythemia
`and polycythemia vera. Blood 2008;112:4061-8.
`21. Kornberg A, Rahimi-Levene N, Yona R, Mor A,
`Rachmilewitz EA. Enhanced generation of monocyte tissue
`factor and increased plasma prothrombin fragment 1+2 lev-
`els in patients with polycythemia vera: mechanism of acti-
`vation of blood coagulation. Am J Hematol 1997;56:5-11.
`22. Passamonti F, Rumi E, Pietra D, la Porta MG, Boveri E,
`
`Pascutto C, et al. Relation between JAK2 (V617F) mutation
`status, granulocyte activation, and constitutive mobilization
`of CD34+ cells into peripheral blood in myeloproliferative
`disorders. Blood 2006;107:3676-82.
`23. Alvarez-Larrán A, Arellano-Rodrigo E, Reverter JC,
`Domingo A, Villamor N, Colomer D, et al. Increased
`platelet, leukocyte, and coagulation activation in primary
`myelofibrosis. Ann Hematol 2008;87:269-76.
`24. Leibundgut EO, Horn MP, Brunold C, Pfanner-Meyer B,
`Marti D, Hirsiger H, et al. Hematopoietic and endothelial
`progenitor cell trafficking in patients with myeloprolifera-
`tive diseases. Haematologica 2006;91:1465-72.
`25. Sozer S, Fiel MI, Schiano T, Xu M, Mascarenhas J, Hoffman
`R. The presence of JAK2V617F mutation in the liver
`endothelial cells of patients with Budd-Chiari syndrome.
`Blood 2009;113:5246-9.
`26. Pieri L, Bogani C, Guglielmelli P, Zingariello M, Rana RA,
`Bartalucci N, et al. The JAK2V617F mutation induces consti-
`tutive activation and agonist hypersensitivity in basophils of
`polycythemia vera. Haematologica 2009;94:1537-475.
`27. Wang J, Ishii T, Zhang W, Sozer S, Dai Y, Mascarenhas J, et
`al. Involvement of mast cells by the malignant process in
`patients with Philadelphia chromosome negative myelopro-
`liferative neoplasms. Leukemia 23:1577-86.
`28. Ishii T, Wang J, Zhang W, Mascarenhas J, Hoffman R, Dai Y,
`et al. Pivotal role of mast cells in pruritogenesis in patients
`with myeloproliferative disorders. Blood 2009;113:5942-50.
`29. Kovanen PT. Mast cells: multipotent local effector cells in
`atherothrombosis. Immunol Rev 2007;217:105-22.
`
`Mantle cell lymphoma
`Stefano A. Pileri1 and Brunangelo Falini2
`
`1Department of Haematology and Oncological Sciences “L. and A. Seràgnoli”, Chair of Pathology and Haemolymphopathology
`Unit, Bologna University School of Medicine; 2Department of Haematology, Laboratory of Hematopathology, Perugia University
`School of Medicine. E-mail: stefano.pileri@unibo.it doi:10.3324/haematol.2009.013359
`
`In 1982, Weisenburger et al. first introduced the con-
`
`cept of mantle-zone lymphoma.1 According to the ter-
`minology used at that time, this was regarded as a
`variant of intermediate lymphocytic lymphoma that pro-
`liferated as wide mantles around non-neoplastic appear-
`ing germinal centers (GC). One year later, Swerdlow et al.
`found that the pattern described above was part of the
`spectrum of the centrocytic lymphoma of the Kiel
`Classification and might correspond to initial lymph
`node involvement by the tumor.2,3 In 1985, Pileri et al.
`reported on 18 cases of small B-cell lymphomas display-
`ing a mantle-fashion growth around reactive GC, which
`turned out to be quite heterogeneous on closer examina-
`tion.4 In fact, at disease presentation 13 of the 18 cases
`displayed cytological and immunological findings consis-
`tent with centrocytic lymphoma, while the remaining
`ones corresponded to neoplasms that nowadays would
`be diagnosed as marginal zone lymphoma. Interestingly,
`the centrocytic lymphomas were CD5+, FMC7+ and
`CD10– and showed progression to a diffuse growth pat-
`tern in follow-up biopsies. Two important conclusions
`were reached: (i) the mantle-fashion growth was pro-
`duced by different types of B-cell lymphoma and could
`not be used as a diagnostic criterion, and (ii) conversely
`to what is reported in the Kiel Classification,3 centrocyt-
`ic lymphomas are derived from a normal counterpart
`other than GC, possibly related to mantle cells. In 1992,
`the International Lymphoma Study Group (ILSG) made
`
`its first experiment to overcome the discrepancies in
`terms of lymphoma classification that had hampered
`communication across the Atlantic for some decades.5 In
`particular, the American and European ILSG members
`agreed on the existence of a tumor derived from mantle
`B cells that was consequently termed mantle-cell lym-
`phoma (MCL). The criteria for its recognition were draft-
`ed and included 2 years later in the Revised European-
`American Lymphoma Classification that led the way to
`the latest edition of the World Health Organization
`(WHO) Classification of Tumours of Haematopoietic
`and Lymphoid Tissues.6,7 The present article will focus on
`the diagnosis and bio-pathology of MCL in the light of
`the criteria of the WHO Classification7 and recent reports
`in the literature including those of Quintinilla-Martinez et
`al., Mozos et al. and Dictor et al. in this issue of the jour-
`nal.8-10 It will not include the still ongoing debate on the
`multiple options proposed for the treatment of MCL
`patients, which often have limited efficacy (comprehen-
`sively reviewed by Ghielmini et al.).11
`
`Definition
`MCL is a distinct entity, representing from 3-10% of
`non-Hodgkin’s lymphomas and more frequently affect-
`ing middle-aged to older males (male:female ratio = 2-
`7:1).7,12 It is listed among peripheral B-cell lymphomas, is
`usually composed of small to medium-sized elements
`with irregular nuclear contours and CCDN1 transloca-
`
`| 1488 |
`
`haematologica | 2009; 94(11)
`
`Coalition for Affordable Drugs IV LLC – Exhibit 1009
`
`

`
`Editorials and Perspectives
`
`tion, and is thought to stem from peripheral B cells of
`the inner mantle-zone of secondary follicles, mostly of
`naïve pre-GC type.7,12 MCL is generally regarded as an
`aggressive, incurable disease with the median survival
`of affected patients being 3-4 years.12
`
`Clinical features
`MCL more often presents in stage III-IV with lym-
`phadenopathy, hepatosplenomegaly, bone-marrow
`involvement, and leukemic spread.7,12 The latter two
`findings can be missed by morphological studies alone
`and are shown by, respectively, immunohistochemistry
`and FACS analysis in most if not all instances.7,12
`Waldayer’s ring and the gastro-intestinal tract are fre-
`quently affected.7,12 Most cases of multiple lymphoma-
`tous polyposis correspond to MCL.
`
`Morphology
`MCL usually consists of small to medium-sized lym-
`phoid elements with irregular nuclear contours, some-
`what dispersed chromatin, inconspicuous nucleoli, and
`scant cytoplasm.7,12,13 Within this context, centroblasts,
`immunoblasts, prolymphocytes and para-immuno-
`blasts are not encountered. This is relevant for the dif-
`ferential diagnosis from follicular lymphoma, lympho-
`plasmacytic
`lymphoma and chronic
`lymphocytic
`leukemia/small lymphocytic lymphoma, which by def-
`inition, contain centroblasts, immunoblasts and prolym-
`phocytes/para-immunoblasts, respectively. Comprised
`within the neoplastic population, hyaline small vessels
`and/or epithelioid histiocytes are frequently found.
`Clusters of plasma cells can be seen: they are usually
`reactive (polytypic at immunohistochemistry), only
`exceptionally representing a feature of tumor differenti-
`ation with obvious monoclonality and tendency to
`accumulate in the center of lymphomatous nodules.14
`The number of mitotic figures varies, although it is usu-
`ally low to moderate (see below).
`In the lymph node, spleen and Waldayer’s ring, neo-
`plastic cells give rise to a mantle-fashion, nodular or –
`more frequently – diffuse growth pattern.7,12,13 Notably,
`repeated biopsies demonstrate that such patterns corre-
`spond to disease progression. In fact, in the early phas-
`es the tumor substitutes the mantle-zone; then, it
`invades reactive GC producing a nodular pattern; final-
`ly, the nodules merge together causing diffuse efface-
`ment of the organ’s structure. The bone marrow can be
`variably involved: paratrabecular infiltrates are the com-
`monest finding.
`Besides the classical type described above, there are
`some cytological and/or morphological variants of MCL
`that can cause diagnostic problems. These are termed
`blastoid, pleomorphic, small cell, and marginal zone-
`like.7,12,13 The first consists of slightly larger cells resem-
`bling lymphoblasts with dispersed chromatin and a
`high mitotic rate. Blastoid MCL is clinically aggressive,
`as is the pleomorphic form, which is characterized by a
`cell population that is highly variable in size and shape,
`with frequent mitotic figures and a proportion of rather
`large elements showing oval to irregular nuclear con-
`tours, pale cytoplasm and at times prominent nucleoli.
`Both the blastoid and pleomorphic forms occur de novo,
`
`rarely representing morphological progression of the
`disease. The small cell variant is composed of small,
`round lymphocytes with more clumped chromatin,
`mimicking chronic lymphocytic leukemia/small lym-
`phocytic lymphoma. Occasionally, the small lym-
`phomatous elements can be almost exclusively restrict-
`ed to the inner mantle zones or to narrow mantles, a
`pattern termed in situ MCL and thought to have a more
`indolent behavior.15 Finally, the marginal zone-like sub-
`type displays prominent foci of cells with abundant pale
`cytoplasm resembling marginal zone or monocytoid B-
`elements.7,12 Notably, these cases are at times character-
`ized by splenomegaly, intrasinusoidal bone-marrow
`infiltration and a leukemic picture in the absence of
`lymph node involvement. They can be easily confused
`with splenic marginal zone lymphoma and seem to
`have a very indolent course. As for in situ MCL, the diag-
`nosis of this rare form of the tumor is feasible only upon
`immunohistochemical detection of cyclin D1 (see
`below).
`
`Immunophenotype
`MCL is characterized by expression of the B-cell
`markers CD19, CD20, CD22, CD79a
`and
`BSAP/PAX5.7,12,13 Notably, CD20 is strongly expressed
`on the surface of neoplastic cells, a feature that can be
`useful for the differential diagnosis from chronic lym-
`phocytic
`leukemia/small
`lymphocytic
`lymphoma,
`which shows weak CD20 positivity in the small cell
`component.13 The search for immunoglobulin (Ig) heavy
`and light chains usually reveals IgM/D positivity with
`more frequent lambda restriction.7,12 Neoplastic cells are
`usually CD5+, FMC-7+, BCL-2+, CD10–, BCL-6–, and
`IFR4–, with occasional weak expression of CD23.7,12,13 In
`the cases with nodular growth, residual GC cells
`(CD10+, BCL-6+) can be detected admixed with the neo-
`plastic population. Furthermore, aberrant phenotypes
`have been reported in MCL (such as negativity for CD5
`and positivity for CD10 and/or BCL-6), sometimes
`associated with the blastoid/pleomorphic variants or
`occurring in conjunction with BCL6/3q27 transloca-
`tions.7 Stains for CD21 and CD35 usually show loose
`follicular dendritic cell meshworks. In the light of this,
`although the phenotypic profile of MCL is distinctive, it
`is not entirely pathognomonic and as such cannot
`always assist in the differentiation of MCL from follicu-
`lar lymphoma, lymphoplasmacytic lymphoma, margin-
`al zone lymphoma and chronic lymphocytic leu-
`kemia/small lymphocytic lymphoma. For this purpose,
`the most useful examination is the strong expression of
`cyclin D1, which is never found in the above mentioned
`categories of B-type non-Hodgkin’s lymphomas.7,12,13
`This attribute is sustained by the t(11;14)(q13;q32)
`translocation (see below).7,12,13 Notably, cyclin D1
`expression is lacking in a small percentage of cases of
`MCL: because of practical and conceptual implications,
`these cases will be treated separately (see below). Last
`but not least, cyclin D1 expression is found in hairy cell
`leukemia (although weak) and in about 16% of plasma
`cell myelomas, also carrying t(11;14)(q13;q32).13 When
`commercially available, the antibody produced by one
`of the authors (Falini)16 and raised against the IRTA-1
`
`haematologica | 2009; 94(11)
`
`| 1489 |
`
`

`
`Editorials and Perspectives
`
`receptor physiologically expressed by marginal zone B-
`cells, could represent a further tool to aid the differential
`diagnosis between MCL (IRTA-1–) and nodal marginal
`zone lymphoma (IRTA-1+ in about 80% of cases).17
`
`The t(11;14)(q13;q32) translocation
`The t(11;14)(q13;q32) translocation is regarded as the
`primary genetic event in MCL.7,12 It can easily be demon-
`strated by fluorescent in situ hybridization (FISH) and
`involves the Ig heavy chain gene (IGH@) at the 14q32
`locus and a region at 11q13 designated BCL1.12 The
`expression of CCND1, located in the latter and encoding
`for cyclin D1, is deregulated by the translocation.7,12 This
`causes over-expression of cyclin D1 both at the mRNA
`and protein levels in MCL. Cyclin D1 plays an important
`role in the cell cycle regulation of G1-S transition follow-
`ing mitotic growth factor signaling.7 Unlike in MCL, in
`normal cells cyclin D1 is transiently expressed and binds
`to CDK4 and CDK6 to form a CDK/cyclin complex able
`to phosphorylate the tumor suppressor gene retinoblas-
`toma (RB1) facilitating cell cycle progression.12 De-
`regulated expression of cyclin D1 is thought to over-
`come the effect of RB1 and p27/kip, leading to the devel-
`opment of MCL.7,12 Some tumors over-express shorter
`cyclin D1 transcripts, usually generated by secondary 3’
`
`rearrangements in the CCD1 locus.7,12 These truncated
`transcripts cause very strong protein expression, high
`proliferative activity and a more aggressive clinical
`course.7,12
`Notably, it has been reported that inactivating muta-
`tions of the ATM and/or CHK2 genes (recorded in 40-
`75% of MCL) are at times found in the germline of
`some patients.7,12 This might be relevant to the develop-
`ment of the tumor: in fact, such mutations cause dereg-
`ulation of the DNA damage response pathway and
`increased genomic instability.7,12 In particular, t(11;14) –
`which should affect immature/naïve B cells as suggest-
`ed by its detection in the peripheral blood of 1-2% of
`healthy individuals – would occur in the setting of a
`population carrying these mutations and lead to consti-
`tutive deregulation of cyclin D1 and early expansions of
`tumor B cells in the mantle zone of lymphoid follicles.
`This might also explain why MCL is one of the malig-
`nant neoplasms with the highest level of genomic insta-
`bility, which might cause additional oncogenic events
`needed for the expansion of MCL cells with classical
`morphology, as well as alterations of genes involved in
`cell cycle regulation and senescence regulatory path-
`ways that would lead to more aggressive variants of
`MCL.7,12,18 In particular, there is a high number of non-
`
`Figure 1. Schematic representation of the postulated steps of the pathogenesis of MCL, shown in conjunction with the main immuno-
`morphological findings. The red immunostain, corresponds to cyclin D1 determined by the alkaline phosphatase anti-alkaline phos-
`phatase complexes technique. The naïve B-lymphocyte is stained for CD20 by immunoperoxidase. The examples of “classic” MCL and
`“blastoid” MCL are stained with Giemsa, while the “pleomorphic” one is stained with hematoxylin and eosin.
`
`| 1490 |
`
`haematologica | 2009; 94(11)
`
`

`
`Editorials and Perspectives
`
`random secondary chromosomal aberrations in MCL,
`including gains of 3q26, 7p21 and 8p24 (MYC), and
`losses of 1p13-p31, 6q23q27, 9p21, 11q22-q23, 13q11-
`q13, 13q14-q34, and 17p13-ter.7,18 Trisomy 12 is detect-
`ed in about 25% of cases. Tetraploid clones are more
`common in the pleomorphic and blastoid variants than
`in the classical type (80%, 36% and 8%, respective-
`ly).7,12 t(8;14)(q24;q32) involving MYC is rare, but asso-
`ciated with a more aggressive clinical course. Finally,
`MCL carries frequent mutations of TP53, homozygous
`deletions of the INK4a/ARF, amplifications of the BMI1
`polycomb and CDK4 genes, and occasional micro-dele-
`tions of the RB1 gene, all producing high proliferative
`activity.7,12
`The possible steps of the molecular pathogenesis of
`MCL are outlined in Figure 1 in relation to immuno-
`morphological findings.
`
`Antigen receptor genes
`Ig genes are rearranged. IGH@ genes are unmutated
`in most cases, although mutations are found in a minor-
`ity of MCL.7,12 The load of the latter is lower than in
`mutated chronic lymphocytic leukemia.7,12 In addition,
`in MCL IGH@ somatic mutations do not correlate with
`ZAP70 expression and survival.7,12 A biased usage of
`IGH@ genes has been reported in some cases, suggest-
`ing that MCL may originate from specific B-cell subsets.
`Unlike in chronic lymphocytic leukemia, in MCL VH3-
`21 usage occurs in unmutated cases and bears a lower
`amount of genomic imbalances and a better prognosis.7
`
`Gene expression profile
`In 2003,
`the Lymphoma/Leukemia Molecular
`Profiling Project group showed that MCL has a specific
`gene signature that differs from the signatures of small
`lymphocytic lymphoma and diffuse large B-cell lym-
`phoma of both the GC B cell-like and activated B cell-
`like types.19 Interestingly, this signature encompassed
`the usual cyclin D1-positive cases and a small subgroup
`of cyclin D1-negative MCL. Besides elucidating the
`pathogenesis of the tumor, the gene expression profile
`(GEP) revealed 20 proliferation-associated genes whose
`expression measurement identified subsets of patients
`whose median survival differed by more than 5 years.
`Differences in cyclin D1 mRNA abundance synergized
`with INK4a/ARF locus deletions to dictate tumor prolif-
`eration rate and survival. Two years later, the same
`group reported on six cases of MCL lacking cyclin D1
`that exhibited the same clinical, morphological and GEP
`features as classical MCL but did not carry the
`t(11;18)(q12;q32), as determined by FISH.20 These cases
`did, however, express either cyclin D2 or cyclin D3 at
`both the mRNA and protein levels, suggesting that up-
`regulation of these cyclins may substitute for cyclin D1
`in the pathogenesis of MCL.
`
`Cyclin D1-negative mantle cell lymphoma
`Since cyclins D2 and D3 are also expressed by normal
`B cells and most, if not all, B-cell non-Hodgkin’s lym-
`phoma,8 although at lower levels,12 these markers can-
`not be confidently used for differentiating cyclin D1-
`negative MCL from other small B-cell tumors.8,12 In
`
`2008, this led Campo et al. to conclude that “the only
`reliable criteria to establish the diagnosis of cyclin D1-
`negative MCL seemed to be the microarray profile that
`is limited to research environments”.12 In this issue of
`the journal, three reports focus on tools that can allow
`the easy identification of such cases and can be applied
`in most histopathology labotatories.8-10 First of all,
`Quintinilla-Martinez et al. propose FISH analysis and/or
`quantitative reverse transcriptase polymerase chain
`reaction as a means for making the differential diagno-
`sis of cyclin D1-negative MCL.8 This approach is based
`on the one hand on previous observations of bone fide
`cyclins D1–/D2+ MCL carrying translocations involving
`the CCND2
`locus with
`either
`the
`IGK@
`[t(2;12)(p12;p13)] or IGH@ locus [t12;14](p13;q32)], not
`recorded in either chronic lymphocytic leukemia or
`CD5+ mantle zone lymphoma, and, on the other hand,
`on the at least 10-times higher mRNA levels of cyclin
`D2 in D1–/D2+ MCL in comparison with normal lym-
`phoid tissue, follicular lymphoma, mantle zone lym-
`phoma and chronic lymphoma and chronic lymphocy-
`tuc leukemia.8 Mozos et al. candidate the expression of
`the neural transcription factor SOX11 as a powerful bio-
`marker for the identification of cyclin D1-negative
`MCL.9 In fact, GEP showed significantly higher expres-
`sion of the SOX11 gene in classical MCL than in
`Burkitt’s lymphoma, diffuse large B-cell lymphoma, pri-
`mary mediastinal B-cell lymphoma and follicular lym-
`phoma.9 However, one third of cases of Burkitt’s lym-
`phoma revealed SOX11 mRNA levels similar to those in
`MCL, thus indicating that SOX11 over-expression is not
`restricted to MCL.9 These findings were corroborated
`by immunohistochemical studies that showed nuclear
`SOX11-positivity in most, if not all, MCL tested, includ-
`ing 12 cyclin D1-negative cases.9 Interestingly, 25% of
`Burkitt’s lymphomas, all examples of B- and T-lym-
`phoblastic
`leukemia/lymphoma,
`one
`classical
`Hodgkin’s
`lymphoma and 2/3 T-prolymphocytic
`leukemias also turned out to be positive.9 Equivalent
`results were reported by Dictor et al., who immunos-
`tained 172 specimens for the SOX11 N and C termini.10
`Their findings indicate that SOX11 can represent a use-
`ful tool for the differentiation of MCL from other small
`B-cell lymphomas in general but is certainly of pivotal
`importance for identifying the cyclin D1-negative cases.
`Of course, it should be integrated in a panel of markers
`in order to avoid any confusion between a potential
`cyclin D1-negative blastoid MCL and lymphoblastic
`leukemia/lymphoma, which may be confused morpho-
`logically but are easily differentiated by the global phe-
`notypic profile.
`
`Prognostic factors
`Apart from the rare forms with an indolent behavior
`reported above, MCL has a particularly poor outcome7,12
`Several attempts have been made to subdivide patients
`into different risk groups. In particular, the European
`MCL Network has shown that the number of mitotic
`figures and Ki-67 marking can allow the identification
`of three subgroups with different proliferation rates
`(mitotic indices: <25, 25-49, and >50/mm2 and Ki-67
`indices: <10%, 10-40%, and <40%, respectively) signif-
`
`haematologica | 2009; 94(11)
`
`| 1491 |
`
`

`
`Editorials and Perspectives
`
`icantly different outcomes.21 Such differences are con-
`served in advanced-stage MCL patients treated with
`anti-CD20 immuno-chemotherapy.22 Interestingly, these
`findings are in keeping with the above reported GEP
`data19 and point to the prognostic value of proliferation
`in MCL. Additional bio-pathological adverse prognosti-
`cators – which may be independent of proliferative
`activity – have been reported, such as blastoid/pleomor-
`phic morphology, trisomy 12, karyotype complexity,
`TP53 mutation/over-expression/loss, gains of chromo-
`some 3q, deletions of 9p, and a series of clinical param-
`eters including overt peripheral blood involvement.7,12
`Recently, a clinical MCL index (MIPI), based on age,
`ECOG, performance status, lactate dehydrogenase val-
`ues and leukocyte count, and a five-gene model (RAN,
`MYC, TNFRSF10B, POLE2, and SLC29A2) applicable to
`both frozen and routine samples and based on quantita-
`tive reverse transcriptase polymerase chain reaction
`analysis, have been proposed as further prognostic indi-
`cators.23,24
`
`This manuscript was supported by grants from BolognAIL,
`Centro Interdipartimentale di Ricerche sul Cancro “Giorgio
`Prodi” and Fondazione Cassa di Risparmio in Bologna
`(Bologna, Italy).
`
`Dr. Stefano A. Pileri is Full Professor of Pathology and
`Director of the Hematopathology Unit at Bologna University.
`Dr. Brunangelo Falini is Full Professor of Hematology at
`Perugia University.
`
`References
`
`1. Weisenburger DD, Kim H, Rappaport H. Mantle-zone lym-
`phoma: a follicular variant of intermediate lymphocytic
`lymphoma. Cancer 1982;49:1429-38.
`2. Swerdlow SH, Habeshaw JA, Murray LJ, Dhaliwal HS,
`Lister TA, Stansfeld AG. Centrocytic lymphoma: a distinct
`clinicopathologic and immunologic entity. A multiparame-
`ter study of 18 cases at diagnosis and relapse. Am J Pathol
`1983;113:181-97.
`3. Lennert K. Malignant lymphomas other than Hodgkin’s dis-
`ease. New York: Springer-Verlag; 1978.
`4. Pileri S, Rivano MT, Gobbi M, Taruscio D, Lennert K.
`Neoplastic and reactive follicles within B-cell malignant
`lymphomas. A morphological and immunological study of
`30 cases. Hematol Oncol 1985;3:243-60.
`5. Banks PM, Chan J, Cleary ML, Delsol G, De Wolf-Peeters C,
`Gatter K, et al. Mantle cell lymphoma. A proposal for unifi-
`cation of morphologic, immunologic, and molecular data.
`Am J Surg Pathol 1992;16:637-40.
`6. Harris NL, Jaffe ES, Stein H, Banks PM, Chan JK, Cleary ML,
`et al. A revised European-American classification of lym-
`phoid neoplasms: a proposal from the International
`Lymphoma Study Group. Blood 1994;84:1361-92.
`7. Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein
`H, et al. WHO classification of tumours of haematopoietic
`and lymphoid tissues. Lyon: IARC, 2008.
`8. Quintanilla-Martinez L, Slotta-Huspenina J, Koch I, Klier M,
`
`His ED, de Leval L, et al. Differential diagnosis of cyclin D2+
`mantle cell lymphoma based on fluorescence in situ
`hybridization and quantitative real-time-polymerase chain
`reaction. Haematologica 2009;94:1595-8.
`9. Mozos A, Royo C, Hartmann E, DeJong S, Baró C, Valera A,
`et al. SOX11 expression is highly specific for mantle cell
`lymphoma and identifies the cyclin D1 negative subtype.
`Haematologica 2009;94:1555-62.
`10. Dictor M, Ek S, Sundberg M, Warenholt J, György C,
`Sernbo S, et al. Strong lymphoid nuclear expression of
`SOX11 transcription factor defines lymphoblastic neo-
`plasms, mantle cell lymphoma and Burkitt’s lymphoma.
`Haematologica 2009;94:1563-8.
`11. Ghielmini M, Zucca E. How I treat mantle cell lymphoma.
`Blood 2009;114:1469-76.
`12. Jares P, Campo E. Advances in the understanding of mantle
`cell lymphoma. Br J Haematol 2008;142:149-65.
`13. Pileri SA, Ascani S, Sabattini E, Fraternali-Orcioni G, Poggi
`S, Piccioli M, et al. The pathologist’s view point. Part I–indo-
`lent lymphomas. Haematologica 2000;85:1291-307.
`14. Young KH, Chan WC, Fu K, Iqbal J, Sanger WG, Ratashak
`A, et al. Mantle cell lymphoma with plasma cell differentia-
`tion. Am J Surg Pathol 2006;30:954-61.
`15. Richard P, Vassallo J, Valmary S, Missoury R, Delsol G,
`Brousset P. “In situ-like” mantle cell lymphoma: a report of
`two cases. J Clin Pathol 2006;59:995-6.
`16. Falini B, Tiacci E, Pucciarini A, Bigerna B, Kurth J,
`Hatzivassiliou G, et al. Expression of the IRTA1 receptor
`identifies intraepithelial and subepithelial marginal zone B
`cells of the mucosa-associated lymphoid tissue (MALT).
`Blood 2003;102:3684-92.
`17. Went P, Ascani S, Strom E, Brorson SH, Musso M, Zinzani
`PL, et al. Nodal marginal-zone lymphoma associated with
`monoclonal light-chain and heavy-chain deposition disease.
`Lancet Oncol 2004;5:381-3.
`18. Salaverria I, Zettl A, Bea S, Moreno V, Valls J, Hartmann E,
`et al. Specific secondary genetic alterations in mantle cell
`lymphoma provide prognostic information independent of
`the gene expression-based proliferation signature. J Clin
`Oncol 2007;25:1216-22.
`19. Rosenwald A, Wright G, Wiestner A, Chan WC, Connors
`JM, Campo E, et al. The proliferation gene expression signa-
`ture is a quantitative integrator of oncogenic events that pre-
`dicts survival in mantle cell lymphoma. Cancer Cell 2003;
`3:185-97.
`20. Fu K, Weisenburger DD, Greiner TC, Dave S, Wright G,
`Rosenwald A, et al. Cyclin D1-negative mantle cell lym-
`phoma: a clinicopathologic study based on gene expression
`profiling. Blood 2005;106:4315-21.
`21. Tiemann M, Schrader C, Klapper W, Dreyling MH, Campo
`E, Norton A, et al. Histopathology, cell proliferation indices
`and clinical outcome in 304 patients with mantle cell lym-
`phoma (MCL): a clinicopathological study from the
`European MCL Network. Br J Haematol 2005;131:29-38.
`22. Determann O, Hoster E, Ott G, Wolfram Bernd H,
`Loddenkemper C, Leo Hansmann M, et al. Ki-67 predicts
`outcome in advanced-stage mantle cell lymphoma patients
`treated with anti-CD20 immunochemotherapy: results
`from randomized trials of the European MCL Network and
`the German Low Grade Lymphoma Study Group. Blood
`2008;111:2385-7.
`23. Hoster E, Dreyling M, Klapper W, Gisselbrecht C, van Hoof
`A, Kluin-Nelemans HC, et al. A new prognostic index
`(MIPI) for patients with advanced-stage mantle cell lym-
`phoma. Blood 2008;111:558-65.
`24. Hartmann E, Fernandez V, Moreno V, Valls J, Hernandez L,
`Bosch F, et al. Five-gene model to predict survival in mantle-
`cell lymphoma using frozen or formalin-fixed, paraffin-
`embedded tissue. J Clin Oncol 2008;26:4966-72.
`
`| 1492 |
`
`haematologica | 2009; 94(11)

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket