ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Pathobiology of Burkitt lymphoma

Pathobiology of Burkitt lymphoma
Literature review current through: Jan 2024.
This topic last updated: Nov 28, 2023.

INTRODUCTION — Burkitt lymphoma (BL) is a highly aggressive B cell neoplasm characterized by the translocation and deregulation of the MYC gene on chromosome 8. Three distinct clinical forms of BL are recognized: endemic, sporadic, and immunodeficiency associated. Although they are histologically identical and have similar clinical behavior, there are differences in epidemiology, clinical presentation, and genetic features among the three forms. (See "Epidemiology, clinical manifestations, pathologic features, and diagnosis of Burkitt lymphoma", section on 'Clinical features'.)

Current lymphoma classification systems consider BL with tissue-only disease and BL with peripheral blood involvement different manifestations of the same disease [1,2]. Classification of BL and other lymphomas is discussed separately. (See "Classification of hematopoietic neoplasms", section on 'Lymphoid neoplasms'.)

The pathogenesis of BL is discussed in this topic.

Clinical manifestations, diagnosis, and treatment of BL are presented separately. (See "Epidemiology, clinical manifestations, pathologic features, and diagnosis of Burkitt lymphoma" and "Treatment of Burkitt leukemia/lymphoma in adults".)

CELL OF ORIGIN — BL is derived from germinal or post-germinal center B cells. The germinal or post-germinal center ancestry of these cells is principally supported by the identification of somatic mutations in the variable region of the immunoglobulin genes (IgV), which serve as a marker of germinal center transit, as well as the tumor cell immunophenotype. Some studies have demonstrated ongoing somatic mutation, a process that is normally restricted to germinal center B cells. (See "Normal B and T lymphocyte development" and "Epidemiology, clinical manifestations, pathologic features, and diagnosis of Burkitt lymphoma", section on 'Other genetic abnormalities'.)

The different clinical subtypes of BL (ie, endemic, nonendemic, and immunodeficiency-related) likely arise from B cells at different stages of development. Specifically, Epstein-Barr virus (EBV)-negative BLs show a low level of somatic hypermutation of their variable region heavy chain genes and no signs of antigen selection, whereas EBV-positive (generally endemic or AIDS-related) BLs have significantly higher levels of somatic hypermutation and evidence of antigen selection [3]. These findings suggest that EBV-negative BL may arise from an early centroblast, while EBV-positive BL may arise from a memory B cell or late germinal center B cell. This difference in cell of origin may also relate to the observed difference in MYC translocation breakpoints described below. Despite these differences, these clinical subtypes may have a common pathogenesis, based on similarities in gene expression [4]. (See 'Chromosomal translocations' below.)

PATHOGENESIS

Overview — The development of BL is dependent on the constitutive expression of the MYC gene located at chromosome 8q24, which encodes the MYC protein transcription factor. MYC modulates the expression of target genes that regulate many cellular processes, including cell growth, division, death, metabolism, adhesion, and motility [5,6]. Cells can generate high levels of MYC protein through various mechanisms, as described in the sections below.

The vast majority of BL have rearrangements of the MYC gene. However, additional factors beyond MYC translocation must occur for the development of BL. In support of this idea, a small percentage of HIV-positive persons and healthy controls have MYC translocations in B lymphocytes isolated from enlarged lymph nodes, a finding that does not appear to predict the later emergence of lymphoma [7]. (See "Epidemiology, clinical manifestations, pathologic features, and diagnosis of Burkitt lymphoma", section on 'Diagnosis'.)

MYC in normal cells — The MYC gene encodes a ubiquitously expressed nuclear phosphoprotein that functions as a pleiotropic transcriptional regulator of cell proliferation, differentiation, and apoptosis [5]. In vivo, MYC is found mainly in heterodimeric complexes with the related protein MAX; these heterodimers bind to the "E box consensus sequence" and directly activate transcription. The MYC-MAX interaction is required for MYC to stimulate transcription and cell proliferation [8-10]. Remarkably, there appear to be thousands of bona fide MYC/MAX binding sites in the genome that permit MYC to regulate the expression of numerous genes, leading to the idea that it may be better viewed as a global regulator of chromatin and gene expression than as a conventional transcription factor [11,12]. As a result, cells that express high levels of MYC produce far more ribonucleic acid (RNA) per cell than those that do not [13]. The net effect of this enhanced gene expression is to support both cell cycle progression and Warburg metabolism, which act together to maximize cell growth and proliferation [14].

MYC activity is normally regulated by the amount of MAX available to form MYC-MAX heterodimers and by competition from complexes formed by MAX with other proteins. MAX can form homodimers (MAX-MAX) and heterodimers with MAD family proteins (ie, MNT, MGA) that are negative regulators of transcription and block MYC-dependent transformation [15,16]. It is postulated that MAD-MAX or MNT-MAX heterodimers inhibit MYC function by either sequestering MAX and preventing the formation of MYC/MAX heterodimers or by competing with MYC/MAX heterodimers for binding to the same target sites. Upregulation of MYC favors MYC-MAX over MNT-MAX complexes at the promoters of target genes, thereby promoting cell cycle progression, while overexpression of MNT inhibits MYC-induced cell cycle entry, consistent with a hypothesis of competitive binding to MAX [17]. In non-Hodgkin lymphomas carrying MYC translocations, it is likely that constitutive expression of MYC increases the ratio of MYC/MAX complexes relative to inhibitory heterodimers, leading to upregulation of a program of gene expression that promotes growth.

The role of MYC in normal germinal center B cells was initially obscure, as most germinal center B cells exhibit low levels of MYC expression. However, conditional Myc knockout mice have established that MYC is essential for the initial proliferative burst that occurs following T cell-dependent antigen-mediated activation of B cells and for the establishment of germinal centers [18,19]. Shortly after entry into germinal centers, antigen-activated B cells downregulate MYC, but a subset of cells re-express MYC, which may contribute to the process of proliferation-associated somatic hypermutation of germinal center B cells.

MYC overexpression — BL is characterized by inappropriately high levels of MYC expression, which may occur through at least one of the following mechanisms [5]:

Chromosomal translocations place MYC under the control of immunoglobulin gene enhancers that are constitutively active in mature B cells.

5' regulatory regions normally flanking the MYC gene body are mutated or removed as a direct result of the translocation.

Mutations of the MYC coding sequence result in amino acid substitutions that stabilize MYC protein by decreasing its proteosome-mediated degradation, thereby increasing its half-life.

Substantial experimental evidence has shown that constitutive expression of MYC influences the growth of B cells in vitro and in vivo, consistent with a role in B cell lymphomagenesis. As examples:

In vitro, the expression of a MYC gene in Epstein-Barr virus (EBV)-immortalized human B cells leads to their malignant transformation [20].

In vivo, transgenic overexpression of MYC in B cell lineage cells in mice leads to the development of B cell malignancy [21].

Gene expression profiling of primary BL has confirmed the overexpression of MYC target genes identified in experimental models [22,23].

Chromosomal translocations — In virtually all cases, BL is associated with a translocation between the long arm of chromosome 8, the site of the MYC gene (8q24), and one of three Ig genes [24]:

The Ig heavy chain gene on chromosome 14 – Resulting in the t(8;14)(q24;q32) found in 80 percent of BL.

The kappa light chain gene on chromosome 2 – Resulting in the t(2;8)(p11;q24) found in 15 percent of BL.

The lambda light chain gene on chromosome 22 – Resulting in the t(8;22)(q24;q11) found in 5 percent of BL.

The common effect of these translocations is that the translocated MYC allele is expressed constitutively in tumor cells, as opposed to the tight regulation of MYC levels in normal B cells [25]. (See 'MYC in normal cells' above.)

Although fairly homogeneous at the microscopic level, these translocations display a high degree of molecular heterogeneity. The specific gene breakpoint sites vary not only by translocation but also by clinical context (ie, endemic versus sporadic cases):

The breakpoint sites on chromosome 8 found in t(8;14) are located 5' and centromeric to MYC, whereas the sites found in t(2;8) and t(8;22) map 3' to MYC [24].

In endemic (African) cases, the breakpoint on chromosome 14 involves the heavy chain joining region and the breakpoint in chromosome 8 usually lies adjacent to MYC, while in non-endemic cases, the breakpoint on chromosome 14 involves the heavy chain class-switch region and the breakpoint in chromosome 8 often lies in intron 1 of MYC [26,27].

This molecular heterogeneity precludes the development of sensitive polymerase chain reaction (PCR) based testing for these translocations. Instead, these translocations are identified either by karyotyping of metaphase chromosomes or by fluorescence in situ hybridization (FISH).

Work using mouse models has shed light on the possible mechanisms that lead to MYC translocations. Translocations involving the class-switch region of the immunoglobulin heavy chain gene and MYC occur with surprisingly high frequency in activated B cells undergoing class-switch recombination. These "mistakes" occur during the recombination event that allows B cells to switch from the expression of IgM to the expression of other immunoglobulin types and require the enzyme activation-induced cytidine deaminase (AID) [28], which is an essential co-factor for normal class-switch recombination. In further support of a role for AID in chromosomal rearrangements that lead to lymphomagenesis, infection of mice with malaria (a risk factor for human endemic BL) provokes sustained expansion of AID-expressing germinal center B cells and increases the development of aggressive B cell lymphomas with chromosomal rearrangements bearing the molecular signatures of AID-mediated deoxyribonucleic acid (DNA) damage [29].

Other work has shown that MYC tends to lie in close proximity to the immunoglobulin heavy chain locus in the nuclei of B cells, which may explain in part why MYC is often involved in such translocations. It is hypothesized that most cells bearing these translocations die rapidly, as unbridled MYC expression is a potent inducer of apoptosis (programmed cell death) in normal cells. How BL cells escape this fate is largely unknown; in some tumors, however, loss of TP53 function may abrogate MYC-induced apoptosis. (See 'Other genetic lesions' below.)

Other mechanisms — As mentioned above, up to 5 percent of tumors with features that are otherwise typical of BL lack MYC rearrangements. Other mechanisms leading to the increased expression of MYC include interference with the 5' regulatory regions and/or stabilization of the protein product.

5' regulatory regions normally present within the MYC DNA sequence can be mutated or moved away from MYC as a direct result of the translocation. Release of MYC from normal regulation can result in increased expression. The MYC first exon/first intron border, where MYC regulatory sequences are located, can be either decapitated by the translocations described in the section above or undergo mutation in translocated alleles [24,30].

Alternatively, the transactivation domain of MYC (encoded in exon 2) can acquire mutations that interfere with modulation of MYC activity by p107 [31,32]. Other mutations may stabilize MYC by decreasing proteasomal degradation.

Consequences of MYC overexpression — BL may be the fastest growing human cancer and MYC overexpression is believed to be responsible for many of the alterations that support the rapid growth of BL tumors cells [33]. Included among these MYC-dependent activities are:

Increased glucose utilization and a switch to so-called Warburg metabolism, in which cells mainly rely on glycolysis for ATP production instead of oxidative phosphorylation.

Increased glutamine metabolism, which creates metabolic intermediates that are needed for membrane biogenesis.

Upregulation of rRNA expression and ribosome biogenesis, leading to increased protein synthesis.

Other genetic lesions — In addition to the presence of MYC translocations in the vast majority of BL cases, most tumors contain additional chromosomal abnormalities, the molecular implications of which are poorly understood [34]. Of interest, BL cases appear to have fewer additional genetic lesions than DLBCL or unclassifiable aggressive B cell lymphomas [35].

Other pathogenic mutations in BL have emerged from genomic sequencing:

A study that reported whole genome sequencing of 101 tumors, which included all three categories of BL (ie, endemic, sporadic, immunodeficiency associated) reported likely driver mutations in 72 genes; these alterations involved both coding and non-coding regions [36]. In vitro assays were used to confirm a functional role for a subset of these genes (eg, MYC, IGLL5, BACH2, ID3, and BTG2).

In one study, high-throughput RNA sequencing and RNA interference screening identified recurrent mutations in components of the p53, cyclin D3/CDK6, PI(3) kinase, and tonic BCR signaling pathways [37]. Approximately 70 percent of sporadic BL cases had mutations in TCF3 or ID3. Mutations in CCND3 (a gene encoding cyclin D3, a key regulator of cell cycle progression) were present in 38 percent.

In another study, whole genome sequencing of BL tumor tissue and germline tissue from the same individual identified mutations of potential interest that were further evaluated with exome sequencing of 59 BL cases and 94 DLBCL cases [38]. Mutations in ID3 were identified in 34 percent of BL but were not present in DLBCL.

In a third study, whole genome and transcriptome sequencing of four BL cases identified seven recurrently mutated genes [39]. Further evaluation of these genes in an extension cohort demonstrated ID3 mutations in 36 of 53 (68 percent) of patients with BL but only 6 of 47 (13 percent) of other B cell lymphomas with Ig-MYC translocation.

The Burkitt Lymphoma Genome Sequencing Project employs whole genome sequencing coupled with transcriptome analyses in an effort to comprehensively characterize the genetics of BL [40]. This ongoing work has identified 22 genes with mutations that are deemed to be significant, as judged by their presence in multiple tumors and their likely disruptive effects on gene function. Newly identified mutated genes include DDX3X (which encodes a putative RNA helicase linked to cell cycle progression) and a number of chromatin factors, including chromatin remodeling factors (ARID1A), chromatin "writers" (KMT2D), and even histones (HIST1H1E). How perturbation of these factors contribute to BL pathobiology remains to be determined.

A common theme emerging from these studies is the frequent occurrence in BL of loss of function mutations in ID3 and gain of function mutations in TCF3, which encodes an E-box type transcription factor previously known as E2A that has important roles in lymphoid development. TCF3 appears to stimulate survival through the BCR/PI3K signaling pathway, which upregulates CCND3 (cyclin D3) [41]. Normally, TCF3 also induces its own inhibitor (ID3), but this negative feedback loop is broken in BL by mutations that activate TCF3 and inactivate ID3. TCF3 appears to augment B cell receptor signaling, thereby enhancing pro-survival signaling mediated through the PI-3K pathway [41].

Other tumors with MYC translocations — Other lymphoid tumors may have MYC translocations similar or identical to those found in BL, including diffuse large B cell lymphoma, multiple myeloma, and a subset of unclassifiable B cell lymphomas with some features of BL. In several studies of the latter tumors, cases with "Burkitt-like" features frequently had dual MYC and B cell leukemia/lymphoma 2 (BCL2) translocations (ie, "double hit" lymphomas) and were associated with an extremely poor clinical outcome [42,43]. Another study found that these "double hit" lymphomas could be distinguished from BL by their expression of BCL-2, expression of IRF4/MUM1, a Ki-67 staining fraction <95 percent, and the absence of Epstein-Barr virus [44]. Many of these cases may represent "transformed" follicular lymphomas, in which the MYC translocation is acquired as a secondary event. (See "Pathobiology of follicular lymphoma", section on 'Translocations involving BCL-2'.)

Other Burkitt-like tumors that lack MYC translocations — The 2016 World Health Organization (WHO) classification of lymphoid neoplasms includes the provisional entity Burkitt-like lymphoma with 11q aberration [45]. These tumors lack MYC translocations, tend to occur in children and young adults, and have a gene expression profile and an immunophenotype that resemble those seen in BL. However, Burkitt-like lymphoma with 11q aberration has lower levels of MYC expression and appears to lack mutations in TCF3 and ID3 [46], indicating that it is genetically distinct from BL. Other tumors resembling BL that lack MYC rearrangements and 11q aberrations are best considered variants of diffuse large B-cell lymphoma or high-grade B-cell lymphoma, not otherwise specified, per the WHO classification [45].

Infections

General — Chronic EBV infection appears to play a role in virtually all cases of endemic (African) BL and a minority of sporadic and immunodeficiency-associated BL. However, data suggest that EBV infection alone cannot account for the development of endemic BL and that the etiology is likely polymicrobial [47].

Patients with some other infections are more likely to have a persistent acute phase of initial EBV infection. This prolonged acute phase with its associated B cell expansion may increase the likelihood of acquisition of translocations involving the MYC locus. Proposed organisms associated with persistent EBV infection and BL development include HIV, malaria, and arboviruses [48]. The specific role of these organisms in the pathogenesis of BL remains to be determined.

Initial epidemiologic data demonstrated a high incidence of both Plasmodium falciparum malaria and endemic BL in equatorial Africa and Papua New Guinea [49,50]. Further evidence came from a study that demonstrated that, when compared with age-, sex-, and location-matched controls, cases of childhood endemic BL were more likely to have serologic evidence of a recent malaria infection (anti-HRP-II antibodies) and less likely to have serologic evidence of a long-term malaria infection (anti-SE36 antibodies) [51].

Plausible explanations directly linking falciparum malaria to BL have emerged from several studies. Isolation of tonsillar germinal center B cells from individuals who are chronically infected with P. falciparum reveal an increase in cells expressing activation-induced cytosine deaminase (AID), an enzyme that may cause DNA damage that can lead to chromosomal translocations, including MYC translocations. Furthermore, a higher incidence of EBV-positivity was observed in germinal center B cells isolated from individuals with chronic P. falciparum infections [52]. Mouse models using recurrent P. chabaudi infection demonstrated expansion of germinal center B cells expressing AID and development of aggressive lymphomas in a p53-deficient background [29].

Epstein-Barr virus (EBV) infection — Monoclonal EBV infection is present in virtually all cases of endemic BL, approximately 30 percent of sporadic BL, and 40 percent of immunodeficiency-associated BL. The pathobiologic role of EBV infection in the development of BL is poorly understood.

One hypothesis is that EBV infection stimulates B cell expansion during which process translocations may occur, leading to the overexpression of MYC. Once BL emerges, the EBV infection is thought to have little effect on tumor maintenance. Of interest, EBV infection in BL displays a particular latent infection phenotype characterized by the lack of expression of the EBV transforming proteins LMP-1 and EBNA-2. (See "Virology of Epstein-Barr virus", section on 'Transformation and latent proteins'.)

Whole-genome analysis of 106 cases of endemic and sporadic BL reported that, compared with EBV-negative tumors, EBV-positive tumors had a striking increase in somatic hypermutation and fewer driver mutations (eg, CCND3 mutations) [53]. The increase in somatic hypermutation in EBV-positive cases is consistent with prior work showing that EBV infection increases the expression of activation-induced cytosine deaminase in B cells [54]. These distinctions may also be related to different cells of origin for EBV-positive and EBV-negative BLs. (See 'Cell of origin' above.)

SUMMARY

Description – Burkitt lymphoma (BL) refers to a group of highly aggressive B cell neoplasms with three distinct clinical forms: endemic, sporadic, and immunodeficiency associated. (See "Epidemiology, clinical manifestations, pathologic features, and diagnosis of Burkitt lymphoma", section on 'Clinical features'.)

Cell of origin – BL is derived from germinal or post-germinal center B cells. (See 'Cell of origin' above.)

Role of MYC overexpression – BL development is dependent on constitutive expression of the MYC transcription factor, which is encoded by the MYC gene, located at chromosome 8q24. (See 'Overview' above.)

Tumor cells maintain high expression of MYC through various mechanisms (see 'MYC overexpression' above):

Chromosomal translocations – BL is generally associated with translocation between the long arm of chromosome 8 (the MYC locus) and one of three immunoglobulin (Ig) genes that place the DNA coding sequences for MYC under the control of Ig gene enhancers that are constitutively active in mature B cells. The translocations include t(8;14), t(2;8), and t(8;22). (See 'Chromosomal translocations' above.)

Other mechanisms – In a small percentage of cases, the MYC DNA sequence can be mutated or moved in ways that increase MYC. Examples include increased MYC gene expression and mutations of the coding sequence that modulate MYC activity by increased half-life of the protein product or modulation of interaction with p107. (See 'Other mechanisms' above.)

Inherent genomic instability of germinal center B cells appears to contribute to MYC rearrangements in BL.

Consequences of increased MYC – MYC modulates expression of target genes that regulate many cellular processes, including cell growth, division, and death. Rapid growth is aided by the alteration of MYC targets, including increased glycolysis for ATP production, instead of oxidative phosphorylation (the Warburg effect); increased glutamine metabolism, which creates metabolic intermediates needed for membrane biogenesis; and increased protein synthesis, in association with upregulation of rRNA expression and ribosome biogenesis. (See 'Consequences of MYC overexpression' above.)

Epstein-Barr virus (EBV) infection – Chronic EBV infection plays a role in virtually all cases of endemic (African) BL and in a minority of sporadic and immunodeficiency-associated BL. (See 'Infections' above.)

  1. Campo E, Jaffe ES, Cook JR, et al. The International Consensus Classification of Mature Lymphoid Neoplasms: a report from the Clinical Advisory Committee. Blood 2022; 140:1229.
  2. Alaggio R, Amador C, Anagnostopoulos I, et al. The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Lymphoid Neoplasms. Leukemia 2022; 36:1720.
  3. Bellan C, Lazzi S, Hummel M, et al. Immunoglobulin gene analysis reveals 2 distinct cells of origin for EBV-positive and EBV-negative Burkitt lymphomas. Blood 2005; 106:1031.
  4. Piccaluga PP, De Falco G, Kustagi M, et al. Gene expression analysis uncovers similarity and differences among Burkitt lymphoma subtypes. Blood 2011; 117:3596.
  5. Hecht JL, Aster JC. Molecular biology of Burkitt's lymphoma. J Clin Oncol 2000; 18:3707.
  6. Klapproth K, Wirth T. Advances in the understanding of MYC-induced lymphomagenesis. Br J Haematol 2010; 149:484.
  7. Müller JR, Janz S, Goedert JJ, et al. Persistence of immunoglobulin heavy chain/c-myc recombination-positive lymphocyte clones in the blood of human immunodeficiency virus-infected homosexual men. Proc Natl Acad Sci U S A 1995; 92:6577.
  8. Blackwood EM, Eisenman RN. Max: a helix-loop-helix zipper protein that forms a sequence-specific DNA-binding complex with Myc. Science 1991; 251:1211.
  9. Blackwood EM, Lüscher B, Eisenman RN. Myc and Max associate in vivo. Genes Dev 1992; 6:71.
  10. Gu W, Cechova K, Tassi V, Dalla-Favera R. Opposite regulation of gene transcription and cell proliferation by c-Myc and Max. Proc Natl Acad Sci U S A 1993; 90:2935.
  11. Eilers M, Eisenman RN. Myc's broad reach. Genes Dev 2008; 22:2755.
  12. Nie Z, Hu G, Wei G, et al. c-Myc is a universal amplifier of expressed genes in lymphocytes and embryonic stem cells. Cell 2012; 151:68.
  13. Lovén J, Orlando DA, Sigova AA, et al. Revisiting global gene expression analysis. Cell 2012; 151:476.
  14. Ward PS, Thompson CB. Metabolic reprogramming: a cancer hallmark even warburg did not anticipate. Cancer Cell 2012; 21:297.
  15. Ayer DE, Kretzner L, Eisenman RN. Mad: a heterodimeric partner for Max that antagonizes Myc transcriptional activity. Cell 1993; 72:211.
  16. Zervos AS, Gyuris J, Brent R. Mxi1, a protein that specifically interacts with Max to bind Myc-Max recognition sites. Cell 1993; 72:223.
  17. Walker W, Zhou ZQ, Ota S, et al. Mnt-Max to Myc-Max complex switching regulates cell cycle entry. J Cell Biol 2005; 169:405.
  18. Calado DP, Sasaki Y, Godinho SA, et al. The cell-cycle regulator c-Myc is essential for the formation and maintenance of germinal centers. Nat Immunol 2012; 13:1092.
  19. Stamm O, Latscha U, Janecek P, Campana A. Development of a special electrode for continuous subcutaneous pH measurement in the infant scalp. Am J Obstet Gynecol 1976; 124:193.
  20. Lombardi L, Newcomb EW, Dalla-Favera R. Pathogenesis of Burkitt lymphoma: expression of an activated c-myc oncogene causes the tumorigenic conversion of EBV-infected human B lymphoblasts. Cell 1987; 49:161.
  21. Adams JM, Harris AW, Pinkert CA, et al. The c-myc oncogene driven by immunoglobulin enhancers induces lymphoid malignancy in transgenic mice. Nature 1985; 318:533.
  22. Dave SS, Fu K, Wright GW, et al. Molecular diagnosis of Burkitt's lymphoma. N Engl J Med 2006; 354:2431.
  23. Hummel M, Bentink S, Berger H, et al. A biologic definition of Burkitt's lymphoma from transcriptional and genomic profiling. N Engl J Med 2006; 354:2419.
  24. Dalla-Favera R, Bregni M, Erikson J, et al. Human c-myc onc gene is located on the region of chromosome 8 that is translocated in Burkitt lymphoma cells. Proc Natl Acad Sci U S A 1982; 79:7824.
  25. ar-Rushdi A, Nishikura K, Erikson J, et al. Differential expression of the translocated and the untranslocated c-myc oncogene in Burkitt lymphoma. Science 1983; 222:390.
  26. Pelicci PG, Knowles DM 2nd, Magrath I, Dalla-Favera R. Chromosomal breakpoints and structural alterations of the c-myc locus differ in endemic and sporadic forms of Burkitt lymphoma. Proc Natl Acad Sci U S A 1986; 83:2984.
  27. Neri A, Barriga F, Knowles DM, et al. Different regions of the immunoglobulin heavy-chain locus are involved in chromosomal translocations in distinct pathogenetic forms of Burkitt lymphoma. Proc Natl Acad Sci U S A 1988; 85:2748.
  28. Ramiro AR, Jankovic M, Callen E, et al. Role of genomic instability and p53 in AID-induced c-myc-Igh translocations. Nature 2006; 440:105.
  29. Robbiani DF, Deroubaix S, Feldhahn N, et al. Plasmodium Infection Promotes Genomic Instability and AID-Dependent B Cell Lymphoma. Cell 2015; 162:727.
  30. Cesarman E, Dalla-Favera R, Bentley D, Groudine M. Mutations in the first exon are associated with altered transcription of c-myc in Burkitt lymphoma. Science 1987; 238:1272.
  31. Bhatia K, Huppi K, Spangler G, et al. Point mutations in the c-Myc transactivation domain are common in Burkitt's lymphoma and mouse plasmacytomas. Nat Genet 1993; 5:56.
  32. Gu W, Bhatia K, Magrath IT, et al. Binding and suppression of the Myc transcriptional activation domain by p107. Science 1994; 264:251.
  33. Dang CV. MYC on the path to cancer. Cell 2012; 149:22.
  34. Toujani S, Dessen P, Ithzar N, et al. High resolution genome-wide analysis of chromosomal alterations in Burkitt's lymphoma. PLoS One 2009; 4:e7089.
  35. Seegmiller AC, Garcia R, Huang R, et al. Simple karyotype and bcl-6 expression predict a diagnosis of Burkitt lymphoma and better survival in IG-MYC rearranged high-grade B-cell lymphomas. Mod Pathol 2010; 23:909.
  36. Panea RI, Love CL, Shingleton JR, et al. The whole-genome landscape of Burkitt lymphoma subtypes. Blood 2019; 134:1598.
  37. Schmitz R, Young RM, Ceribelli M, et al. Burkitt lymphoma pathogenesis and therapeutic targets from structural and functional genomics. Nature 2012; 490:116.
  38. Love C, Sun Z, Jima D, et al. The genetic landscape of mutations in Burkitt lymphoma. Nat Genet 2012; 44:1321.
  39. Richter J, Schlesner M, Hoffmann S, et al. Recurrent mutation of the ID3 gene in Burkitt lymphoma identified by integrated genome, exome and transcriptome sequencing. Nat Genet 2012; 44:1316.
  40. Grande BM, Gerhard DS, Griner NB, et al. Burkitt Lymphoma Genome Sequencing Project (BLGSP): Integrative Genomic and Transcriptomic Characterization of Burkitt Lymphoma. Blood (ASH Annual Meeting Abstracts) 2017; 130:39.
  41. Sander S, Calado DP, Srinivasan L, et al. Synergy between PI3K signaling and MYC in Burkitt lymphomagenesis. Cancer Cell 2012; 22:167.
  42. Macpherson N, Lesack D, Klasa R, et al. Small noncleaved, non-Burkitt's (Burkit-Like) lymphoma: cytogenetics predict outcome and reflect clinical presentation. J Clin Oncol 1999; 17:1558.
  43. Johnson NA, Savage KJ, Ludkovski O, et al. Lymphomas with concurrent BCL2 and MYC translocations: the critical factors associated with survival. Blood 2009; 114:2273.
  44. Snuderl M, Kolman OK, Chen YB, et al. B-cell lymphomas with concurrent IGH-BCL2 and MYC rearrangements are aggressive neoplasms with clinical and pathologic features distinct from Burkitt lymphoma and diffuse large B-cell lymphoma. Am J Surg Pathol 2010; 34:327.
  45. World health organization classification of tumours of haematopoietic and lymphoid tissues, revised 4th edition, Swerdlow SH, Campo E, Harris NL, et al. (Eds), IARC, Lyon 2017.
  46. Gonzalez-Farre B, Ramis-Zaldivar JE, Salmeron-Villalobos J, et al. Burkitt-like lymphoma with 11q aberration: a germinal center-derived lymphoma genetically unrelated to Burkitt lymphoma. Haematologica 2019; 104:1822.
  47. Chene A, Donati D, Orem J, et al. Endemic Burkitt's lymphoma as a polymicrobial disease: new insights on the interaction between Plasmodium falciparum and Epstein-Barr virus. Semin Cancer Biol 2009; 19:411.
  48. World Health Organization Classification of Tumours of Haematopoietic and Lymphoid Tissues, Swerdlow SH, Campo E, Harris NL, et al. (Eds), IARC Press, Lyon 2008.
  49. Parkin DM, Sitas F, Chirenje M, et al. Part I: Cancer in Indigenous Africans--burden, distribution, and trends. Lancet Oncol 2008; 9:683.
  50. Morrow RH Jr. Epidemiological evidence for the role of falciparum malaria in the pathogenesis of Burkitt's lymphoma. IARC Sci Publ 1985; :177.
  51. Aka P, Vila MC, Jariwala A, et al. Endemic Burkitt lymphoma is associated with strength and diversity of Plasmodium falciparum malaria stage-specific antigen antibody response. Blood 2013; 122:629.
  52. Torgbor C, Awuah P, Deitsch K, et al. A multifactorial role for P. falciparum malaria in endemic Burkitt's lymphoma pathogenesis. PLoS Pathog 2014; 10:e1004170.
  53. Grande BM, Gerhard DS, Jiang A, et al. Genome-wide discovery of somatic coding and noncoding mutations in pediatric endemic and sporadic Burkitt lymphoma. Blood 2019; 133:1313.
  54. Kalchschmidt JS, Bashford-Rogers R, Paschos K, et al. Epstein-Barr virus nuclear protein EBNA3C directly induces expression of AID and somatic mutations in B cells. J Exp Med 2016; 213:921.
Topic 4723 Version 20.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟