Abstract

Simple SummaryThe pathogenetic mechanisms and peculiar tropism of primary CNS lymphoma (PCNSL) of the central nervous system (CNS) have been the subject of debate for decades. Hypothesis-driven targeted molecular studies have revealed that PCNSLs derived from self-/polyreactive B cells that have escaped developmental control mechanisms. The early acquisition of activating mutations targeting the B cell receptor pathway provides a survival advantage. The failure of the germinal center (GC) reaction and its checkpoints increases tumor B cell affinity for the CNS. During this faulty GC reaction, PCNSL tumor cells acquire further oncogenic alterations converging on the Toll-like receptor, B cell receptor, and NF-κB pathway. These activated pathways sustain proliferation. Concomitantly, cells become unable to complete terminal B cell differentiation, becoming trapped within the vicious cycle of the GC reaction as low-affinity IgM+ B cells related to memory cells.Primary lymphoma of the central nervous system (PCNSL, CNS) is a specific diffuse large B cell lymphoma (DLBCL) entity confined to the CNS. Key to its pathogenesis is a failure of B cell differentiation and a lack of appropriate control at differentiation stages before entrance and within the germinal center (GC). Self-/polyreactive B cells rescued from apoptosis by MYD88 and/or CD79B mutations accumulate a high load of somatic mutations in their rearranged immunoglobulin (IG) genes, with ongoing somatic hypermutation (SHM). Furthermore, the targeting of oncogenes by aberrant SHM (e.g., PIM1, PAX5, RHOH, MYC, BTG2, KLHL14, SUSD2), translocations of the IG and BCL6 genes, and genomic instability (e.g., gains of 18q21; losses of 9p21, 8q12, 6q21) occur in these cells in the course of their malignant transformation. Activated Toll-like receptor, B cell receptor (BCR), and NF-κB signaling pathways foster lymphoma cell proliferation. Hence, tumor cells are arrested in a late B cell differentiation stage, corresponding to late GC exit B cells, which are genetically related to IgM+ memory cells. Paradoxically, the GC reaction increases self-/polyreactivity, yielding increased tumor BCR reactivity for multiple CNS proteins, which likely contributes to CNS tropism of the lymphoma. The loss of MHC class I antigen expression supports tumor cell immune escape. Thus, specific and unique interactions of the tumor cells with resident CNS cells determine the hallmarks of PCNSL.

Highlights

  • (1) What is the cellular origin of these tumor cells? (2) What are the pathogenetically relevant genetic, epigenetic, transcriptional, and proteomic alterations of these tumor cells? (3) What are the reasons for the tropism and exclusive manifestation of this tumor in the central nervous system (CNS)?

  • We propose that the term “primary CNS lymphoma (PCNSL)” should be reserved for the most common molecular subtype of PCNS-diffuse large B cell lymphoma (DLBCL), namely, PCNS-DLBCL with MYD88 and B cell receptor (BCR) signaling pathway mutations, which is nowadays described as the MCD/C5/MYD88 molecular group

  • PCNSLs are characterized by their unique geno- and phenotype among DLBCLs, which results from the failure of B cell differentiation and the lack of appropriate control of differentiation steps

Read more

Summary

Historic Background

The exclusive manifestation of malignant lymphoma in the central nervous system (CNS) has remained mysterious and has been frequently debated. The term “PCNSL” has been modified to “PCNS-DLBCL” to precisely define this entity; this terminology appreciates the specific nature of the target organ, i.e., the immunoprivileged CNS, and delineates PCNS-DLBCL from other lymphoma entities that may be present in the CNS The latter mainly include low-grade B cell lymphomas, mucosa-associated lymphatic tissue (MALT) lymphoma of the dura, and various immunodeficiency-related hematopoietic lesions, all of which may manifest in the CNS either exclusively or as part of a generalized disease. Current diagnostic work-up includes (1) the phenotypic identification of the tumor cells of PCNSL as late germinal center (GC) exit B cells and (2) careful clinical staging to exclude the possibility of extracerebral DLBCL In unequivocal cases, this algorithm correctly identifies PCNSL corresponding to the MCD/C5/MYD88 DLBCL subgroup when investigated by sequencing technologies

Molecular Pathogenesis of PCNSL
A Faulty GC Reaction as Key Process for PCNSL Pathogenesis
Tumor Cell Adaptation to the CNS Microenvironment in PCNSL
Role of Preclinical Animal Models in Studying PCNSL Pathogenesis
Findings
Conclusions and Perspectives
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call