Abstract
More than 50 subtypes of B-cell non-Hodgkin lymphoma (B-NHL) are recognized in the most recent World Health Organization classification of 2016. The current treatment paradigm, however, is largely based on ‘one-size-fits-all’ immune-chemotherapy. Unfortunately, this therapeutic strategy is inadequate for a significant number of patients. As such, there is an indisputable need for novel, preferably targeted, therapies based on a biologically driven classification and risk stratification. Sequencing studies identified mutations in the MYD88 gene as an important oncogenic driver in B-cell lymphomas. MYD88 mutations constitutively activate NF-κB and its associated signaling pathways, thereby promoting B-cell proliferation and survival. High frequencies of the hotspot MYD88(L265P) mutation are observed in extranodal diffuse large B-cell lymphoma and Waldenström macroglobulinemia, thereby demonstrating this mutation’s potential as a disease marker. In addition, the presence of mutant MYD88 predicts survival outcome in B-NHL subtypes and it provides a therapeutic target. Early clinical trials targeting MYD88 have shown encouraging results in relapsed/refractory B-NHL. Patients with these disorders can benefit from analysis for the MYD88 hotspot mutation in liquid biopsies, as a minimally invasive method to demonstrate treatment response or resistance. Given these clear clinical implications and the crucial role of MYD88 in lymphomagenesis, we expect that analysis of this gene will increasingly be used in routine clinical practice, not only as a diagnostic classifier, but also as a prognostic and therapeutic biomarker directing precision medicine. This review focuses on the pivotal mechanistic role of mutated MYD88 and its clinical implications in B-NHL.Introduction
With the introduction of high-throughput, next-generation sequencing, many studies have aimed to explain the diverse biology, clinical course, prognosis, and therapeutic response of B-cell non-Hodgkin lymphoma (B-NHL). This has increased our knowledge of lymphomagenesis by identifying many novel somatic alterations that affect signaling pathways involved in several B-NHL subtypes. In this rapidly evolving molecular landscape, it is important to translate newly obtained genetic knowledge directly into clinical benefit for patients.1
Ngo et al. were the first to identify an oncogenic, non-synonymous, gain-of-function mutation in myeloid differentiation primary response 88 (MYD88), leading to an amino-acid change of leucine to proline at position 265 (NM_002468.5, also referred to as position 273 in NM_001172567) of MYD88 [MYD88(L265P)].2 Other recurrent mutations in MYD88 were likewise identified; however, the impact of these mutations has been difficult to establish due to their low prevalence.3 This review, therefore, focuses on the present understanding of the role of MYD88(L265P) in NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) activation and its association with the B-cell receptor (BCR) cascade. In addition, we address the clinical importance of MYD88(L265P), including its prevalence across B-NHL subtypes, its predictive significance in patients’ outcome, and its potential as a therapeutic target.
Oncogenic mechanisms of MYD88(L265P)
Canonical NF-κB signaling
In normal physiology, MYD88 acts as a signaling adaptor in the canonical NF-κB pathway (Figure 1). This pathway is activated upon recognition of pathogen-associated molecular patterns (PAMP) by receptors containing a toll/interleukin-1 receptor (TIR) domain, such as toll-like receptors (TLR) and the interleukin receptors 1 (IL-1R) and 18 (IL-18R). After ligand binding, the TIR domain of these receptors interacts with the TIR domain of MYD884 and this process initiates the formation of the so-called ‘myddosome complex’. For this complex, activated MYD88 recruits IL-1R associated kinase 4 (IRAK4), a serine-threonine kinase, and together they phosphorylate IRAK1 or IRAK2. Phosphorylated IRAK1 and IRAK2 interact with tumor necrosis factor receptor-associated factor 6 (TRAF6), resulting in activation of transforming growth factor beta-activated kinase 1 (TAK1).5 Activated TAK1 continues signaling through the mitogen-activated protein kinase (MAPK) signaling cascade and cooperates with TAK1-binding protein (TAB) to activate the inhibitor of the NF-κB kinase (IKK) complex.
The IKK complex consists of the kinase subunits IKKa and IKKβ and the regulatory subunit NF-κB essential modulator. After activation, this complex phosphorylates the inhibitor of NF-κB (IκB) proteins that are bound to NF-κB, which prevent migration of NF-κB to the nucleus. Phosphorylation of these IκB proteins results in ubiquitylation and proteasomal degradation of IκB and release of the NF-κB subunits. Subsequently, the NF-κB subunits, including RELA (p65)-p50 in the classical pathway and RELB-p52 in the alternative pathway, migrate to the nucleus where they bind to specific DNA-binding sites and induce increased expression of genes involved in B-cell proliferation and survival. In addition, expression of these genes is increased through interactions between the NF-κB subunits and other transcription factors, such as E1A binding protein P300 (EP300) and CREB binding protein (CREBBP).6
In the case of MYD88(L265P), the TIR domain of MYD88, in which L265P resides, is more highly activated compared with wildtype MYD88 and this increases downstream signaling and formation of the myddosome complex.2 Henceforth, MYD88(L265P) preferentially and constitutively recruits IRAK1 for the myddosome and, together with IRAK4, was found to be essential for survival of activated B-cell (ABC) diffuse large B-cell lymphoma (DLBCL) cell lines with MYD88(L265P).872 In addition, IRAK1 was shown to be co-immunoprecipitated with MYD88 in chronic lymphocytic leukemia (CLL) cells with MYD88(L265P) and stimulation of IL-1R and TLR induced a 5-fold to 150-fold increase of cytokine secretion compared to that of CLL cells with wildtype MYD88.9 However, Ansell et al.7 identified that in Waldenström macroglobulinemia (WM) cell lines, the myddosome complex consisted of IRAK4, TRAF6, and MYD88, but not IRAK1. The authors hypothesized that this difference in complex formation was instigated by the heterozygous nature of MYD88(L265P) in WM and the homozygous nature in DLBCL, which was strengthened by the finding that downstream signaling of TAK1 phosphorylation was highest in the DLBCL cell line with homozygous MYD88(L265P).7 Furthermore, the stabilizing effect of heat shock protein 110 (HSP110) on the myddosome complex, due to interference with the proteasomal degradation of MYD88, is stronger in ABC-DLBCL cell lines with MYD88(L265P) than in those with wild-type MYD88.10 As MYD88(L265P) constitutively activates the NF-κB pathway, it is regarded as an important oncogenic driver in B-NHL.1272
B-cell receptor signaling
In addition to the canonical NF-κB pathway, the BCR pathway plays an important role in B-cell survival and proliferation and oncogenesis of B-NHL with MYD88 mutations (Figure 1). In normal physiology, stimulation of the BCR activates NF-κB, as well as the phosphoinositide 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR), and nuclear factor of activated T cells (NFAT) pathways. After antigen recognition by the BCR, Lck/Yes-related novel protein tyrosine kinase (LYN) is released from its inactive state through dephosphorylation of the C-terminal regulatory tyrosine by cluster of differentiation 45 (CD45) or an exogenous ligand for the Src-homology 2 (SH2) and SH3 domains of LYN, such as CD19. Activated LYN consecutively phosphorylates the immunoreceptor tyrosine-based activation motif (ITAM) domains of the coupled CD79A and CD79B heterodimers. These double-phosphorylated ITAM domains provide a docking site for the SH2 domains of spleen tyrosine kinase (SYK), which is activated by autophosphorylation or through transphosphorylation by LYN. LYN and SYK then activate Bruton tyrosine kinase (BTK) by phosphorylation, which is recruited to the membrane through interaction between the pleckstrin homology (PH) domain of BTK and phosphatidylinositol-3, 4, 5-triphosphate (PIP3) of the PI3K pathway or through interaction between the SH2 domain of BTK with the B-cell linker protein (BLNK) adapter molecule that also recruits phospholipase Cg2 (PLCg2) to the membrane.13 BTK activates PLCg2, initiating activation of the NF-κB pathway through formation of CBM complex, consisting of caspase recruitment domain family member 11 (CARD11), BCL10, and mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1). In addition, BTK activates the MAPK and PI3K pathways14 and PLCg2 triggers the NFAT pathway through calcineurin. The CBM complex subsequently attracts TRAF6, TAK1, and TAB, and promotes the degradation of IκB, which leads to the release of NF-κB subunits.151454
BTK is an integral protein in the BCR signaling cascade and has been found to be preferentially complexed to MYD88 in WM cells with MYD88(L265P) and not in MYD88 wildtype cells. Inhibition of BTK resulted in a decrease of the formation of this MYD88-BTK complex, but lacked effect on IRAK4/IRAK1 activity and vice versa, indicating a potential necessity of dual inhibition of IRAK and BTK for WM with MYD88(L265P).1816 MYD88 is frequently mutated in patients who also harbor a mutation in the 196 tyrosine residue in the ITAM domain of CD79B (NM_000626) and these patients seem to benefit most from BTK-inhibition treatment.19 The exact consequence of these double mutations in B-NHL is unclear, but Phelan et al.8 recently provided new insight into the mechanism of combined MYD88 and BCR-pathway activation as they identified a MYD88-TLR9-BCR (My-T-BCR) supercomplex. This supercomplex is generated by constitutive trafficking of the BCR towards endolysosomes that contain TLR9 and interacts with mTOR and the CBM complex, thereby promoting lymphomagenesis by activation of the mTOR and NF-κB pathways. Its presence was demonstrated in cell lines and biopsies of ABC-DLBCL, primary DLBCL of the central nervous system, and lymphoplasmacytic lymphoma and correlated with responsiveness to BTK inhibition. On the other hand, the supercomplex was not identified in CLL or mantle cell lymphoma, suggesting a different mechanism of BCR signaling in these entities. Therefore, the My-T-BCR supercomplex could potentially be used as a biomarker for predicting the efficacy of BTK inhibitors, as a classifier of B-NHL subtypes, or as a novel therapeutic target via inhibition of TLR9.8
Autocrine signaling
As described, increased formation of the myddosome complex with IRAK1, as well as activation of the BCR pathway, caused by interactions of BTK with MYD88(L265P), CD79B mutations, and the My-T-BCR supercomplex, result in constitutive activation of the NF-κB pathway. NF-κB not only activates the transcription of genes involved in cell survival and proliferation, but also results in autocrine signaling with IL-6 and IL-10. One consequence of this autocrine signaling loop is the phosphorylation of Janus kinase 1 (JAK1) and, subsequently, signal transducer and activator of transcription 3 (STAT3) with the assembly of a STAT3/STAT3 complex. This complex increases transcription of genes involved in several signaling cascades, including the PI3K/AKT/mTOR, E2F/G2M cell-cycle checkpoint, JAK/STAT, and NF-κB pathways. In addition, STAT3 activity represses the proapoptotic type I interferon (IFN) signaling pathway by downregulating IFN-regulatory factor 7 (IRF7), IRF9, STAT1, and STAT2 expression.2032
Another consequence of IL-6 signaling is the aberrant expression of hematopoietic cell kinase (HCK), as identified in primary WM cells and B-NHL cell lines.21 Increased levels of HCK promote lymphomagenesis, as HCK knockdown in B-NHL cell lines reduces survival and lowers the activity of the BCR, PI3K/AKT, and MAPK/ERK (extracellular signal-regulated kinases) pathways. Furthermore, BTK- and HCK-inhibition treatment of ABC-DLBCL and WM cells with MYD88(L265P) decreased HCK expression, whereas mutant HCK(T333M) (NM_002110.4) attenuated this effect. These findings suggest that HCK is downstream of MYD88(L265P) and that HCK should be regarded as a potential therapeutic target in B-NHL with MYD88(L265P).
Prevalence
The described oncogenic mechanisms largely depend on the prevalence of MYD88(L265P) in B-NHL. Several studies, using Sanger sequencing, allele-specific polymerase chain reaction (PCR) analysis, or (targeted) next-generation sequencing, have demonstrated that the occurrence of MYD88(L265P) varies highly among the different subtypes of B-NHL (Table 1).108221832 The highest prevalence of MYD88(L265P) is found in lymphoplasmacytic lymphoma/WM, with approximately 85% of the patients being affected.372218 In DLBCL, the prevalence of MYD88(L265P) is highest (range, 44% to 73%) in extran-odal DLBCL, in immune-privileged sites,96 such as primary DLBCL of the central nervous system968886232218 and primary testicular lymphoma,108962322 primary cutaneous DLBCL, leg type,91897122 orbital/vitreoretinal DLBCL,989722 intravascular large B-cell lymphoma,95 and primary breast DLBCL.9922 The high prevalence of MYD88(L265P) in extranodal site-specific lymphomas, lymphoplasmacytic lymphoma, and WM may provide an indication for the origin of these lymphomas. Interestingly, B-NHL entities with a high prevalence of MYD88(L265P) are characterized by a monoclonal immunoglobulin M. Furthermore, the high occurrence of MYD88(L265P) in extranodal DLBCL may imply that B cells need to gain this mutation for survival and manifestation in extranodal sites.
In DLBCL in general, a recent meta-analysis by Lee et al.,22 comprising 18 studies with a total of 2002 DLBCL patients, documented that 255 of 1236 (21%) cases of ABC-DLBCL harbored MYD88(L265P), compared with 44 of 766 (6%) cases of germinal center B-cell-like (GCB) DLBCL. Large sequencing studies, such as those by Reddy et al.,80 Schmitz et al.,81 Chapuy et al.,77 and Intlekofer et al.,79 have compared with 44 of 766 (6%) cases of GCB DLBCL with archaic cell-of-origin classification, based on immunohistochemistry or gene expression profiling, and have shown that MYD88(L265P) and other mutations transcend these classifications and should be put into context with emerging genomic classification systems. These large sequencing studies underscore the need to evaluate the status of not only MYD88, but also other genes involved in B-cell lymphomagenesis for diagnosis and during treatment with targeted therapies, as proposed by Sujobert et al.109
Overall, these results identify MYD88(L265P) as a diagnostic classifier for specific B-NHL subtypes. This is supported by a recent study by our group that identified MYD88 mutations as an independent marker, in a cohort of 250 patients with DLBCL, in addition to the routinely used MYC and BCL2 and/or BCL6 rearrangements and Epstein-Barr virus status (according to the 2016 World Health Organization classification110).83 Furthermore, MYD88(L265P) is absent in primary mediastinal large B-cell lymphoma9432 and primary cutaneous follicle center lymphoma,7371 and rarely present in hairy cell leukemia (1.1%),59573022 plasma cell myeloma (1.5%),10710643232218 Burkitt lymphoma (1.5%),742 follicular lymphoma (1.9%),6867232218 and CLL (2.5%).523828242218
Prognostic impact
In addition to its role as a diagnostic classifier, the prognostic value of MYD88(L265P) has been a topic of many studies involving B-NHL patients. Lee et al. performed a meta-analysis of three studies with accurate multivariate hazard ratios to investigate the prognostic value of MYD88(L265P) in DLBCL.22 This analysis, involving a total of 275 DLBCL patients, showed that DLBCL patients with MYD88(L265P) had a statistically significant inferior overall survival compared with DLBCL patients with wildtype MYD88. In addition, MYD88(L265P) was significantly associated with older age, high International Prognostic Index (IPI)-score risk groups, and extranodal localization. We also demonstrated this association of MYD88(L265P) with an inferior survival in our recent study in which we evaluated MYD88 status, together with CD79B, MYC, BCL2, BCL6 and Epstein-Barr virus status and clinical characteristics in 250 DLBCL patients.83 Additionally, we showed that the performance of the IPI score is improved by adding MYD88(L265P) as a poor risk factor.
The correlation of MYD88 mutations with an inferior overall survival is also observed in several subtypes of extranodal DLBCL, such as primary cutaneous DLBCL, leg type111 and immune-privileged DLBCL.1128322 On the other hand, in a study by Xu et al.,84 MYD88 mutations were significantly more frequent in DLBCL patients who were refractory to chemotherapy with R-CHOP (rituximab, cyclophosphamide, doxorubicin, vincristine and prednisone) (28%) compared with DLBCL patients who were chemosensitive (15%), but no statistically significant correlation with overall survival was found. The actual prognostic value of MYD88 in DLBCL requires further investigation, as other studies identified no effect of MYD88(L265P) on the survival of DLBCL patients.11411222
In other subtypes of B-NHL, such as CLL, splenic marginal zone lymphoma, and WM, MYD88(L265P9 is associated with a superior survival compared with wildtype MYD88.11611545 In WM, approximately 30-40% of patients present with concomitantly mutated MYD88 and CXCR4, a gene involved in homing of B cells in the bone marrow, and these patients present with a greater disease burden and reduced progression-free and overall survival.118117 With regards to CLL, Improgo et al.39 stated that MYD88(L265P) occurs mainly in patients with mutated IGHV or chromosome 13q deletions and both alterations are associated with a superior survival. Furthermore, WM patients with wildtype MYD88 had an increased risk of disease transformation, ibrutinib resistance and shorter overall survival.1181179
Targeted therapies
The oncogenic activity of MYD88(L265P), as well as its high frequency in several B-NHL subtypes, ensure that MYD88 and its affiliated signaling pathways are very interesting for targeted therapeutic strategies. As reviewed by Yu et al.18 and Weber et al.,119 several targets are conceivable for direct or indirect inhibition, such as IRAK1 and IRAK4 in the myddosome-complex, TAK1 in downstream signaling, BTK in the BCR pathway, TLR9 in the My-T-BCR supercomplex, and components of the concurrently activated PI3K/AKT/mTOR and HCK pathways (Figure 2).
Of these targets, inhibition of BTK has been the most extensively studied, regardless of the fact that BTK is not a MYD88(L265P)-specific target and is not directly involved with the myddosome complex. The BTK inhibitor ibrutinib is approved as treatment for CLL, mantle cell lymphoma, relapsed/refractory marginal zone lymphoma, and WM by the United States Food and Drug Administration (FDA). Additionally, the FDA permitted the combined use of ibrutinib and rituximab as the first non-chemotherapeutic regimen for WM patients. In early clinical trials in patients with relapsed/refractory DLBCL and primary DLBCL of the central nervous system, ibrutinib elicited an overall response rate of 80-85% in those with MYD88(L265P) alone or in combination with mutated CD79B.12019 Furthermore, in a randomized phase III trial, ibrutinib plus R-CHOP improved the overall survival of DLBCL patients younger than 60 years regardless of the cell-of-origin.121 Nonetheless, ibrutinib tends to produce many off-target effects and acquisition of resistance to the drug is common. For instance, ibrutinib resistance can be caused by the C481S mutation in BTK (NM_000061), which hampers the interaction between ibrutinib and BTK,122 but also by mutations in PLCg2,123 CARD11,120 and CXCR4.124 Given these drawbacks of ibrutinib, next-generation BTK inhibitors, such as acalabrutinib and zanubrutinib, are being developed and used for research. Studies demonstrated that acalabrutinib achieved an overall response rate of 95% in relapsed CLL125 and 81% in relapsed mantle cell lymphoma,126 and this medicine is now approved as treatment for mantle cell lymphoma by the United States FDA. Zanubrutinib achieved an overall response rate of 90% in WM, and was also shown to be well tolerated and to overcome the ibrutinib resistance mechanism induced by CXCR4 mutations.127
In addition to studies on BTK inhibition, several phase I/II clinical trials have investigated the response of novel therapeutic targets (in)directly involved with MYD88 in patients with B-NHL. In relapsed/refractory WM, mTOR inhibition with everolimus produced an overall response rate of 50%.128 In several subtypes of relapsed/refractory B-NHL, PI3K inhibition with parsaclisib produced overall response rates ranging between 20% and 78%, with a low risk of adverse events and improved long-term outcomes.129 In in vitro assays, enzastaurin, a protein kinase C inhibitor, reduced proliferation and viability of DLBCL cells by regulation of the PI3K, MAPK, and JAK/STAT pathways; however, it also increased phosphorylation of BTK, suggesting the need for simultaneous treatment of enzastaurin with BTK inhibition.130 Patients with DLBCL are currently being recruited into a phase III clinical trial in which enzastaurin is combined with R-CHOP (NCT03263026).
The clinical trials mentioned above focus on therapeutic targets that are directly or indirectly involved with MYD88 activity; however these targets are not specific for MYD88(L265P) and patients are selected irrespective of the mutational status of MYD88. The lack of biomarkers in these clinical trials is a potential weakness, especially regarding the evolving field of genetics and precision medicine. Novel drugs targeting the oncogenic mechanisms of MYD88(L265P), such as inhibition of the interaction between TLR9 and MYD88 in the My-T-BCR supercomplex8 and between MYD88 and IRAK4 in the myddo-some,131 or direct inhibition of IRAK43911 or TAK1,7 would be interesting for B-NHL patients with MYD88(L265P) and have shown promising results in in vitro and in vivo studies. In addition, the use of immunomodulatory oligonucleotides (IMO) such as IMO 8400, an antagonist of TLR7, TLR8, and TLR9, could be an interesting targeted treatment for MYD88(L265P) B-NHL and especially for ABC-DLBCL with the My-T-BCR supercomplex. IMO-8400 has mainly been investigated in immune-mediated inflammatory diseases and only two phase I/II clinical trials with MYD88(L265P)-positive DLBCL and WM have been performed, showing that IMO-8400 is well tolerated in these patients (NCT02252146, NCT02363439, https://www.ider-apharma.com/wp-content/uploads/2015/12/IMO-8400-WM-ASH-Poster.pdf). More research is required on the MYD88(L265P)-specificity of the above-mentioned targets in order to determine their role in the treatment of B-NHL patients with MYD88(L265P) and, thereby, improve personalized medicine.
An alternative therapeutic approach for these patients, as reviewed by Weber et al.,119 is the induction of a T-cell mediated immune response towards tumor-specific neoepitopes that are derived from MYD88(L265P). In in vitro experiments, such neoepitopes, presented by major histocompatibility class I molecules, prompted a cytotoxic CD8 T-cell response.132 These tumor-specific T cells can be harvested from healthy donors133 or patients with B-NHL and primed to elicit a tumor-specific cytotoxic effect or theoretically used as a model for chimeric antigen receptor (CAR) T-cell therapy. Furthermore, in vitro assays of DLBCL showed that MYD88(L265P) tumor cells develop resistance against T-cell mediated cytotoxicity via upregulation of IL-10 and STAT3 and that inhibition of either IL-10 or STAT3 significantly attenuates this gain of resistance.134 To our knowledge, currently no clinical trials are underway to investigate this intriguing treatment concept.
Liquid biopsy
Until now, comprehensive genomic analysis for accurate diagnosis and classification of B-NHL has been based on DNA isolated from lymphoma tissues. For most patients, the collection of this tissue is a highly invasive procedure with the risk of severe complications.135 An alternative and less invasive method of sampling is the so-called ‘liquid biopsy’, using blood plasma or cerebrospinal fluid, instead of lymphoma tissue. These fluids contain circulating tumor DNA (ctDNA) that is secreted or released during apoptosis or necrosis of the tumor cells, and may harbor somatic mutations, such as MYD88(L265P). Besides being a less invasive method of sampling, ctDNA allows detection of spatial differences between lymphoma cells spread throughout the body, which is not possible with tissue biopsies.
The high frequency of MYD88(L265P) in several B-NHL subtypes make this mutation perfectly appropriate for screening by ctDNA, as already demonstrated in DLBCL,136 primary DLBCL of the central nervous system,137 and intravascular large B-cell lymphoma.95 With the highly sensitive and specific method of digital droplet PCR (ddPCR), even low amounts of ctDNA can be detected, potentially providing information about minimal residual disease, clonal evolution over time, and spatial differences between the lymphoma cells. As demonstrated in patients with DLBCL and WM, ddPCR analysis of liquid biopsies can aid in monitoring the disease course, because of the highly sensitive identification and quantification of the variant allele frequency of MYD88(L265P).13831
An alternative technique for ctDNA analysis is targeted next-generation sequencing. The benefit of this technique over ddPCR is the possibility of identifying multiple variants at the same time, as was shown by Bohers et al.139 and Kurtz et al.140 in liquid biopsies from 30 and 217 DLBCL patients, respectively. The mutational burden of most of their patients, with a median of 117 variants per patient, was sufficient for disease monitoring. This novel way of disease monitoring could enhance evaluation of treatment responses (Figure 3). In their studies, the tumor burden, as measured by positron emission tomography-computed tomography scans, was significantly correlated with the variant allele frequency of ctDNA both during and after treatment.140139 Given this recent progress in ctDNA analysis, liquid biopsies are a minimally invasive method for evaluation of the molecular profile and can be used for analysis of tumor burden, disease progression, and treatment efficacy in patients with B-NHL.
Conclusions and future perspectives
Routine diagnostics in B-NHL are moving forward from classical morphology and immunohistochemistry towards the implementation of genetic analysis. In several subtypes of B-NHL subtype, MYD88(L265P) plays a crucial role as a driver of lymphomagenesis and can be used as a diagnostic classifier, as well as a prognostic factor and predictive biomarker. B-NHL with MYD88(L265P) can be (in)directly targeted by several novel therapeutic strategies and prospective clinical trials investigating these strategies are ongoing. We expect that that these theranostic strategies will be guided by analysis of MYD88(L265P) in liquid biopsies to monitor disease progression and determine response to therapy. Altogether, given the significant clinical relevance of MYD88(L265P), we advocate evaluation of MYD88 mutational status in routine diagnostics of B-NHL.
Footnotes
- Check the online version for the most updated information on this article, online supplements, and information on authorship & disclosures: www.haematologica.org/content/104/12/2337
- Received May 17, 2019.
- Accepted September 19, 2019.
References
- Vermaat JS, Pals ST, Younes A. Precision medicine in diffuse large B-cell lymphoma: hitting the target. Haematologica. 2015; 100(8):989-993. PubMedhttps://doi.org/10.3324/haematol.2015.128371Google Scholar
- Ngo VN, Young RM, Schmitz R. Oncogenically active MYD88 mutations in human lymphoma. Nature. 2011; 470(7332):115-119. PubMedhttps://doi.org/10.1038/nature09671Google Scholar
- Dubois S, Viailly PJ, Bohers E. Biological and clinical relevance of associated genomic alterations in MYD88 L265P and non-L265P-mutated diffuse large B-cell lymphoma: analysis of 361 cases. Clin Cancer Res. 2017; 23(9):2232-2244. PubMedhttps://doi.org/10.1158/1078-0432.CCR-16-1922Google Scholar
- Deguine J, Barton GM. MyD88: a central player in innate immune signaling. F1000Prime Rep. 2014; 6:97. PubMedhttps://doi.org/10.12703/p6-97Google Scholar
- Lin SC, Lo YC, Wu H. Helical assembly in the MyD88-IRAK4-IRAK2 complex in TLR/IL-1R signalling. Nature. 2010; 465(7300):885-890. PubMedhttps://doi.org/10.1038/nature09121Google Scholar
- Perkins ND. The diverse and complex roles of NF-kappaB subunits in cancer. Nat Rev Cancer. 2012; 12(2):121-132. PubMedhttps://doi.org/10.1038/nrc3204Google Scholar
- Ansell SM, Hodge LS, Secreto FJ. Activation of TAK1 by MYD88 L265P drives malignant B-cell growth in non-Hodgkin lymphoma. Blood Cancer J. 2014; 4:e183. PubMedhttps://doi.org/10.1038/bcj.2014.4Google Scholar
- Phelan JD, Young RM, Webster DE. A multiprotein supercomplex controlling oncogenic signalling in lymphoma. Nature. 2018; 560(7718):387-391. https://doi.org/10.1038/s41586-018-0290-0Google Scholar
- Puente XS, Pinyol M, Quesada V. Whole-genome sequencing identifies recurrent mutations in chronic lymphocytic leukaemia. Nature. 2011; 475(7354):101-105. PubMedhttps://doi.org/10.1038/nature10113Google Scholar
- Boudesco C, Verhoeyen E, Martin L. HSP110 sustains chronic NF-kappaB signaling in activated B-cell diffuse large B-cell lymphoma through MyD88 stabilization. Blood. 2018; 132(5):510-520. PubMedhttps://doi.org/10.1182/blood-2017-12-819706Google Scholar
- Ni HW, Shirazi F, Baladandayuthapani V. Targeting myddosome signaling in Waldenstrom’s macroglobulinemia with the interleukin-1 receptor-associated kinase 1/4 inhibitor R191. Clin Cancer Res. 2018; 24(24):6408-6420. PubMedhttps://doi.org/10.1158/1078-0432.CCR-17-3265Google Scholar
- Rousseau S, Martel G. Gain-of-function mutations in the toll-like receptor pathway: TPL2-mediated ERK1/ERK2 MAPK activation, a path to tumorigenesis in lymphoid neoplasms?. Front Cell Dev Biol. 2016; 4:50. Google Scholar
- Kurosaki T, Hikida M. Tyrosine kinases and their substrates in B lymphocytes. Immunol Rev. 2009; 228(1):132-148. PubMedhttps://doi.org/10.1111/j.1600-065X.2008.00748.xGoogle Scholar
- Xu Y, Xu L, Zhao M. No receptor stands alone: IgG B-cell receptor intrinsic and extrinsic mechanisms contribute to anti body memory. Cell Res. 2014; 24(6):651-664. PubMedhttps://doi.org/10.1038/cr.2014.65Google Scholar
- Knittel G, Liedgens P, Korovkina D, Pallasch CP, Reinhardt HC. Rewired NFkappaB signaling as a potentially actionable feature of activated B-cell-like diffuse large B-cell lymphoma. Eur J Haematol. 2016; 97(6):499-510. https://doi.org/10.1111/ejh.12792Google Scholar
- Yang G, Zhou Y, Liu X. A mutation in MYD88 (L265P) supports the survival of lymphoplasmacytic cells by activation of Bruton tyrosine kinase in Waldenstrom macroglobulinemia. Blood. 2013; 122(7):1222-1232. PubMedhttps://doi.org/10.1182/blood-2012-12-475111Google Scholar
- Wang JQ, Jeelall YS, Humburg P. Synergistic cooperation and crosstalk between MYD88(L265P) and mutations that dysregulate CD79B and surface IgM. J Exp Med. 2017; 214(9):2759-2776. PubMedhttps://doi.org/10.1084/jem.20161454Google Scholar
- Yu X, Li W, Deng Q. MYD88 L265P mutation in lymphoid malignancies. Cancer Res. 2018; 78(10):2457-2462. PubMedhttps://doi.org/10.1158/0008-5472.CAN-18-0215Google Scholar
- Wilson WH, Young RM, Schmitz R. Targeting B cell receptor signaling with ibrutinib in diffuse large B cell lymphoma. Nat Med. 2015; 21(8):922-926. PubMedhttps://doi.org/10.1038/nm.3884Google Scholar
- Lu L, Zhu F, Zhang M. Gene regulation and suppression of type I interferon signaling by STAT3 in diffuse large B cell lymphoma. Proc Natl Acad Sci U S A. 2018; 115(3):E498-E505. PubMedhttps://doi.org/10.1073/pnas.1715118115Google Scholar
- Yang G, Buhrlage SJ, Tan L. HCK is a survival determinant transactivated by mutated MYD88, and a direct target of ibrutinib. Blood. 2016; 127(25):3237-3252. PubMedhttps://doi.org/10.1182/blood-2016-01-695098Google Scholar
- Lee JH, Jeong H, Choi JW, Oh H, Kim YS. Clinicopathologic significance of MYD88 L265P mutation in diffuse large B-cell lymphoma: a meta-analysis. Sci Rep. 2017; 7(1):1785. Google Scholar
- Onaindia A, Medeiros LJ, Patel KP. Clinical utility of recently identified diagnostic, prognostic, and predictive molecular biomarkers in mature B-cell neoplasms. Mod Pathol. 2017; 30(10):1338-1366. https://doi.org/10.1038/modpathol.2017.58Google Scholar
- Baer C, Dicker F, Kern W, Haferlach T, Haferlach C. Genetic characterization of MYD88-mutated lymphoplasmacytic lymphoma in comparison with MYD88-mutated chronic lymphocytic leukemia. Leukemia. 2017; 31(6):1355-1362. Google Scholar
- Ballester LY, Loghavi S, Kanagal-Shamanna R. Clinical validation of a CXCR4 mutation screening assay for Waldenstrom macroglobulinemia. Clin Lymphoma Myeloma Leuk. 2016; 16(7):395-403. Google Scholar
- Cilla N, Vercruyssen M, Ameye L. Diagnostic approach of an IgM monoclonal gammopathy and clinical importance of gene MYD88 L265P mutation. Rev Med Brux. 2018. Google Scholar
- Fang H, Kapoor P, Gonsalves WI. Defining lymphoplasmacytic lymphoma: does MYD88L265P define a pathologically distinct entity among patients with an IgM paraprotein and bone marrow-based low-grade B-cell lymphomas with plasmacytic differentiation?. Am J Clin Pathol. 2018; 150(2):168-176. Google Scholar
- Insuasti-Beltran G, Gale JM, Wilson CS, Foucar K, Czuchlewski DR. Significance of MYD88 L265P mutation status in the sub-classification of low-grade B-cell lymphoma/leukemia. Arch Pathol Lab Med. 2015; 139(8):1035-1041. Google Scholar
- Martinez-Lopez A, Curiel-Olmo S, Mollejo M. MYD88 (L265P) somatic mutation in marginal zone B-cell lymphoma. Am J Surg Pathol. 2015; 39(5):644-651. PubMedhttps://doi.org/10.1097/PAS.0000000000000411Google Scholar
- Ondrejka SL, Lin JJ, Warden DW, Durkin L, Cook JR, Hsi ED. MYD88 L265P somatic mutation: its usefulness in the differential diagnosis of bone marrow involvement by B-cell lymphoproliferative disorders. Am J Clin Pathol. 2013; 140(3):387-394. PubMedhttps://doi.org/10.1309/AJCP10ZCLFZGYZIPGoogle Scholar
- Drandi D, Genuardi E, Dogliotti I. Highly sensitive MYD88(L265P) mutation detection by droplet digital polymerase chain reaction in Waldenstrom macroglobulinemia. Haematologica. 2018; 103(6):1029-1037. PubMedhttps://doi.org/10.3324/haematol.2017.186528Google Scholar
- Poulain S, Roumier C, Decambron A. MYD88 L265P mutation in Waldenstrom macroglobulinemia. Blood. 2013; 121(22):4504-4511. PubMedhttps://doi.org/10.1182/blood-2012-06-436329Google Scholar
- Varettoni M, Boveri E, Zibellini S. Clinical and molecular characteristics of lymphoplasmacytic lymphoma not associated with an IgM monoclonal protein: a multicentric study of the rete ematologica lombarda (REL) network. Am J Hematol. 2019. Google Scholar
- Xu L, Hunter ZR, Tsakmaklis N. Clonal architecture of CXCR4 WHIM-like mutations in Waldenstrom macroglobulinaemia. Br J Haematol. 2016; 172(5):735-744. PubMedhttps://doi.org/10.1111/bjh.13897Google Scholar
- Xu L, Hunter ZR, Yang G. Detection of MYD88 L265P in peripheral blood of patients with Waldenstrom’s macroglobulinemia and IgM monoclonal gammopathy of undetermined significance. Leukemia. 2014; 28(8):1698-1704. Google Scholar
- Treon SP, Cao Y, Xu L, Yang G, Liu X, Hunter ZR. Somatic mutations in MYD88 and CXCR4 are determinants of clinical presentation and overall survival in Waldenstrom macroglobulinemia. Blood. 2014; 123(18):2791-2796. PubMedhttps://doi.org/10.1182/blood-2014-01-550905Google Scholar
- Abeykoon JP, Paludo J, King RL. MYD88 mutation status does not impact overall survival in Waldenstrom macroglobulinemia. Am J Hematol. 2018; 93(2):187-194. Google Scholar
- Ali YB, Foad RM, Abdel-Wahed E. Lack of associations between TLR9 and MYD88 gene polymorphisms and risk of chronic lymphocytic leukemia. Asian Pac J Cancer Prev. 2017; 18(12):3245-3250. Google Scholar
- Improgo MR, Tesar B, Klitgaard JL. MYD88 L265P mutations identify a prognostic gene expression signature and a pathway for targeted inhibition in CLL. Br J Haematol. 2019; 184(6):925-936. Google Scholar
- Jiang M, Li J, Zhou J, Xing C, Xu JJ, Guo F. High-resolution melting analysis for rapid and sensitive MYD88 screening in chronic lymphocytic leukemia. Oncol Lett. 2019; 18(1):814-821. Google Scholar
- Leeksma AC, Taylor J, Wu B. Clonal diversity predicts adverse outcome in chronic lymphocytic leukemia. Leukemia. 2019; 33(2):390-402. Google Scholar
- Maleki Y, Alahbakhshi Z, Heidari Z. NOTCH1, SF3B1, MDM2 and MYD88 mutations in patients with chronic lymphocytic leukemia. Oncol Lett. 2019; 17(4):4016-4023. Google Scholar
- Patkar N, Subramanian PG, Deshpande P. MYD88 mutant lymphoplasmacytic lymphoma/Waldenstrom macroglobulinemia has distinct clinical and pathological features as compared to its mutation negative counterpart. Leuk Lymphoma. 2015; 56(2):420-425. PubMedhttps://doi.org/10.3109/10428194.2014.924123Google Scholar
- Putowski M, Podgorniak M, Pirog M. Prognostic impact of NOTCH1, MYD88, and SF3B1 mutations in Polish patients with chronic lymphocytic leukemia. Pol Arch Intern Med. 2017; 127(4):238-244. Google Scholar
- Qin SC, Xia Y, Miao Y. MYD88 mutations predict unfavorable prognosis in chronic lymphocytic leukemia patients with mutated IGHV gene. Blood Cancer J. 2017; 7(12):651. Google Scholar
- Quijada-Alamo M, Hernandez-Sanchez M, Robledo C. Next-generation sequencing and FISH studies reveal the appearance of gene mutations and chromosomal abnormalities in hematopoietic progenitors in chronic lymphocytic leukemia. J Hematol Oncol. 2017; 10(1):83. Google Scholar
- Rigolin GM, Saccenti E, Bassi C. Extensive next-generation sequencing analysis in chronic lymphocytic leukemia at diagnosis: clinical and biological correlations. J Hematol Oncol. 2016; 9(1):88. Google Scholar
- Rizzo D, Chauzeix J, Trimoreau F. IgM peak independently predicts treatment-free survival in chronic lymphocytic leukemia and correlates with accumulation of adverse oncogenetic events. Leukemia. 2015; 29(2):337-345. Google Scholar
- Rossi D, Rasi S, Spina V. Integrated mutational and cytogenetic analysis identifies new prognostic subgroups in chronic lymphocytic leukemia. Blood. 2013; 121(8):1403-1412. PubMedhttps://doi.org/10.1182/blood-2012-09-458265Google Scholar
- Sutton LA, Young E, Baliakas P. Different spectra of recurrent gene mutations in subsets of chronic lymphocytic leukemia harboring stereotyped B-cell receptors. Haematologica. 2016; 101(8):959-967. PubMedhttps://doi.org/10.3324/haematol.2016.141812Google Scholar
- Vollbrecht C, Mairinger FD, Koitzsch U. Comprehensive analysis of disease-related genes in chronic lymphocytic leukemia by multiplex PCR-based next generation sequencing. PLoS One. 2015; 10(6):e0129544. Google Scholar
- Wu SJ, Lin CT, Agathangelidis A. Distinct molecular genetics of chronic lymphocytic leukemia in Taiwan: clinical and pathogenetic implications. Haematologica. 2017; 102(6):1085-1090. PubMedhttps://doi.org/10.3324/haematol.2016.157552Google Scholar
- Puente XS, Bea S, Valdes-Mas R. Non-coding recurrent mutations in chronic lymphocytic leukaemia. Nature. 2015; 526(7574):519-524. PubMedhttps://doi.org/10.1038/nature14666Google Scholar
- Agathangelidis A, Ljungstrom V, Scarfo L. Highly similar genomic landscapes in monoclonal B-cell lymphocytosis and ultra-stable chronic lymphocytic leukemia with low frequency of driver mutations. Haematologica. 2018; 103(5):865-873. PubMedhttps://doi.org/10.3324/haematol.2017.177212Google Scholar
- Clipson A, Wang M, de Leval L. KLF2 mutation is the most frequent somatic change in splenic marginal zone lymphoma and identifies a subset with distinct genotype. Leukemia. 2015; 29(5):1177-1185. PubMedhttps://doi.org/10.1038/leu.2014.330Google Scholar
- Traverse-Glehen A, Bachy E, Baseggio L. Immunoarchitectural patterns in splenic marginal zone lymphoma: correlations with chromosomal aberrations, IGHV mutations, and survival. A study of 76 cases. Histopathology. 2013; 62(6):876-893. PubMedhttps://doi.org/10.1111/his.12092Google Scholar
- Jallades L, Baseggio L, Sujobert P. Exome sequencing identifies recurrent BCOR alterations and the absence of KLF2, TNFAIP3 and MYD88 mutations in splenic diffuse red pulp small B-cell lymphoma. Haematologica. 2017; 102(10):1758-1766. PubMedhttps://doi.org/10.3324/haematol.2016.160192Google Scholar
- Maitre E, Bertrand P, Maingonnat C. New generation sequencing of targeted genes in the classical and the variant form of hairy cell leukemia highlights mutations in epigenetic regulation genes. Oncotarget. 2018; 9(48):28866-28876. Google Scholar
- Staiger AM, Ott MM, Parmentier S. Allele-specific PCR is a powerful tool for the detection of the MYD88 L265P mutation in diffuse large B cell lymphoma and decalcified bone marrow samples. Br J Haematol. 2015; 171(1):145-148. Google Scholar
- Hamadeh F, MacNamara SP, Aguilera NS, Swerdlow SH, Cook JR. MYD88 L265P mutation analysis helps define nodal lymphoplasmacytic lymphoma. Mod Pathol. 2015; 28(4):564-574. PubMedhttps://doi.org/10.1038/modpathol.2014.120Google Scholar
- King RL, Gonsalves WI, Ansell SM. Lymphoplasmacytic lymphoma With a non-IgM paraprotein shows clinical and pathologic heterogeneity and may harbor MYD88 L265P mutations. Am J Clin Pathol. 2016; 145(6):843-851. PubMedhttps://doi.org/10.1093/ajcp/aqw072Google Scholar
- Varettoni M, Zibellini S, Boveri E. A risk-stratification model based on the initial concentration of the serum monoclonal protein and MYD88 mutation status identifies a subset of patients with IgM monoclonal gammopathy of undetermined significance at high risk of progression to Waldenstrom macroglobulinaemia or other lymphoproliferative disorders. Br J Haematol. 2019. Google Scholar
- Angelova EA, Li S, Wang W. IgM plasma cell myeloma in the era of novel therapy: a clinicopathological study of 17 cases. Hum Pathol. 2019; 84:321-334. Google Scholar
- Li ZM, Rinaldi A, Cavalli A. MYD88 somatic mutations in MALT lymphomas. Br J Haematol. 2012; 158(5):662-664. PubMedhttps://doi.org/10.1111/j.1365-2141.2012.09176.xGoogle Scholar
- Moody S, Escudero-Ibarz L, Wang M. Significant association between TNFAIP3 inactivation and biased immunoglobulin heavy chain variable region 4-34 usage in mucosa-associated lymphoid tissue lymphoma. J Pathol. 2017; 243(1):3-8. Google Scholar
- Gurth M, Bernard V, Bernd HW, Schemme J, Thorns C. Nodal marginal zone lymphoma: mutation status analyses of CD79A, CD79B, and MYD88 reveal no specific recurrent lesions. Leuk Lymphoma. 2017; 58(4):979-981. Google Scholar
- Hung SS, Meissner B, Chavez EA. Assessment of capture and amplicon-based aspproaches for the development of a targeted next-generation sequencing pipeline to personalize lymphoma management. J Mol Diagn. 2018; 20(2):203-214. Google Scholar
- Okosun J, Bodor C, Wang J. Integrated genomic analysis identifies recurrent mutations and evolution patterns driving the initiation and progression of follicular lymphoma. Nat Genet. 2014; 46(2):176-181. PubMedhttps://doi.org/10.1038/ng.2856Google Scholar
- Ozawa MG, Bhaduri A, Chisholm KM. A study of the mutational landscape of pediatric-type follicular lymphoma and pediatric nodal marginal zone lymphoma. Mod Pathol. 2016; 29(10):1212-1220. Google Scholar
- Louissaint A, Schafernak KT, Geyer JT. Pediatric-type nodal follicular lymphoma: a biologically distinct lymphoma with frequent MAPK pathway mutations. Blood. 2016; 128(8):1093-1100. PubMedhttps://doi.org/10.1182/blood-2015-12-682591Google Scholar
- Menguy S, Beylot-Barry M, Parrens M. Primary cutaneous large B-cell lymphomas: relevance of the 2017 World Health Organization classification: clinicopathological and molecular analyses of 64 cases. Histopathology. 2019; 74(7):1067-1080. Google Scholar
- Menguy S, Gros A, Pham-Ledard A. MYD88 somatic mutation is a diagnostic criterion in primary cutaneous large B-cell lymphoma. J Invest Dermatol. 2016; 136(8):1741-1744. Google Scholar
- Pham-Ledard A, Cappellen D, Martinez F, Vergier B, Beylot-Barry M, Merlio JP. MYD88 somatic mutation is a genetic feature of primary cutaneous diffuse large B-cell lymphoma, leg type. J Invest Dermatol. 2012; 132(8):2118-2120. PubMedhttps://doi.org/10.1038/jid.2012.102Google Scholar
- Shin SY, Lee ST, Kim HY. Detection of MYD88 L265P in patients with lymphoplas-macytic lymphoma/Waldenstrom macro -globulinemia and other B-cell non-Hodgkin lymphomas. Blood Res. 2016; 51(3):181-186. Google Scholar
- Aggarwal V, Das A, Bal A. MYD88, CARD11, and CD79B oncogenic mutations are rare events in the indian cohort of de novo nodal diffuse large B-cell lymphoma. Appl Immunohistochem Mol Morphol. 2019; 27(4):311-318. Google Scholar
- Cao Y, Zhu T, Zhang P. Mutations or copy number losses of CD58 and TP53 genes in diffuse large B cell lymphoma are independent unfavorable prognostic factors. Oncotarget. 2016; 7(50):83294-83307. Google Scholar
- Chapuy B, Stewart C, Dunford AJ. Molecular subtypes of diffuse large B cell lymphoma are associated with distinct pathogenic mechanisms and outcomes. Nat Med. 2018; 24(5):679-690. Google Scholar
- Fogliatto L, Grokoski KC, Strey YM. Prognostic impact of MYD88 mutation, proliferative index and cell origin in diffuse large B cell lymphoma. Hematol Transfus Cell Ther. 2019; 41(1):50-56. Google Scholar
- Intlekofer AM, Joffe E, Batlevi CL. Integrated DNA/RNA targeted genomic profiling of diffuse large B-cell lymphoma using a clinical assay. Blood Cancer J. 2018; 8(6):60. Google Scholar
- Reddy A, Zhang J, Davis NS. Genetic and functional drivers of diffuse large B cell lymphoma. Cell. 2017; 171(2):481-494. PubMedhttps://doi.org/10.1016/j.cell.2017.09.027Google Scholar
- Schmitz R, Wright GW, Huang DW. Genetics and pathogenesis of diffuse large B-cell lymphoma. N Engl J Med. 2018; 378(15):1396-1407. PubMedhttps://doi.org/10.1056/NEJMoa1801445Google Scholar
- Tadic L, Marjanovic G, Macukanovic-Golubovic L. The importance of Myd88 L265P mutation, clinical and immunohistochemical prognostic factors for the survival of patients with diffuse large B-cell non-Hodgkin lymphoma treated by immunochemotherapy in southeast Serbia. J BUON. 2016; 21(5):1259-1267. Google Scholar
- Vermaat JS, Somers SF, de Wreede LC. MYD88 mutations identify a molecular subgroup of diffuse large B-cell lymphoma with an unfavourable prognosis. Haematologica. 2019. Google Scholar
- Xu PP, Zhong HJ, Huang YH. B-cell function gene mutations in diffuse large B-cell lymphoma: a retrospective cohort study. EBioMedicine. 2017; 16:106-114. Google Scholar
- Ren W, Ye X, Su H. Genetic landscape of hepatitis B virus-associated diffuse large B-cell lymphoma. Blood. 2018; 131(24):2670-2681. PubMedhttps://doi.org/10.1182/blood-2017-11-817601Google Scholar
- Nayyar N, White MD, Gill CM. MYD88 L265P mutation and CDKN2A loss are early mutational events in primary central nervous system diffuse large B-cell lymphomas. Blood Adv. 2019; 3(3):375-383. PubMedhttps://doi.org/10.1182/bloodadvances.2018027672Google Scholar
- Sethi TK, Kovach AE, Grover NS. Clinicopathologic correlates of MYD88 L265P mutation and programmed cell death (PD-1) pathway in primary central nervous system lymphoma. Leuk Lymphoma. 2019;1-10. Google Scholar
- Zheng M, Perry AM, Bierman P. Frequency of MYD88 and CD79B mutations, and MGMT methylation in primary central nervous system diffuse large B-cell lymphoma. Neuropathology. 2017; 37(6):509-516. Google Scholar
- Ducharme O, Beylot-Barry M, Pham-Ledard A. Mutations of the B-cell receptor pathway confer chemoresistance in primary cutaneous diffuse large B-cell lymphoma leg-type. J Invest Dermatol. 2019. Google Scholar
- Mareschal S, Pham-Ledard A, Viailly PJ. Identification of somatic mutations in primary cutaneous diffuse large B-cell lymphoma, leg type by massive parallel sequencing. J Invest Dermatol. 2017; 137(9):1984-1994. Google Scholar
- Pham-Ledard A, Prochazkova-Carlotti M, Andrique L. Multiple genetic alterations in primary cutaneous large B-cell lymphoma, leg type support a common lymphomagenesis with activated B-cell-like diffuse large B-cell lymphoma. Mod Pathol. 2014; 27(3):402-411. Google Scholar
- Kataoka K, Miyoshi H, Sakata S. Frequent structural variations involving programmed death ligands in Epstein-Barr virus-associated lymphomas. Leukemia. 2019; 33(7):1687-1699. Google Scholar
- Ohata Y, Tatsuzawa A, Ohyama Y. A distinctive subgroup of oral EBV+ B-cell neoplasm with polymorphous features is potentially identical to EBV+ mucocutaneous ulcer. Hum Pathol. 2017; 69:129-139. Google Scholar
- Gebauer N, Hardel TT, Gebauer J. Activating mutations affecting the NF-kappa B pathway and EZH2-mediated epigenetic regulation are rare events in primary mediastinal large B-cell lymphoma. Anticancer Res. 2014; 34(10):5503-5507. PubMedGoogle Scholar
- Schrader AMR, Jansen PM, Willemze R. High prevalence of MYD88 and CD79B mutations in intravascular large B-cell lymphoma. Blood. 2018; 131(18):2086-2089. PubMedhttps://doi.org/10.1182/blood-2017-12-822817Google Scholar
- Kraan W, Horlings HM, van Keimpema M. High prevalence of oncogenic MYD88 and CD79B mutations in diffuse large B-cell lymphomas presenting at immune-privileged sites. Blood Cancer J. 2013; 3:e139. PubMedhttps://doi.org/10.1038/bcj.2013.28Google Scholar
- Carreno E, Clench T, Steeples LR. Clinical spectrum of vitreoretinal lymphoma and its association with MyD88 L265P mutation. Acta Ophthalmol. 2019; 97(1):e138-e139. Google Scholar
- Yonese I, Takase H, Yoshimori M. CD79B mutations in primary vitreoretinal lymphoma: diagnostic and prognostic potential. Eur J Haematol. 2019; 102(2):191-196. Google Scholar
- Franco F, Gonzalez-Rincon J, Lavernia J. Mutational profile of primary breast diffuse large B-cell lymphoma. Oncotarget. 2017; 8(61):102888-102897. Google Scholar
- de Groen RAL, Ibramoglu MS, van Eijk R. Molecular profiling of primary bone lymphomas reveals frequent mutations in EZH2 and other epigenetic genes: Implications for targeted treatment. Hemasphere. 2019; 3:212. Google Scholar
- Hallas C, Preukschas M, Tiemann M. Immunohistochemical distinction of ABC and GCB in extranodal DLBCL is not reflected in mutation patterns. Leuk Res. 2019; 76:107-111. Google Scholar
- Xu Y, Li J, Ouyang J. Prognostic relevance of protein expression, clinical factors, and MYD88 mutation in primary bone lymphoma. Oncotarget. 2017; 8(39):65609-65619. Google Scholar
- Brenner I, Roth S, Flossbach L, Wobser M, Rosenwald A, Geissinger E. Lack of myeloid differentiation primary response protein MyD88 L265P mutation in primary cutaneous marginal zone lymphoma. Br J Dermatol. 2015; 173(6):1527-1528. Google Scholar
- Wobser M, Maurus K, Roth S. Myeloid differentiation primary response 88 mutations in a distinct type of cutaneous marginal-zone lymphoma with a nonclass-switched immunoglobulin M immunophenotype. Br J Dermatol. 2017; 177(2):564-566. Google Scholar
- Zhu D, Ikpatt OF, Dubovy SR. Molecular and genomic aberrations in Chlamydophila psittaci negative ocular adnexal marginal zone lymphomas. Am J Hematol. 2013; 88(9):730-735. Google Scholar
- Je EM, Yoo NJ, Lee SH. Absence of MYD88 gene mutation in acute leukemias and multiple myelomas. Eur J Haematol. 2012; 88(3):273-274. PubMedGoogle Scholar
- Mori N, Ohwashi M, Yoshinaga K. L265P mutation of the MYD88 gene is frequent in Waldenstrom’s macroglobulinemia and its absence in myeloma. PLoS One. 2013; 8(11):e80088. PubMedhttps://doi.org/10.1371/journal.pone.0080088Google Scholar
- Kraan W, van Keimpema M, Horlings HM. High prevalence of oncogenic MYD88 and CD79B mutations in primary testicular diffuse large B-cell lymphoma. Leukemia. 2014; 28(3):719-720. PubMedhttps://doi.org/10.1038/leu.2013.348Google Scholar
- Sujobert P, Le Bris Y, Leval L. The need for a consensus next-generation sequencing panel for mature lymphoid malignancies. Hemasphere. 2018; 3(1)Google Scholar
- Swerdlow SH, Campo E, Pileri SA. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood. 2016; 127(20):2375-2390. PubMedhttps://doi.org/10.1182/blood-2016-01-643569Google Scholar
- Pham-Ledard A, Beylot-Barry M, Barbe C. High frequency and clinical prognostic value of MYD88 L265P mutation in primary cutaneous diffuse large B-cell lymphoma, leg-type. JAMA Dermatol. 2014; 150(11):1173-1179. Google Scholar
- Takano S, Hattori K, Ishikawa E. MyD88 mutation in elderly predicts poor prognosis in primary central nervous system lymphoma: multi-institutional analysis. World Neurosurg. 2018; 112:e69-e73. Google Scholar
- Yu S, Luo H, Pan M. High frequency and prognostic value of MYD88 L265P mutation in diffuse large B-cell lymphoma with R-CHOP treatment. Oncol Lett. 2018; 15(2):1707-1715. Google Scholar
- Lee YS, Liu J, Fricano KA. Lack of a Prognostic impact of the MyD88 L265P mutation for diffuse large B cell lymphoma patients undergoing autologous stem cell transplantation. Biol Blood Marrow Transplant. 2017; 23(12):2199-2204. Google Scholar
- Parry M, Rose-Zerilli MJ, Ljungstrom V. Genetics and prognostication in splenic marginal zone lymphoma: revelations from deep sequencing. Clin Cancer Res. 2015; 21(18):4174-4183. PubMedhttps://doi.org/10.1158/1078-0432.CCR-14-2759Google Scholar
- Gertz MA. Waldenstrom macroglobulinemia: 2019 update on diagnosis, risk stratification, and management. Am J Hematol. 2019; 94(2):266-276. Google Scholar
- Treon SP, Gustine J, Xu L. MYD88 wild-type Waldenstrom macroglobulinaemia: differential diagnosis, risk of histological transformation, and overall survival. Br J Haematol. 2018; 180(3):374-380. Google Scholar
- Hunter ZR, Xu L, Tsakmaklis N. Insights into the genomic landscape of MYD88 wild-type Waldenstrom macroglobulinemia. Blood Adv. 2018; 2(21):2937-2946. PubMedhttps://doi.org/10.1182/bloodadvances.2018022962Google Scholar
- Weber ANR, Cardona Gloria Y, Cinar O, Reinhardt HC, Pezzutto A, Wolz OO. Oncogenic MYD88 mutations in lymphoma: novel insights and therapeutic possibilities. Cancer Immunol Immunother. 2018; 67(11):1797-1807. Google Scholar
- Grommes C, Pastore A, Palaskas N. Ibrutinib unmasks critical role of bruton tyrosine kinase in primary CNS pymphoma. Cancer Discov. 2017; 7(9):1018-1029. PubMedhttps://doi.org/10.1158/2159-8290.CD-17-0613Google Scholar
- Younes A, Sehn LH, Johnson P. Randomized phase III trial of ibrutinib and rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone in non-germinal center B-cell diffuse large B-cell lymphoma. J Clin Oncol. 2019; 37(15):1285-1295. Google Scholar
- Chen JG, Liu X, Munshi M. BTK(Cys481Ser) drives ibrutinib resistance via ERK1/2 and protects BTK(wild-type) MYD88-mutated cells by a paracrine mechanism. Blood. 2018; 131(18):2047-2059. PubMedhttps://doi.org/10.1182/blood-2017-10-811752Google Scholar
- Woyach JA, Furman RR, Liu TM. Resistance mechanisms for the Bruton’s tyrosine kinase inhibitor ibrutinib. N Engl J Med. 2014; 370(24):2286-2294. PubMedhttps://doi.org/10.1056/NEJMoa1400029Google Scholar
- Cao Y, Hunter ZR, Liu X. CXCR4 WHIM-like frameshift and nonsense mutations promote ibrutinib resistance but do not supplant MYD88(L265P) -directed survival signalling in Waldenstrom macroglobulinaemia cells. Br J Haematol. 2015; 168(5):701-707. PubMedhttps://doi.org/10.1111/bjh.13200Google Scholar
- Byrd JC, Harrington B, O’Brien S. Acalabrutinib (ACP-196) in relapsed chronic lymphocytic leukemia. N Engl J Med. 2016; 374(4):323-332. PubMedhttps://doi.org/10.1056/NEJMoa1509981Google Scholar
- Wang M, Rule S, Zinzani PL. Acalabrutinib in relapsed or refractory mantle cell lymphoma (ACE-LY-004): a single-arm, multicentre, phase 2 trial. Lancet. 2018; 391(10121):659-667. Google Scholar
- Trotman J, Opat S, Marlton P. Bruton’s tyrosine kinase (Btk) inhibitor Bgb-3111 demonstrates high very good partial response (VGPR) rate in patients with Waldenström macroglobulinemia (Wm). Hematol Oncol. 2017; 35(S2):70-71. Google Scholar
- Ghobrial IM, Witzig TE, Gertz M. Long-term results of the phase II trial of the oral mTOR inhibitor everolimus (RAD001) in relapsed or refractory Waldenstrom Macroglobulinemia. Am J Hematol. 2014; 89(3):237-242. PubMedhttps://doi.org/10.1002/ajh.23620Google Scholar
- Forero-Torres A, Ramchandren R, Yacoub A. Parsaclisib, a potent and highly selective PI3Kdelta inhibitor, in patients with relapsed or refractory B-cell malignancies. Blood. 2019; 133(16):1742-1752. PubMedhttps://doi.org/10.1182/blood-2018-08-867499Google Scholar
- He Y, Li J, Ding N. Combination of enzastaurin and ibrutinib synergistically induces anti-tumor effects in diffuse large B cell lymphoma. J Exp Clin Cancer Res. 2019; 38(1):86. Google Scholar
- Liu X, Hunter ZR, Xu L. Targeting myddosome assembly in Waldenstrom macroglobulinaemia. Br J Haematol. 2017; 177(5):808-813. Google Scholar
- Nelde A, Walz JS, Kowalewski DJ. HLA class I-restricted MYD88 L265P- derived peptides as specific targets for lymphoma immunotherapy. Oncoimmunology. 2017; 6(3):e1219825. Google Scholar
- Nielsen JS, Chang AR, Wick DA. Mapping the human T cell repertoire to recurrent driver mutations in MYD88 and EZH2 in lymphoma. Oncoimmunology. 2017; 6(7):e1321184. Google Scholar
- Qiu H, Gong S, Xu L. MYD88 L265P mutation promoted malignant B cell resistance against T cell-mediated cytotoxicity via upregulating the IL-10/STAT3 cascade. Int Immunopharmacol. 2018; 64:394-400. Google Scholar
- Camus V, Jardin F, Tilly H. The value of liquid biopsy in diagnosis and monitoring of diffuse large B-cell lymphoma: recent developments and future potential. Expert Rev Mol Diagn. 2017; 17(6):557-566. Google Scholar
- Scherer F, Kurtz DM, Newman AM. Distinct biological subtypes and patterns of genome evolution in lymphoma revealed by circulating tumor DNA. Sci Transl Med. 2016; 8(364):364ra155. PubMedhttps://doi.org/10.1126/scitranslmed.aai8545Google Scholar
- Hattori K, Sakata-Yanagimoto M, Kusakabe M. Genetic evidence implies that primary and relapsed tumors arise from common precursor cells in primary central nervous system lymphoma. Cancer Sci. 2019; 110(1):401-407. Google Scholar
- Camus V, Sarafan-Vasseur N, Bohers E. Digital PCR for quantification of recurrent and potentially actionable somatic mutations in circulating free DNA from patients with diffuse large B-cell lymphoma. Leuk Lymphoma. 2016; 57(9):2171-2179. Google Scholar
- Bohers E, Viailly PJ, Becker S. Noninvasive monitoring of diffuse large B-cell lymphoma by cell-free DNA high-throughput targeted sequencing: analysis of a prospective cohort. Blood Cancer J. 2018; 8(8):74. Google Scholar
- Kurtz DM, Scherer F, Jin MC. Circulating tumor DNA measurements as early outcome predictors in diffuse large B-cell lymphoma. J Clin Oncol. 2018; 36(28):2845-2853. Google Scholar
- Witzig TE, Reeder CB, LaPlant BR. A phase II trial of the oral mTOR inhibitor everolimus in relapsed aggressive lymphoma. Leukemia. 2011; 25(2):341-347. PubMedhttps://doi.org/10.1038/leu.2010.226Google Scholar