Abstract

Diffuse large B-cell lymphoma (DLBCL) encompasses a diverse spectrum of aggressive B-cell lymphomas with remarkable genetic heterogeneity and myriad clinical presentations. Multiplatform genomic analyses of DLBCL have identified oncogenic drivers within genetic subtypes that allow for pathologic subclassification of tumors into discrete entities with shared immunophenotypic, genetic, and clinical features. Robust classification of lymphoid tumors establishes a foundation for precision medicine and enables the identification of novel therapeutic vulnerabilities within biologically homogeneous entities. Most cases of DLBCL involving the central nervous system (CNS), vitreous, and testis exhibit immunophenotypic features suggesting an activated B-cell (ABC) origin. Shared molecular features include frequent comutations of MYD88 (L265P) and CD79B and frequent genetic alterations promoting immune evasion, which are hallmarks of the MCD/C5/MYD88 genetic subtype of DLBCL. Clinically, these lymphomas primarily arise within anatomic sanctuary sites and have a predilection for remaining confined to extranodal sites and strong CNS tropism. Given the shared clinical and molecular features, the umbrella term primary large B-cell lymphoma of immune-privileged sites (IP-LBCL) was proposed. Other extranodal DLBCL involving the breast, adrenal glands, and skin are often ABC DLBCL but are more heterogeneous in their genomic profile and involve anatomic sites that are not considered immune privileged. In this review, we describe the overlapping clinical, pathologic, and molecular features of IP-LBCL and highlight important considerations for diagnosis, staging, and treatment. We also discuss potential therapeutic vulnerabilities of IP-LBCL including sensitivity to inhibitors of Bruton tyrosine kinase, immunomodulatory agents, and immunotherapy.

Diffuse large B-cell lymphoma (DLBCL) is not a singular disease but describes a broad spectrum of aggressive B-cell lymphomas that can involve any organ system in the body. As the genetic basis underlying the clinical diversity emerges,1-4 the classification of lymphoid tumors evolves with the purpose of identifying discrete DLBCL subtypes that share common immunophenotypic, genetic, and clinical features.5,6 A subset of DLBCL demonstrate a strong predilection for involving extranodal anatomic regions including immune-privileged sanctuary sites such as the central nervous system (CNS), vitreous, and testis. Furthermore, these tumors frequently remain within immune-privileged sites at the time of relapse, including the CNS, suggesting they have unique genetic and/or microenvironmental selective pressures. Recent studies have demonstrated that primary DLBCL of the CNS (PCNSL), primary DLBCL of the testis (PTL), and primary vitreoretinal lymphoma (PVRL) share biologic and clinical features.7-11 Thus, the umbrella term primary large B-cell lymphomas involving immune-privileged sites (IP-LBCL) was proposed in the fifth edition of the World Health Organization classification of lymphoid neoplasms.6 Importantly, the term IP-LBCL is reserved for tumors that arise in immunocompetent hosts, and DLBCL arising in the CNS or testis in the setting of inherited or acquired immunodeficiencies have a different underlying biology and are classified separately.12,13 Notably, the International Consensus Classification system also debated creating an umbrella term for “extranodal lymphoma activated B-cell (ABC) type” to describe DLBCL that arise in immune privileged sites along with other lymphomas that share clinical and biologic features including intravascular LBCL (IVLBCL), primary cutaneous DLBCL, leg-type, and primary DLBCL of the breast or adrenals.

Here, we describe the biologic and clinical features of IP-LBCL and emphasize special considerations for staging and treatment. We have included IVLBCL given its strong biologic overlap with IP-LBCL and its confinement to the intravascular niche that may also represent a site of immune privilege. We discuss therapeutic vulnerabilities of these entities and highlight clinical trials testing the efficacy of inhibitors of Bruton tyrosine kinase (BTK), immunomodulatory agents, and immunotherapy.

IP-LBCL includes uncommon or rare lymphomas that share overlapping clinical features but with important differences (Table 1). One central unifying clinical feature is that they arise restricted to extranodal anatomic sites at diagnosis and also frequently remain within extranodal sites at relapse.14 Furthermore, these lesions either arise within the CNS or have a strong predilection for CNS progression, which can occur many years after treatment.14-16 “Immune privileged” refers to anatomic sanctuary sites such as the brain, cerebrospinal fluid (CSF), eyes, and the testes based on studies that demonstrated tissue allografted into these sites was less likely to undergo graft rejection.17 Tumor cells within these immune-privileged sites are thereby postulated to be less susceptible to antitumor immune responses from T cells and natural killer cells. Given the unique clinical features of IP-LBCL, special considerations are required for diagnosis and staging that differ from nodal-based lymphomas (Table 2).

Table 1.

Clinical features of IP-LBCL

PCNSLPVRLPTLIVLBCL 
Incidence ∼4% of malignant brain tumors; ∼1700 cases annually in the United States A rare subset of PCNSL; ∼50 cases annually in United States <5% of testicular cancers; commonest testicular malignancy in men aged >60 y Rare 
Age Median age 55 to 60 y; incidence rising in those aged >70 y Median age 50 to 60 y Median age 65 to 68 y Median age 60 to 70 y 
Presentation Focal neurologic deficits, cognitive/behavioral changes, or increased intracranial pressure Blurry vision and floaters; impaired visual acuity; symptoms precede diagnosis for years Firm, painless testicular mass; 40% associated with hydrocele Fever of unknown origin common, rapid weight loss; progressive neurologic signs; hemophagocytosis-associated variant 
Behavior Aggressive Indolent Aggressive Aggressive 
CNS involvement 100% 100% involve the eye; 60%-80% progress to CNS ∼25% relapse in CNS (can be late); most CNS progressions involve brain parenchyma Common 
Anatomic sites >90% involve brain parenchyma; 30% to 40% are multifocal; 15%-25% with concurrent brain/CSF; may relapse outside of CNS Infiltration of the vitreous; may also affect the retina 5%-10% involve contralateral testis; 20% to 30% have concurrent disease at extranodal sites Largely restricted to lumina of small blood vessels; the bone marrow, spleen, liver, skin, and CNS 
PCNSLPVRLPTLIVLBCL 
Incidence ∼4% of malignant brain tumors; ∼1700 cases annually in the United States A rare subset of PCNSL; ∼50 cases annually in United States <5% of testicular cancers; commonest testicular malignancy in men aged >60 y Rare 
Age Median age 55 to 60 y; incidence rising in those aged >70 y Median age 50 to 60 y Median age 65 to 68 y Median age 60 to 70 y 
Presentation Focal neurologic deficits, cognitive/behavioral changes, or increased intracranial pressure Blurry vision and floaters; impaired visual acuity; symptoms precede diagnosis for years Firm, painless testicular mass; 40% associated with hydrocele Fever of unknown origin common, rapid weight loss; progressive neurologic signs; hemophagocytosis-associated variant 
Behavior Aggressive Indolent Aggressive Aggressive 
CNS involvement 100% 100% involve the eye; 60%-80% progress to CNS ∼25% relapse in CNS (can be late); most CNS progressions involve brain parenchyma Common 
Anatomic sites >90% involve brain parenchyma; 30% to 40% are multifocal; 15%-25% with concurrent brain/CSF; may relapse outside of CNS Infiltration of the vitreous; may also affect the retina 5%-10% involve contralateral testis; 20% to 30% have concurrent disease at extranodal sites Largely restricted to lumina of small blood vessels; the bone marrow, spleen, liver, skin, and CNS 

IVLBCL is currently categorized as a discrete biologic entity in both the World Health Organization and International Consensus Classification systems.

Table 2.

Diagnosis and staging of IP-LBCL

PCNSLPVRLPTLIVLBCL
Diagnostic biopsy procedures     
Stereotactic brain biopsy Procedure of choice    
Vitreous fluid biopsy  Procedure of choice   
Orchiectomy   Procedure of choice  
Biopsy affected organ (the liver, bone marrow, or skin)    Procedure of choice 
Random skin biopsies    Consider 
Adjunctive tests (CSF or vitreous)     
Flow cytometry, cytology Recommended Recommended Recommended Recommended 
MYD88 mutation testing Consider Consider Consider Consider 
Immunoglobulin gene rearrangement Consider Consider Consider Consider 
ctDNA Consider Consider Consider Consider 
Cytokines (interleukin-6, interleukin-10) Consider Consider Not recommended Not recommended 
Laboratory tests     
Complete blood count, LDH, and liver and renal function Recommended Recommended Recommended Recommended 
HIV screening Recommended Recommended Recommended Recommended 
Hepatitis B screening Recommended Recommended Recommended Recommended 
Hepatitis C screening Recommended Recommended Recommended Recommended 
EBV serology Recommended Not recommended Not recommended Not recommended 
Staging procedures     
MRI of brain with gadolinium Essential Recommended Consider Recommended 
Dilated eye examination Recommended Essential Not recommended Not recommended 
Testicular ultrasound Consider Consider Essential Not recommended 
FDG-PET of body Recommended Recommended Recommended Recommended 
Peripheral blood flow cytometry Not recommended Not recommended Consider Consider 
MRI of spine Consider Not recommended Not recommended Not Recommended 
CT of body with contrast Consider Consider Consider Consider 
Bone marrow biopsy, aspirate Consider Not recommended Recommended Recommended 
PCNSLPVRLPTLIVLBCL
Diagnostic biopsy procedures     
Stereotactic brain biopsy Procedure of choice    
Vitreous fluid biopsy  Procedure of choice   
Orchiectomy   Procedure of choice  
Biopsy affected organ (the liver, bone marrow, or skin)    Procedure of choice 
Random skin biopsies    Consider 
Adjunctive tests (CSF or vitreous)     
Flow cytometry, cytology Recommended Recommended Recommended Recommended 
MYD88 mutation testing Consider Consider Consider Consider 
Immunoglobulin gene rearrangement Consider Consider Consider Consider 
ctDNA Consider Consider Consider Consider 
Cytokines (interleukin-6, interleukin-10) Consider Consider Not recommended Not recommended 
Laboratory tests     
Complete blood count, LDH, and liver and renal function Recommended Recommended Recommended Recommended 
HIV screening Recommended Recommended Recommended Recommended 
Hepatitis B screening Recommended Recommended Recommended Recommended 
Hepatitis C screening Recommended Recommended Recommended Recommended 
EBV serology Recommended Not recommended Not recommended Not recommended 
Staging procedures     
MRI of brain with gadolinium Essential Recommended Consider Recommended 
Dilated eye examination Recommended Essential Not recommended Not recommended 
Testicular ultrasound Consider Consider Essential Not recommended 
FDG-PET of body Recommended Recommended Recommended Recommended 
Peripheral blood flow cytometry Not recommended Not recommended Consider Consider 
MRI of spine Consider Not recommended Not recommended Not Recommended 
CT of body with contrast Consider Consider Consider Consider 
Bone marrow biopsy, aspirate Consider Not recommended Recommended Recommended 

CT, computed tomography EBV, Epstein-Barr virus; LDH, lactate dehydrogenase.

PCNSL

PCNSL is a rare and aggressive B-cell lymphoma confined to the brain, spine, CSF, or eyes at diagnosis.18,19 It constitutes ∼4% of newly diagnosed malignant brain tumors, with a slight male predominance.20 The incidence of PCNSL is rising among immunocompetent people particularly in individuals aged >70 years.21,22 Notably, CNS lymphomas that arise in the setting of immunodeficiency including human immunodeficiency virus and after solid organ transplantation are commonly associated with Epstein-Barr virus, have a different genetic landscape, and are considered separate entities.12,13 The clinical presentation may be an acute onset of focal neurologic deficits or more insidious with subtle cognitive or behavioral changes that develop over weeks. The natural history is highly aggressive and neurologic symptoms often progress rapidly. PCNSL involves the brain parenchyma in >90% of cases, with most lesions being supratentorial and 30% to 40% of cases showing multiple brain lesions.23 Concomitant involvement of the CSF or eyes occurs in 15% to 25% of cases, but isolated leptomeningeal involvement is rare. PCNSL may involve anatomic sites outside of the CNS at relapse.24 

Stereotactic needle biopsy of a brain parenchymal lesion is the diagnostic procedure of choice and surgical resection is not indicated. Empiric corticosteroid therapy before biopsy is strongly discouraged because it may result in the disappearance of the lesions, which is not sufficient evidence to diagnose lymphoma.25 The CSF should be evaluated with flow cytometry (preferred) and cytology in all patients unless contraindicated because of increased intracranial pressure. In cases in which biopsy is challenging, the demonstration of tumor cells in the CSF using flow cytometry or cytology can be diagnostic. MYD88 mutation and immunoglobin gene rearrangement testing in CSF can also be considered as adjunct diagnostic procedures.26 Circulating tumor DNA (ctDNA) can be detected in the CSF of virtually all patients and may emerge as a useful biomarker for diagnosis and/or prognosis.27 Interestingly, ctDNA can also be detected in the plasma of most patients, which may have prognostic relevance by identifying a high-risk group at diagnosis and as a noninvasive biomarker that can monitor treatment response.27 

Magnetic resonance imaging (MRI) scans of the brain with gadolinium are essential for tumor characterization, and spine MRI can be considered in patients with localizing symptoms. Staging procedures for PCNSL focus on ruling out the presence of systemic DLBCL, and fluorodeoxyglucose (FDG)-positron emission tomography (PET) scans are the most sensitive to evaluate for extranodal disease.28 All patients should have a dilated eye examination, and a testicular ultrasound should be considered in men. Bone marrow biopsy with aspirate have limited clinical utility in the modern era with routine use of FDG-PET scans.

PVRL

PVRL is a rare variant of PCNSL in which tumor cells are confined to the vitreous or retina without overt CNS involvement.29 Patients present with the subacute onset of blurry vision, floaters, or decreased visual acuity that mimics posterior uveitis. PVRL often has an indolent course and symptoms can precede diagnosis for years. Similar to PCNSL, empiric therapy with corticosteroids may transiently improve symptoms but may delay diagnosis confirmation. The diagnosis of PVRL can be challenging and relies on a combination of immunocytochemical or flow cytometry from vitrectomy specimens. Intravitreal levels of interleukin-10 and interleukin-6 may also assist in diagnosis.30MYD88 (L265P) mutation analysis in the vitreous may aid in diagnosis but is not widely available.31 Next-generation sequencing panels for ctDNA from the vitreous are also emerging as adjunctive diagnostic tools.11,27 

Ruling out CNS disease is critical and best performed with a contrasted MRI of the brain and CSF flow cytometry. Up to 80% of patients with PVRL will ultimately progress to the CNS, but systemic spread is rare. FDG-PET scans of the body are used to rule out systemic lymphomas with secondary involvement of intraocular structures; this is uncommon except for PTL and primary breast lymphoma, which can progress to the eye in isolation or with concomitant CNS relapse.32 

PTL

PTL is an uncommon lymphoma, but it is the most common testicular malignancy in men aged ≥60 years.33 The classic presentation is painless testicular enlargement and a hydrocele may coexist in up to 40% of cases.34 Most cases are localized to the testis, but PTL includes cases of systemic spread that commonly remain confined to extranodal sites including the CNS, skin, lung, soft tissues, and the contralateral testis.35 The diagnostic procedure of choice is an orchiectomy. FDG-PET scans are used to rule out systemic disease and bilateral testicular ultrasounds are essential. Most PTL cases are localized at diagnosis, with up to 10% involving the contralateral testis and ∼25% of cases with systemic involvement.36 Despite the limited stage, the clinical course of PTL is aggressive and demonstrates a continuous rate of relapse.35-37 Most relapses involve extranodal sites such as the contralateral testis or CNS that can occur many years after therapy36,38 and typically involve the brain parenchyma.15 For this reason, all patients with PTL should have a lumbar puncture with CSF studies, and a contrasted brain MRI should be considered. The biologic reasons for the CNS tropism of PTL remains unclear, but a recent study suggested that tumors with BCL6 or PDL1/2 rearrangements were associated with a higher risk of CNS relapse.39 The management of patients with PTL includes measures to prevent CNS relapse, and a recent prospective study demonstrated that intensive CNS prophylaxis including both intrathecal and intravenous methods was feasible and no CNS relapses were observed.37 Contralateral testis irradiation is administered to prevent local recurrence.

IVLBCL

IVLBCL is a rare and aggressive form of extranodal DLBCL characterized by tumor cells growing predominantly within the lumen of small blood vessels without associated lymphadenopathy, although extravascular location of tumor cells has been described.40-42 The diagnosis is challenging and often delayed because imaging scans are unrevealing and biopsy of an affected organ is required. Common symptoms are nonspecific including generalized fatigue, anorexia, rapid deterioration of performance status, weight loss, night sweats, neurologic symptoms, and fever of unknown origin.43 Random skin biopsies can be diagnostic of IVLBCL in cases of fever of unknown origin with elevated lactate dehydrogenase levels, poor performance status, and unexplained cytopenias, and may avoid unacceptably prolonged delays in treatment initiation.44 The natural history of IVLBCL is very aggressive and often results in progressive neurologic compromise without therapy. Cases isolated to the skin appear to have a better prognosis, likely because of earlier diagnosis and therapeutic intervention. A hemophagocytic syndrome–associated variant has been described, which more commonly involves the bone marrow and is often associated with fever and hepatosplenomegaly.45,46 Conventional staging procedures are not reliable and IVLBCL should be considered disseminated at diagnosis. Bone marrow biopsy, brain MRI with contrast, and lumbar puncture with CSF studies are recommended.40 ctDNA levels are often very high in IVLBCL and may emerge as an effective method for studying tumor genetics and monitoring therapeutic response.47 

Histologically, on conventional hematoxylin and eosin stains, these tumors are not unique (Figure 1). A common feature of cases presenting in the CNS and testis is the ABC or non–germinal center B-cell (non-GCB) profile using the Hans algorithm. Gene expression profiling studies are confirmatory of the ABC derivation. The tumor cells express B-cell–associated antigens including CD20, CD19, and CD79a. They are typically positive for MUM1/IRF4 but negative for CD10. BCL6 is usually positive and BCL2 is more variably expressed. Although MYC rearrangement is not a feature of these lesions, MYC protein is often positive. A common feature of DLBCL presenting in immune privileged sites is loss of expression of major histocompatibility complex (MHC) class 1 and class 2.2 Consistent with the failure of an immune response, infiltrating immune cells including T cells are generally few in number.

Figure 1.

Unifying clinical and biologic features of IP-LBCL and IVLBCL. (A) The involved anatomic sites are commonly restricted to extranodal regions at both diagnosis and upon relapse and include the CNS, eye, and testis. (B) The pathologic subtypes included under the proposed umbrella term IP-LBCL include primary CNS lymphoma, primary testicular lymphoma, and PVRL. IVLBCL is also an extranodal ABC lymphoma that shares features with IP-LBCL but does not arise in an anatomic site that is considered to be immune privileged. Primary CNS lymphoma, original magnification ×1000; primary testicular lymphoma, original magnification ×200; primary vitreoretinal lymphoma, original magnification ×1000; intravascular large B-cell lymphoma, original magnification ×400. (C) The molecular biology of IP-LBCL mostly closely resembles that of an ABC phenotype and the MCD genetic subtype of DLBCL. Four main biologic cornerstones that have been described in IP-LBCL include (1) activating mutations of MYD88 (L265P) and CD79B that form the MYD88-TLR9-BCR (My-T-BCR) complex and promote chronic active BCR signaling; (2) inactivating mutations of class 1 and 2 HLA expression as well as CD58, which promote immune escape; (3) loss of CDKN2A, which promotes unregulated cell cycle activation; and (4) inactivating mutations in transcription factors such as PRDM1 and TBL1XR1, that promote ongoing proliferation.

Figure 1.

Unifying clinical and biologic features of IP-LBCL and IVLBCL. (A) The involved anatomic sites are commonly restricted to extranodal regions at both diagnosis and upon relapse and include the CNS, eye, and testis. (B) The pathologic subtypes included under the proposed umbrella term IP-LBCL include primary CNS lymphoma, primary testicular lymphoma, and PVRL. IVLBCL is also an extranodal ABC lymphoma that shares features with IP-LBCL but does not arise in an anatomic site that is considered to be immune privileged. Primary CNS lymphoma, original magnification ×1000; primary testicular lymphoma, original magnification ×200; primary vitreoretinal lymphoma, original magnification ×1000; intravascular large B-cell lymphoma, original magnification ×400. (C) The molecular biology of IP-LBCL mostly closely resembles that of an ABC phenotype and the MCD genetic subtype of DLBCL. Four main biologic cornerstones that have been described in IP-LBCL include (1) activating mutations of MYD88 (L265P) and CD79B that form the MYD88-TLR9-BCR (My-T-BCR) complex and promote chronic active BCR signaling; (2) inactivating mutations of class 1 and 2 HLA expression as well as CD58, which promote immune escape; (3) loss of CDKN2A, which promotes unregulated cell cycle activation; and (4) inactivating mutations in transcription factors such as PRDM1 and TBL1XR1, that promote ongoing proliferation.

Close modal

The phenotype of IVLBCL is more heterogeneous.40 Although these tumors are often non-GCB by the Hans algorithm, CD5 is often positive, which is not generally a feature of tumors arising in the CNS or testis. Fewer data are available regarding the phenotype of vitreo-retinal lesions. The cells recovered from the vitreous are often markedly degenerate, and difficult to characterize by immunohistochemistry or flow cytometry.

Although histological and clinical heterogeneity of systemic DLBCL have long been appreciated, only recently have genetic studies been sufficiently powered to resolve these differences into distinct molecular subtypes. Gene expression studies first demonstrated that DLBCL consists of 2 main subtypes that share transcriptional patterns with either GCBs, so called GCB DLBCL, or with in vitro ABCs, so called ABC DLBCL. The ABC subtype was found to have high NF-κB activity and worse overall survival in response to cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP)-based therapies.48-51 This gene expression classification system, termed “cell of origin,” is an incredibly robust classification that yields greater reproducibility across tissue preparation and gene expression profiling platforms.52-54 Patients with ABCs show an enrichment for unique genomic alterations in genes including MYD88, CD79B, PIM1, and CDKN2A, among others; whereas patients with GCBs have higher mutation frequencies in EZH2, MEF2B, GNA13, BCL2, and SGK1.55-59 Subsequent functional studies have identified unique survival mechanisms used by each subtype, none perhaps as clinically relevant as the discovery of self-antigen–driven chronic active B-cell receptor (BCR) signaling in ABC DLBCL, which is sensitive to BTK inhibition.56,60 As such, since the revised fourth edition of the World Health Organization classification, studies to determine the cell of origin for newly diagnosed DLBCL have been recommended, with this guidance retained in the newer classification systems.5,6 IP-LBCL are heavily skewed to the non-GCB subtype, a histological method that approximates the ABC DLBCL cell-of-origin classification.61 

More recently, genetic studies of de novo DLBCL have described upward of 8 molecular subtypes of DLBCL. Despite using different sequencing approaches and statistical methods, each of the 3 models contains highly overlapping molecular features for a given genetic subtype.1-4 Notably, MYD88 (L265P) and CD79B immunoreceptor tyrosine-based activation motif (ITAM) mutations are hallmark features of the MCD/C5/MYD88 subtypes. Other MCD features are also frequently found in IP-LBCL including mutations and homozygous deletions in CDKN2A and HLA class 1 genes; evidence of somatic hypermutation targeting BTG1, BTG2, KLHL14, and PIM1; gains and amplifications in BCL2; as well as missense mutations in TBL1XR1.7,11,62-68 The MCD subtype has >30 genetic features (mutations, translocations, and DNA copy number alterations). Although the functional consequence of each have yet to be determined, many hallmarks of cancer are represented within the mutated gene list of MCD DLBCL including resisting cell death (BCL2), evading growth suppressors (CDK2NA), phenotypic plasticity (IRF4, PRDM1, SPIB, and TBL1XR1), and immune evasion (HLA-A, HLA-B, HLA-C, TAP1, CD58, and PDCD1LG2/CD274).

Given high expression of programmed death-ligand 1 (PD-L1)/PD-L269 and the success of immune checkpoint blockade therapy in Hodgkin lymphoma,70 the observation of amplification of chromosome 9p24, containing the PD-L1/PD-L2 loci in a subset MCD and IP-LBCL cases warrants a brief discussion of checkpoint blockade in IP-LBCL. In general, PD-L1 staining is higher in ABC DLBCL than in GCB DLBCL71 and in Epstein-Barr virus–positive DLBCL than in Epstein-Barr virus–negative DLBCL,72 however overall frequencies remain <20%.2 Within PCNSL and PTL, genetic alteration of the locus was reported in nearly 50% of cases7 and early case reports of relapsed cases of PD-L1–positive PCNSL and PTL showed high response rates to programmed death protein 1 blockade.73 Such high frequencies of PD-L1/PD-L2 have not been observed in subsequent studies8,68 and prospective clinical trials have shown disappointing results.74,75 

Conversely, other genomic alterations common to IP-LBCL are clinically actionable using BTK inhibitors, namely, the co-occurrence of MYD88 (L265P) and CD79B ITAM mutations. At first blush, this appears to be a contradiction. MYD88 is an innate immune signaling adapter protein that facilitates activation of downstream NF-κB, interferon regulatory factor (IRF), and MAPK signaling, whereas CD79B is 1 of 2 immunoglobulin containing, signaling competent components of the BCR. One might predict then that BTK inhibitors would be less effective in patients with DLBCL with both MYD88 (L265P) and CD79B mutations compared with those with mutant CD79B alone because of activation of a compensatory pathway driving NF-κB. Yet, double-mutant tumors are, in fact, the most sensitive to BTK inhibitors60 and patients with MCD subtype, in general, demonstrate the greatest benefit of combining BTK inhibitors with chemotherapy.76 

This apparent clinical contradiction was resolved with the discovery of the “My-T-BCR” supercomplex, named for the colocalization of MYD88 (L265P) with TLR9 and the BCR. The My-T-BCR is the site of NF-κB activation and exceptionally sensitive to BTK inhibition possibly because of autophagic degradation of mutant MYD88 by BTK inhibitors.77 The My-T-BCR complex was found in ABC and PCNSL tumors, and its presence correlated with clinical response to BTK inhibition.78 Thus, the BCR and MYD88 are not independent pathways that can activate NF-κB in MCD DLBCL but rather both proteins required to form a multiprotein supercomplex that mediates oncogenic signaling.

Other frequently mutated MCD genes may further promote this mode of signaling. For instance, KLHL14 is a Kelch-like BTB-containing subunit of the Cullin-RING E3 ligase complex. KLHL14 is localized to the cytoplasmic face of the endoplasmic reticulum in which it was recently shown to interact with CD79A, CD79B, and immunoglobulin M.79 Missense and nonsense mutations in KLHL14 are associated with MCD DLBCL, but only missense mutations were found to impair the growth of MCD DLBCL cell lines and decreased BCR expression when ectopically expressed. Conversely, mimicking KLHL14 nonsense mutations with CRISPR-mediated knockout of KLHL14 resulted in higher levels of surface BCR, increased My-T-BCR interactions, and resulted in a relative growth advantage to MCD DLBCL cell lines treated with ibrutinib. Taken together, these results implicate KLHL14 in a critical quality control mechanism of BCR expression and highlight the central role of My-T-BCR signaling to MCD-like lymphomas.

The clinical management of patients with IP-LBCL differs significantly when the CNS is involved compared with situations in which strategies to prevent CNS progression/relapse are considered. A thorough discussion is beyond the scope of this manuscript and is better reviewed elsewhere,18,19,80,81 but we emphasize important aspects of clinical management specific to IP-LBCL and highlight rational targeted agents being studied in PCNSL and PVRL, the prototypical MCD/C5/MYD88 tumor, because these therapies may ultimately prove clinical utility across all IP-LBCLs (Table 3).

Table 3.

Novel agents for relapsed or refractory IP-LBCL

Study design (no. of patients)Response rateLong-term efficacyReference
BTK inhibitors     
Ibrutinib Phase 1 (N = 13) ORR = 77%, CR = 38% Median PFS = 4.6 mo 102  
 Phase 2 (N = 52) ORR = 52%, CR = 19% Median PFS = 4.8 mo 103  
Tirabrutinib Phase 2 (N = 44) ORR = 64%, CR = 34% Median PFS = 2.9 mo 105  
Immunomodulatory agents     
Lenalidomide Phase 1 (N = 7) ORR = 86%, CR = 14% Not reported 110  
Lenalidomide with rituximab Phase 2 (N = 45) ORR = 36%, CR = 29% Median PFS = 7.8 mo 111  
Pomalidomide with dexamethasone Phase 1 (N = 25) ORR = 48%, CR = 32% Median PFS = 5.3 mo 112  
Immune checkpoint inhibitors     
Nivolumab Phase 2 PCNSL (N = 47) ORR = 6% Median PFS = 1.4 mo Clinicaltrials.gov [NCT02857426] 
 Phase 2 PTL (N = 19) ORR = 26% Median PFS = 1.7 mo Clinicaltrials.gov [NCT02857426] 
Pembrolizumab Phase 2 PCNSL (N = 50) ORR = 12% Median PFS = 2.6 mo 75  
CAR T-cell therapy     
Tisangenlecleucel Phase 1/2 (N = 12) ORR = 58%, CR = 50% Not reported 113  
Axicabtagene ciloleucel Phase 1 (N = 9) ORR = 86%, CR = 86% Not reported 114  
Study design (no. of patients)Response rateLong-term efficacyReference
BTK inhibitors     
Ibrutinib Phase 1 (N = 13) ORR = 77%, CR = 38% Median PFS = 4.6 mo 102  
 Phase 2 (N = 52) ORR = 52%, CR = 19% Median PFS = 4.8 mo 103  
Tirabrutinib Phase 2 (N = 44) ORR = 64%, CR = 34% Median PFS = 2.9 mo 105  
Immunomodulatory agents     
Lenalidomide Phase 1 (N = 7) ORR = 86%, CR = 14% Not reported 110  
Lenalidomide with rituximab Phase 2 (N = 45) ORR = 36%, CR = 29% Median PFS = 7.8 mo 111  
Pomalidomide with dexamethasone Phase 1 (N = 25) ORR = 48%, CR = 32% Median PFS = 5.3 mo 112  
Immune checkpoint inhibitors     
Nivolumab Phase 2 PCNSL (N = 47) ORR = 6% Median PFS = 1.4 mo Clinicaltrials.gov [NCT02857426] 
 Phase 2 PTL (N = 19) ORR = 26% Median PFS = 1.7 mo Clinicaltrials.gov [NCT02857426] 
Pembrolizumab Phase 2 PCNSL (N = 50) ORR = 12% Median PFS = 2.6 mo 75  
CAR T-cell therapy     
Tisangenlecleucel Phase 1/2 (N = 12) ORR = 58%, CR = 50% Not reported 113  
Axicabtagene ciloleucel Phase 1 (N = 9) ORR = 86%, CR = 86% Not reported 114  

CR, complete response rate; ORR, overall response rate.

Treatment of PCNSL

Treatment of PCNSL differs significantly from the treatment of systemic DLBCL because standard anthracycline-based chemotherapy regimens do not reliably cross the blood–brain barrier and the prognosis is worse. As such, high-dose methotrexate (HD-MTX) chemotherapy regimens form the cornerstone of therapy.82-85 The rate of complete response to standard induction regimens is only ∼50%, so most patients are offered postremission consolidation. Younger patients who are deemed suitable for dose-intensive therapy are often treated with high-dose chemotherapy with autologous stem cell transplantation.86,87 A recent randomized phase 2 study of 140 patients aged <60 years showed that thiotepa-based conditioning followed by autologous stem cell transplantation resulted in an 8-year progression-free survival (PFS) of 67% and was a superior consolidation strategy than whole-brain radiotherapy.83 Older patients and those not deemed suitable for dose-intensive therapy have a much poorer prognosis because few alternative therapies exist with reliable penetration across the blood–brain barrier.88-91 Furthermore, the actual rate of cure may not be captured from clinical trials because late recurrences are common.16,92 Few effective salvage therapies exist, and those with chemotherapy-refractory disease have a grave prognosis.93 

PVRL

PVRL has an indolent course and systemic therapy is generally not indicated at diagnosis. Clinical management incorporates localized therapy to prevent progressive vision loss but this does not prevent CNS relapse.94 Treatment algorithms derive mostly from small retrospective series leading to significant variance in practice patterns.95 In situations of unilateral involvement, localized therapy consists of intravitreal injections of either MTX or rituximab.96,97 An alternative local therapy is radiotherapy, but this risks inducing retinopathy and does not prevent CNS relapse. Bilateral involvement leads to consideration of more intensive therapy such as HD-MTX, but the benefit of this approach is unknown. HD-MTX has been used after intravitreal MTX to prevent CNS relapse, but the benefit to prevention of CNS progression remains unproven. Prospective studies testing the role of novel targeted agents and immunotherapy are needed.

Treatment of PTL

Orchiectomy is required for the localized management of PTL but is not definitive therapy and systemic therapy is indicated even with limited stage disease because of the high risk of systemic relapse.34,35 Standard chemotherapy regimens for DLBCL such as rituximab plus CHOP (R-CHOP) should be given and the benefit of anthracycline-based chemotherapy has been shown.35 The management of patients with PTL includes measures to prevent CNS relapse, and a recent prospective study demonstrated that intensive CNS prophylaxis including both intrathecal and intravenous methods was feasible and no CNS relapses were observed.37 Contralateral testis irradiation is administered to prevent local recurrence.37 

A common source of treatment failure for PTL is related to the risk of CNS relapse, which occurs in 15% to 30% of patients.38 Prospective studies have included intrathecal MTX as CNS prophylaxis and the rate of CNS progression was only 6%.98 Other studies have used HD-MTX as CNS prophylaxis, which may be a suitable option in some patients.99 A recent prospective study demonstrated that intensive CNS prophylaxis including both intrathecal and intravenous methods was feasible and no CNS relapses were observed.37 Involvement of the contralateral testicle occurs in up to 10% of cases, which should prompt the use of testicular radiation. Retrospective series have suggested that contralateral scrotal radiation is effective for reducing the risk of local recurrence and this strategy is recommended.35,37 Hypogonadism is a risk of this approach and testosterone replacement should be considered.

Treatment of IVLBCL

Given the challenges of accurate staging of IVLBCL, it should be considered as disseminated disease in all cases. Anthracycline-based chemotherapy regimens are the mainstay of treatment, with special attention given to the risk of CNS involvement and progression. As in IP-LBCL, the risk of CNS progression in IVLBCL is sufficiently high to warrant consideration of CNS prophylaxis as part of frontline management. A prospective phase 2 study of 38 patients with IVLBCL and no CNS involvement tested R-CHOP every 14 days with intrathecal chemotherapy alternating with rituximab plus HD-MTX as an intensive strategy to prevent CNS disease suggested that CNS prophylaxis may reduce the risk of CNS recurrence in IVLBCL.100 

Novel treatment approaches for IP-LBCL

The fundamental question is whether an improved understanding of the genetic profile of IP-LBCL can be translated to improved clinical outcomes. Multiple rational targeted agents and immunotherapy approaches have been tested in PCNSL and PVRL and multiple combinations are being tested.

Ibrutinib is a first-generation BTK inhibitor that interferes with chronic active BCR signaling and initially demonstrated selective activity in relapsed or refractory DLBCL tumors that harbor both MYD88L265P and CD79B mutations.60 BTK was thereby identified as highly rational therapeutic target in PCNSL because this genetic profile is characteristic. Indeed, early clinical studies demonstrated very high response rates to ibrutinib monotherapy,101-103 but resistance developed rapidly to monotherapy,104 which results in a median PFS of only 3 to 5 months.102,103,105 To overcome the problem of acquired resistance, ibrutinib has been added to chemotherapy including HD-MTX with rituximab106 and anthracycline-based chemotherapy regimens.101 This strategy resulted in significantly higher rates of complete response, and many combination regimens using ibrutinib as well as second generation BTK inhibitors including zanubrutinib, acalabrutinib, tirabrutinib, and orelabrutinib are now being tested in ongoing clinical trials.

Understanding the potential role of BTK inhibitors across IP-LBCL subtypes is more difficult because PTL is uncommon and related entities such as IVLBCL are both uncommon and frequently excluded from clinical trials that require measurable disease. The most compelling data supporting a possible role of BTK inhibitors for IP-LBCL comes from a randomized phase 3 study testing frontline therapy of R-CHOP with ibrutinib compared with R-CHOP with placebo in non-GCB DLBCL.107 This study did not meet its primary end point and no benefit was observed with the use of ibrutinib across the entire study population. Intriguingly, patients aged <60 years had improvements in both PFS and overall survival, and a subsequent translational study demonstrated that the genetic subtypes MCD and N1 had survival of 100% when treated with ibrutinib plus R-CHOP.76 

Other novel targeted agents that are rational for PCNSL and PVRL are the immunomodulatory drugs, lenalidomide and pomalidomide, which downregulate the transcription factor interferon regulatory factor 4 and augment interferon β production to kill ABC DLBCL cell lines.108,109 Both lenalidomide and pomalidomide have been tested as combination therapy for relapsed or refractory PCNSL and PVRL.110-112 These agents show clinical activity and may improve outcomes as part of combination therapy, many of which are being tested in ongoing studies.

Novel immunotherapy approaches including chimeric antigen receptor (CAR) T-cell therapies targeting CD19 are effective in chemotherapy-refractory systemic DLBCL and have been explored in PCNSL.113,114 These early studies have demonstrated that effector CAR T cells successfully cross the blood–brain barrier and are clinically active in CNS tumors. A summary of 128 patients with CNS lymphoma treated with CAR T-cell therapy showed acceptable safety and suggested that the rates of complete response compare favorably with those in systemic DLBCL.115 Immune checkpoint inhibitors targeting PD-1 showed early promise in retrospective studies for PCNSL and PTL,73 but prospective studies have been disappointing.74,75 

The umbrella term IP-LBCL includes cases of DLBCL that involve the CNS, vitreous, and testis, that share unique clinical features including a strong predilection to involve extranodal sites, including the CNS, and are commonly of the MCD/C5/MYD88 genetic subtype. The relevance of their origins within immune-privileged sites is unclear and other entities also appear closely related as extranodal ABC lymphomas, particularly IVLBCL. The recognition of IP-LBCL as a distinct group of tumors allows for the investigation of therapeutic vulnerabilities and clinical trials can report results within the context of these subtypes.

The authors thank patients and their families who participate in clinical trials testing novel agents in aggressive B-cell lymphomas. Ethan Tyler is acknowledged for illustration support.

This work is supported by the Intramural Research Program of the NIH: National Cancer Institute, Center for Cancer Research.

Contribution: M.R wrote the first version of this manuscript with input from J.D.P. and E.S.J., who edited the final version of the manuscript.

Conflict-of interest disclosure: The authors declare no competing financial interests.

Correspondence: Mark Roschewski, National Cancer Institute, National Institutes of Health, Lymphoid Malignancies Branch, CCR 9000 Rockville Pike, Bldg 10, Room 12c442, 10 Center Dr, Bethesda, MD 20892; email: mark.roschewski@nih.gov.

1.
Schmitz
R
,
Wright
GW
,
Huang
DW
, et al
.
Genetics and pathogenesis of diffuse large B-cell lymphoma
.
N Engl J Med
.
2018
;
378
(
15
):
1396
-
1407
.
2.
Wright
GW
,
Huang
DW
,
Phelan
JD
, et al
.
A probabilistic classification tool for genetic subtypes of diffuse large B cell lymphoma with therapeutic implications
.
Cancer Cell
.
2020
;
37
(
4
):
551
-
568.e14
.
3.
Chapuy
B
,
Stewart
C
,
Dunford
AJ
, et al
.
Molecular subtypes of diffuse large B cell lymphoma are associated with distinct pathogenic mechanisms and outcomes
.
Nat Med
.
2018
;
24
(
5
):
679
-
690
.
4.
Lacy
SE
,
Barrans
SL
,
Beer
PA
, et al
.
Targeted sequencing in DLBCL, molecular subtypes, and outcomes: a Haematological Malignancy Research Network report
.
Blood
.
2020
;
135
(
20
):
1759
-
1771
.
5.
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
(
11
):
1229
-
1253
.
6.
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
(
7
):
1720
-
1748
.
7.
Chapuy
B
,
Roemer
MG
,
Stewart
C
, et al
.
Targetable genetic features of primary testicular and primary central nervous system lymphomas
.
Blood
.
2016
;
127
(
7
):
869
-
881
.
8.
Minderman
M
,
Amir
A
,
Kraan
W
, et al
.
Immune evasion in primary testicular and central nervous system lymphomas: HLA loss rather than 9p24.1/PD-L1/PD-L2 alterations
.
Blood
.
2021
;
138
(
13
):
1194
-
1197
.
9.
Radke
J
,
Ishaque
N
,
Koll
R
, et al
.
The genomic and transcriptional landscape of primary central nervous system lymphoma
.
Nat Commun
.
2022
;
13
(
1
):
2558
.
10.
Elfrink
S
,
de Winde
CM
,
van den Brand
M
, et al
.
High frequency of inactivating tetraspanin C D37 mutations in diffuse large B-cell lymphoma at immune-privileged sites
.
Blood
.
2019
;
134
(
12
):
946
-
950
.
11.
Bonzheim
I
,
Sander
P
,
Salmeron-Villalobos
J
, et al
.
The molecular hallmarks of primary and secondary vitreoretinal lymphoma
.
Blood Adv
.
2022
;
6
(
5
):
1598
-
1607
.
12.
Gandhi
MK
,
Hoang
T
,
Law
SC
, et al
.
EBV-associated primary CNS lymphoma occurring after immunosuppression is a distinct immunobiological entity
.
Blood
.
2021
;
137
(
11
):
1468
-
1477
.
13.
Guney
E
,
Lucas
CHG
,
Singh
K
, et al
.
Molecular profiling identifies at least 3 distinct types of posttransplant lymphoproliferative disorder involving the CNS
.
Blood Adv
.
2023
;
7
(
13
):
3307
-
3311
.
14.
Los-de Vries
GT
,
Stathi
P
,
Rutkens
R
, et al
.
Large B-cell lymphomas of immune-privileged sites relapse via parallel clonal evolution from a common progenitor B cell
.
Cancer Res
.
2023
;
83
(
11
):
1917
-
1927
.
15.
Fonseca
R
,
Habermann
TM
,
Colgan
JP
, et al
.
Testicular lymphoma is associated with a high incidence of extranodal recurrence
.
Cancer
.
2000
;
88
(
1
):
154
-
161
.
16.
Ambady
P
,
Holdhoff
M
,
Bonekamp
D
,
Wong
F
,
Grossman
SA
.
Late relapses in primary CNS lymphoma after complete remissions with high-dose methotrexate monotherapy
.
CNS Oncol
.
2015
;
4
(
6
):
393
-
398
.
17.
Barker
CF
,
Billingham
RE
.
Immunologically privileged sites
.
Adv Immunol
.
1977
;
25
:
1
-
54
.
18.
Schaff
LR
,
Grommes
C
.
Primary central nervous system lymphoma
.
Blood
.
2022
;
140
(
9
):
971
-
979
.
19.
Ferreri
AJM
,
Calimeri
T
,
Cwynarski
K
, et al
.
Primary central nervous system lymphoma
.
Nat Rev Dis Primers
.
2023
;
9
(
1
):
1
-
29
.
20.
Ostrom
QT
,
Price
M
,
Neff
C
, et al
.
CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2015-2019
.
Neuro Oncol
.
2022
;
24
(
suppl 5
):
v1
-
v95
.
21.
Villano
JL
,
Koshy
M
,
Shaikh
H
,
Dolecek
TA
,
McCarthy
BJ
.
Age, gender, and racial differences in incidence and survival in primary CNS lymphoma
.
Br J Cancer
.
2011
;
105
(
9
):
1414
-
1418
.
22.
Mendez
JS
,
Ostrom
QT
,
Gittleman
H
, et al
.
The elderly left behind-changes in survival trends of primary central nervous system lymphoma over the past 4 decades
.
Neuro Oncol
.
2018
;
20
(
5
):
687
-
694
.
23.
Shiels
MS
,
Pfeiffer
RM
,
Besson
C
, et al
.
Trends in primary central nervous system lymphoma incidence and survival in the U.S
.
Br J Haematol
.
2016
;
174
(
3
):
417
-
424
.
24.
Jahnke
K
,
Thiel
E
,
Martus
P
, et al
.
Relapse of primary central nervous system lymphoma: clinical features, outcome and prognostic factors
.
J Neuro Oncol
.
2006
;
80
(
2
):
159
-
165
.
25.
Bromberg
JE
,
Siemers
MD
,
Taphoorn
MJ
.
Is a "vanishing tumor" always a lymphoma?
.
Neurology
.
2002
;
59
(
5
):
762
-
764
.
26.
Ferreri
AJM
,
Calimeri
T
,
Lopedote
P
, et al
.
MYD88 L265P mutation and interleukin-10 detection in cerebrospinal fluid are highly specific discriminating markers in patients with primary central nervous system lymphoma: results from a prospective study
.
Br J Haematol
.
2021
;
193
(
3
):
497
-
505
.
27.
Mutter
JA
,
Alig
SK
,
Esfahani
MS
, et al
.
Circulating tumor DNA profiling for detection, risk stratification, and classification of brain lymphomas
.
J Clin Oncol
.
2023
;
41
(
9
):
1684
-
1694
.
28.
Abrey
LE
,
Batchelor
TT
,
Ferreri
AJ
, et al
.
Report of an international workshop to standardize baseline evaluation and response criteria for primary CNS lymphoma
.
J Clin Oncol
.
2005
;
23
(
22
):
5034
-
5043
.
29.
Grimm
SA
,
Pulido
JS
,
Jahnke
K
, et al
.
Primary intraocular lymphoma: an International Primary Central Nervous System Lymphoma Collaborative Group Report
.
Ann Oncol
.
2007
;
18
(
11
):
1851
-
1855
.
30.
Fisson
S
,
Ouakrim
H
,
Touitou
V
, et al
.
Cytokine profile in human eyes: contribution of a new cytokine combination for differential diagnosis between intraocular lymphoma or uveitis
.
PLoS One
.
2013
;
8
(
2
):
e52385
.
31.
Bonzheim
I
,
Giese
S
,
Deuter
C
, et al
.
High frequency of MYD88 mutations in vitreoretinal B-cell lymphoma: a valuable tool to improve diagnostic yield of vitreous aspirates
.
Blood
.
2015
;
126
(
1
):
76
-
79
.
32.
Karakawa
A
,
Taoka
K
,
Kaburaki
T
, et al
.
Clinical features and outcomes of secondary intraocular lymphoma
.
Br J Haematol
.
2018
;
183
(
4
):
668
-
671
.
33.
Gundrum
JD
,
Mathiason
MA
,
Moore
DB
,
Go
RS
.
Primary testicular diffuse large B-cell lymphoma: a population-based study on the incidence, natural history, and survival comparison with primary nodal counterpart before and after the introduction of rituximab
.
J Clin Oncol
.
2009
;
27
(
31
):
5227
-
5232
.
34.
Vitolo
U
,
Ferreri
AJ
,
Zucca
E
.
Primary testicular lymphoma
.
Crit Rev Oncol Hematol
.
2008
;
65
(
2
):
183
-
189
.
35.
Zucca
E
,
Conconi
A
,
Mughal
TI
, et al
.
Patterns of outcome and prognostic factors in primary large-cell lymphoma of the testis in a survey by the International Extranodal Lymphoma Study Group
.
J Clin Oncol
.
2003
;
21
(
1
):
20
-
27
.
36.
Deng
L
,
Xu-Monette
ZY
,
Loghavi
S
, et al
.
Primary testicular diffuse large B-cell lymphoma displays distinct clinical and biological features for treatment failure in rituximab era: a report from the International PTL Consortium
.
Leukemia
.
2016
;
30
(
2
):
361
-
372
.
37.
Conconi
A
,
Chiappella
A
,
Ferreri
AJ
, et al
.
IELSG30 phase 2 trial: intravenous and intrathecal CNS prophylaxis in primary testicular diffuse large B-cell lymphoma
.
Blood Adv
.
2024
;
8
(
6
):
1541
-
1549
.
38.
Kridel
R
,
Telio
D
,
Villa
D
, et al
.
Diffuse large B-cell lymphoma with testicular involvement: outcome and risk of CNS relapse in the rituximab era
.
Br J Haematol
.
2017
;
176
(
2
):
210
-
221
.
39.
Twa
DDW
,
Lee
DG
,
Tan
KL
, et al
.
Genomic predictors of central nervous system relapse in primary testicular diffuse large B-cell lymphoma
.
Blood
.
2021
;
137
(
9
):
1256
-
1259
.
40.
Ponzoni
M
,
Ferreri
AJ
,
Campo
E
, et al
.
Definition, diagnosis, and management of intravascular large B-cell lymphoma: proposals and perspectives from an international consensus meeting
.
J Clin Oncol
.
2007
;
25
(
21
):
3168
-
3173
.
41.
Ponzoni
M
,
Campo
E
,
Nakamura
S
.
Intravascular large B-cell lymphoma: a chameleon with multiple faces and many masks
.
Blood
.
2018
;
132
(
15
):
1561
-
1567
.
42.
Gonzalez-Farre
B
,
Ramis-Zaldivar
JE
,
Castrejon de Anta
N
, et al
.
Intravascular large B-cell lymphoma genomic profile is characterized by alterations in genes regulating NF-kappaB and immune checkpoints
.
Am J Surg Pathol
.
2023
;
47
(
2
):
202
-
211
.
43.
Shimada
K
,
Kinoshita
T
,
Naoe
T
,
Nakamura
S
.
Presentation and management of intravascular large B-cell lymphoma
.
Lancet Oncol
.
2009
;
10
(
9
):
895
-
902
.
44.
MacGillivary
ML
,
Purdy
KS
.
Recommendations for an approach to random skin biopsy in the diagnosis of intravascular B-cell lymphoma
.
J Cutan Med Surg
.
2023
;
27
(
1
):
44
-
50
.
45.
Ferreri
AJ
,
Campo
E
,
Seymour
JF
, et al
.
Intravascular lymphoma: clinical presentation, natural history, management and prognostic factors in a series of 38 cases, with special emphasis on the 'cutaneous variant'
.
Br J Haematol
.
2004
;
127
(
2
):
173
-
183
.
46.
Murase
T
,
Nakamura
S
,
Kawauchi
K
, et al
.
An Asian variant of intravascular large B-cell lymphoma: clinical, pathological and cytogenetic approaches to diffuse large B-cell lymphoma associated with haemophagocytic syndrome
.
Br J Haematol
.
2000
;
111
(
3
):
826
-
834
.
47.
Shimada
K
,
Yoshida
K
,
Suzuki
Y
, et al
.
Frequent genetic alterations in immune checkpoint-related genes in intravascular large B-cell lymphoma
.
Blood
.
2021
;
137
(
11
):
1491
-
1502
.
48.
Alizadeh
AA
,
Eisen
MB
,
Davis
RE
, et al
.
Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling
.
Nature
.
2000
;
403
(
6769
):
503
-
511
.
49.
Davis
RE
,
Brown
KD
,
Siebenlist
U
,
Staudt
LM
.
Constitutive nuclear factor kappaB activity is required for survival of activated B cell-like diffuse large B cell lymphoma cells
.
J Exp Med
.
2001
;
194
(
12
):
1861
-
1874
.
50.
Wright
G
,
Tan
B
,
Rosenwald
A
,
Hurt
EH
,
Wiestner
A
,
Staudt
LM
.
A gene expression-based method to diagnose clinically distinct subgroups of diffuse large B cell lymphoma
.
Proc Natl Acad Sci U S A
.
2003
;
100
(
17
):
9991
-
9996
.
51.
Rosenwald
A
,
Wright
G
,
Chan
WC
, et al
.
The use of molecular profiling to predict survival after chemotherapy for diffuse large-B-cell lymphoma
.
N Engl J Med
.
2002
;
346
(
25
):
1937
-
1947
.
52.
Scott
DW
,
Mottok
A
,
Ennishi
D
, et al
.
Prognostic significance of diffuse large B-cell lymphoma cell of origin determined by digital gene expression in formalin-fixed paraffin-embedded tissue biopsies
.
J Clin Oncol
.
2015
;
33
(
26
):
2848
-
2856
.
53.
Scott
DW
,
Wright
GW
,
Williams
PM
, et al
.
Determining cell-of-origin subtypes of diffuse large B-cell lymphoma using gene expression in formalin-fixed paraffin-embedded tissue
.
Blood
.
2014
;
123
(
8
):
1214
-
1217
.
54.
Ahmed
S
,
Glover
P
,
Taylor
J
, et al
.
Comparative analysis of gene expression platforms for cell-of-origin classification of diffuse large B-cell lymphoma shows high concordance
.
Br J Haematol
.
2021
;
192
(
3
):
599
-
604
.
55.
Ngo
VN
,
Young
RM
,
Schmitz
R
, et al
.
Oncogenically active MYD88 mutations in human lymphoma
.
Nature
.
2011
;
470
(
7332
):
115
-
119
.
56.
Davis
RE
,
Ngo
VN
,
Lenz
G
, et al
.
Chronic active B-cell-receptor signalling in diffuse large B-cell lymphoma
.
Nature
.
2010
;
463
(
7277
):
88
-
92
.
57.
Morin
RD
,
Mendez-Lago
M
,
Mungall
AJ
, et al
.
Frequent mutation of histone-modifying genes in non-Hodgkin lymphoma
.
Nature
.
2011
;
476
(
7360
):
298
-
303
.
58.
Pasqualucci
L
,
Trifonov
V
,
Fabbri
G
, et al
.
Analysis of the coding genome of diffuse large B-cell lymphoma
.
Nat Genet
.
2011
;
43
(
9
):
830
-
837
.
59.
Lohr
JG
,
Stojanov
P
,
Lawrence
MS
, et al
.
Discovery and prioritization of somatic mutations in diffuse large B-cell lymphoma (DLBCL) by whole-exome sequencing
.
Proc Natl Acad Sci U S A
.
2012
;
109
(
10
):
3879
-
3884
.
60.
Wilson
WH
,
Young
RM
,
Schmitz
R
, et al
.
Targeting B cell receptor signaling with ibrutinib in diffuse large B cell lymphoma
.
Nat Med
.
2015
;
21
(
8
):
922
-
926
.
61.
Hans
CP
,
Weisenburger
DD
,
Greiner
TC
, et al
.
Confirmation of the molecular classification of diffuse large B-cell lymphoma by immunohistochemistry using a tissue microarray
.
Blood
.
2004
;
103
(
1
):
275
-
282
.
62.
Bodor
C
,
Alpar
D
,
Marosvari
D
, et al
.
Molecular subtypes and genomic profile of primary central nervous system lymphoma
.
J Neuropathol Exp Neurol
.
2020
;
79
(
2
):
176
-
183
.
63.
Gonzalez-Aguilar
A
,
Idbaih
A
,
Boisselier
B
, et al
.
Recurrent mutations of MYD88 and TBL1XR1 in primary central nervous system lymphomas
.
Clin Cancer Res
.
2012
;
18
(
19
):
5203
-
5211
.
64.
Nakamura
T
,
Tateishi
K
,
Niwa
T
, et al
.
Recurrent mutations of CD79B and MYD88 are the hallmark of primary central nervous system lymphomas
.
Neuropathol Appl Neurobiol
.
2016
;
42
(
3
):
279
-
290
.
65.
Yamada
S
,
Ishida
Y
,
Matsuno
A
,
Yamazaki
K
.
Primary diffuse large B-cell lymphomas of central nervous system exhibit remarkably high prevalence of oncogenic MYD88 and CD79B mutations
.
Leuk Lymphoma
.
2015
;
56
(
7
):
2141
-
2145
.
66.
Braggio
E
,
Van Wier
S
,
Ojha
J
, et al
.
Genome-wide analysis uncovers novel recurrent alterations in primary central nervous system lymphomas
.
Clin Cancer Res
.
2015
;
21
(
17
):
3986
-
3994
.
67.
Fukumura
K
,
Kawazu
M
,
Kojima
S
, et al
.
Genomic characterization of primary central nervous system lymphoma
.
Acta Neuropathol
.
2016
;
131
(
6
):
865
-
875
.
68.
Nayyar
N
,
White
MD
,
Gill
CM
, et al
.
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
.
69.
Green
MR
,
Monti
S
,
Rodig
SJ
, et al
.
Integrative analysis reveals selective 9p24.1 amplification, increased PD-1 ligand expression, and further induction via JAK2 in nodular sclerosing Hodgkin lymphoma and primary mediastinal large B-cell lymphoma
.
Blood
.
2010
;
116
(
17
):
3268
-
3277
.
70.
Ansell
SM
,
Lesokhin
AM
,
Borrello
I
, et al
.
PD-1 blockade with nivolumab in relapsed or refractory Hodgkin's lymphoma
.
N Engl J Med
.
2015
;
372
(
4
):
311
-
319
.
71.
Kiyasu
J
,
Miyoshi
H
,
Hirata
A
, et al
.
Expression of programmed cell death ligand 1 is associated with poor overall survival in patients with diffuse large B-cell lymphoma
.
Blood
.
2015
;
126
(
19
):
2193
-
2201
.
72.
Chen
BJ
,
Chapuy
B
,
Ouyang
J
, et al
.
PD-L1 expression is characteristic of a subset of aggressive B-cell lymphomas and virus-associated malignancies
.
Clin Cancer Res
.
2013
;
19
(
13
):
3462
-
3473
.
73.
Nayak
L
,
Iwamoto
FM
,
LaCasce
A
, et al
.
PD-1 blockade with nivolumab in relapsed/refractory primary central nervous system and testicular lymphoma
.
Blood
.
2017
;
129
(
23
):
3071
-
3073
.
74.
A study of nivolumab in relapsed/refractory primary central nervous system lymphoma (PCNSL) and relapsed/refractory primary testicular lymphoma (PTL)
. 2021. ClinicalTrials.gov identifier: NCT02857426. Updated 24 November. Accessed 19 March 2024. https://clinicaltrials.gov/study/NCT02857426.
75.
Hoang-Xuan
K
,
Houot
R
,
Soussain
C
, et al
.
First results of the Acsé Pembrolizumab phase II in the primary CNS lymphoma (PCNSL) cohort [abstract]
.
Blood
.
2020
;
136
(
suppl 1
):
15
-
16
.
76.
Wilson
WH
,
Wright
GW
,
Huang
DW
, et al
.
Effect of ibrutinib with R-CHOP chemotherapy in genetic subtypes of DLBCL
.
Cancer Cell
.
2021
;
39
(
12
):
1643
-
1653.e3
.
77.
Phelan
JD
,
Scheich
S
,
Choi
J
, et al
.
Response to Bruton's tyrosine kinase inhibitors in aggressive lymphomas linked to chronic selective autophagy
.
Cancer Cell
.
2024
;
42
(
2
):
238
-
252.e9
.
78.
Phelan
JD
,
Young
RM
,
Webster
DE
, et al
.
A multiprotein supercomplex controlling oncogenic signalling in lymphoma
.
Nature
.
2018
;
560
(
7718
):
387
-
391
.
79.
Choi
J
,
Phelan
JD
,
Wright
GW
, et al
.
Regulation of B cell receptor-dependent NF-kappaB signaling by the tumor suppressor KLHL14
.
Proc Natl Acad Sci U S A
.
2020
;
117
(
11
):
6092
-
6102
.
80.
Bobillo
S
,
Khwaja
J
,
Ferreri
AJM
,
Cwynarski
K
.
Prevention and management of secondary central nervous system lymphoma
.
Haematologica
.
2023
;
108
(
3
):
673
-
689
.
81.
Simard
J
,
Roschewski
M
.
SOHO state of the art updates and next questions: prophylaxis and management of secondary CNS lymphoma
.
Clin Lymphoma Myeloma Leuk
.
2022
;
22
(
10
):
709
-
717
.
82.
Rubenstein
JL
,
Hsi
ED
,
Johnson
JL
, et al
.
Intensive chemotherapy and immunotherapy in patients with newly diagnosed primary CNS lymphoma: CALGB 50202 (Alliance 50202)
.
J Clin Oncol
.
2013
;
31
(
25
):
3061
-
3068
.
83.
Houillier
C
,
Dureau
S
,
Taillandier
L
, et al
.
Radiotherapy or autologous stem-cell transplantation for primary CNS lymphoma in patients age 60 years and younger: long-term results of the randomized phase II PRECIS study
.
J Clin Oncol
.
2022
;
40
(
32
):
3692
-
3698
.
84.
Omuro
A
,
Correa
DD
,
DeAngelis
LM
, et al
.
R-MPV followed by high-dose chemotherapy with TBC and autologous stem-cell transplant for newly diagnosed primary CNS lymphoma
.
Blood
.
2015
;
125
(
9
):
1403
-
1410
.
85.
Ferreri
AJ
,
Cwynarski
K
,
Pulczynski
E
, et al
.
Chemoimmunotherapy with methotrexate, cytarabine, thiotepa, and rituximab (MATRix regimen) in patients with primary CNS lymphoma: results of the first randomisation of the International Extranodal Lymphoma Study Group-32 (IELSG32) phase 2 trial
.
Lancet Haematol
.
2016
;
3
(
5
):
e217
-
e227
.
86.
Colombat
P
,
Lemevel
A
,
Bertrand
P
, et al
.
High-dose chemotherapy with autologous stem cell transplantation as first-line therapy for primary CNS lymphoma in patients younger than 60 years: a multicenter phase II study of the GOELAMS group
.
Bone Marrow Transplant
.
2006
;
38
(
6
):
417
-
420
.
87.
Abrey
LE
,
Moskowitz
CH
,
Mason
WP
, et al
.
Intensive methotrexate and cytarabine followed by high-dose chemotherapy with autologous stem-cell rescue in patients with newly diagnosed primary CNS lymphoma: an intent-to-treat analysis
.
J Clin Oncol
.
2003
;
21
(
22
):
4151
-
4156
.
88.
David
KA
,
Sundaram
S
,
Kim
SH
, et al
.
Older patients with primary central nervous system lymphoma: survival and prognostication across 20 U.S. cancer centers
.
Am J Hematol
.
2023
;
98
(
6
):
900
-
912
.
89.
Pulczynski
EJ
,
Kuittinen
O
,
Erlanson
M
, et al
.
Successful change of treatment strategy in elderly patients with primary central nervous system lymphoma by de-escalating induction and introducing temozolomide maintenance: results from a phase II study by the Nordic Lymphoma Group
.
Haematologica
.
2015
;
100
(
4
):
534
-
540
.
90.
Fritsch
K
,
Kasenda
B
,
Schorb
E
, et al
.
High-dose methotrexate-based immuno-chemotherapy for elderly primary CNS lymphoma patients (PRIMAIN study)
.
Leukemia
.
2017
;
31
(
4
):
846
-
852
.
91.
Omuro
A
,
Chinot
O
,
Taillandier
L
, et al
.
Methotrexate and temozolomide versus methotrexate, procarbazine, vincristine, and cytarabine for primary CNS lymphoma in an elderly population: an intergroup ANOCEF-GOELAMS randomised phase 2 trial
.
Lancet Haematol
.
2015
;
2
(
6
):
e251
-
e259
.
92.
Biccler
JL
,
Savage
KJ
,
Brown
PDN
, et al
.
Risk of death, relapse or progression, and loss of life expectancy at different progression-free survival milestones in primary central nervous system lymphoma
.
Leuk Lymphoma
.
2019
;
60
(
10
):
2516
-
2523
.
93.
Langner-Lemercier
S
,
Houillier
C
,
Soussain
C
, et al
.
Primary CNS lymphoma at first relapse/progression: characteristics, management, and outcome of 256 patients from the French LOC network
.
Neuro Oncol
.
2016
;
18
(
9
):
1297
-
1303
.
94.
Soussain
C
,
Malaise
D
,
Cassoux
N
.
Primary vitreoretinal lymphoma: a diagnostic and management challenge
.
Blood
.
2021
;
138
(
17
):
1519
-
1534
.
95.
Riemens
A
,
Bromberg
J
,
Touitou
V
, et al
.
Treatment strategies in primary vitreoretinal lymphoma: a 17-center European collaborative study
.
JAMA Ophthalmol
.
2015
;
133
(
2
):
191
-
197
.
96.
de Smet
MD
,
Vancs
VS
,
Kohler
D
,
Solomon
D
,
Chan
CC
.
Intravitreal chemotherapy for the treatment of recurrent intraocular lymphoma
.
Br J Ophthalmol
.
1999
;
83
(
4
):
448
-
451
.
97.
Kim
H
,
Csaky
KG
,
Chan
CC
, et al
.
The pharmacokinetics of rituximab following an intravitreal injection
.
Exp Eye Res
.
2006
;
82
(
5
):
760
-
766
.
98.
Vitolo
U
,
Chiappella
A
,
Ferreri
AJ
, et al
.
First-line treatment for primary testicular diffuse large B-cell lymphoma with rituximab-CHOP, CNS prophylaxis, and contralateral testis irradiation: final results of an international phase II trial
.
J Clin Oncol
.
2011
;
29
(
20
):
2766
-
2772
.
99.
Aviles
A
,
Nambo
MJ
,
Cleto
S
,
Neri
N
,
Huerta-Guzman
J
.
Rituximab and dose-dense chemotherapy in primary testicular lymphoma
.
Clin Lymphoma Myeloma
.
2009
;
9
(
5
):
386
-
389
.
100.
Shimada
K
,
Yamaguchi
M
,
Atsuta
Y
, et al
.
Rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisolone combined with high-dose methotrexate plus intrathecal chemotherapy for newly diagnosed intravascular large B-cell lymphoma (PRIMEUR-IVL): a multicentre, single-arm, phase 2 trial
.
Lancet Oncol
.
2020
;
21
(
4
):
593
-
602
.
101.
Lionakis
MS
,
Dunleavy
K
,
Roschewski
M
, et al
.
Inhibition of B cell receptor signaling by ibrutinib in primary CNS lymphoma
.
Cancer Cell
.
2017
;
31
(
6
):
833
-
843.e5
.
102.
Grommes
C
,
Pastore
A
,
Palaskas
N
, et al
.
Ibrutinib unmasks critical role of Bruton tyrosine kinase in primary CNS lymphoma
.
Cancer Discov
.
2017
;
7
(
9
):
1018
-
1029
.
103.
Soussain
C
,
Choquet
S
,
Blonski
M
, et al
.
Ibrutinib monotherapy for relapse or refractory primary CNS lymphoma and primary vitreoretinal lymphoma: final analysis of the phase II 'proof-of-concept' iLOC study by the Lymphoma study association (LYSA) and the French oculo-cerebral lymphoma (LOC) network
.
Eur J Cancer
.
2019
;
117
:
121
-
130
.
104.
Shaffer
AL
,
Phelan
JD
,
Wang
JQ
, et al
.
Overcoming acquired epigenetic resistance to BTK inhibitors
.
Blood Cancer Discov
.
2021
;
2
(
6
):
630
-
647
.
105.
Narita
Y
,
Nagane
M
,
Mishima
K
, et al
.
Phase I/II study of tirabrutinib, a second-generation Bruton's tyrosine kinase inhibitor, in relapsed/refractory primary central nervous system lymphoma
.
Neuro Oncol
.
2021
;
23
(
1
):
122
-
133
.
106.
Grommes
C
,
Tang
SS
,
Wolfe
J
, et al
.
Phase 1b trial of an ibrutinib-based combination therapy in recurrent/refractory CNS lymphoma
.
Blood
.
2019
;
133
(
5
):
436
-
445
.
107.
Younes
A
,
Sehn
LH
,
Johnson
P
, et al
.
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
.
108.
Tageja
N
.
Lenalidomide - current understanding of mechanistic properties
.
Anti Cancer Agents Med Chem
.
2011
;
11
(
3
):
315
-
326
.
109.
Zhang
LH
,
Kosek
J
,
Wang
M
,
Heise
C
,
Schafer
PH
,
Chopra
R
.
Lenalidomide efficacy in activated B-cell-like subtype diffuse large B-cell lymphoma is dependent upon IRF4 and cereblon expression
.
Br J Haematol
.
2013
;
160
(
4
):
487
-
502
.
110.
Rubenstein
JL
,
Geng
H
,
Fraser
EJ
, et al
.
Phase 1 investigation of lenalidomide/rituximab plus outcomes of lenalidomide maintenance in relapsed CNS lymphoma
.
Blood Adv
.
2018
;
2
(
13
):
1595
-
1607
.
111.
Ghesquieres
H
,
Chevrier
M
,
Laadhari
M
, et al
.
Lenalidomide in combination with intravenous rituximab (REVRI) in relapsed/refractory primary CNS lymphoma or primary intraocular lymphoma: a multicenter prospective 'proof of concept' phase II study of the French Oculo-Cerebral lymphoma (LOC) Network and the Lymphoma Study Association (LYSA)dagger
.
Ann Oncol
.
2019
;
30
(
4
):
621
-
628
.
112.
Tun
HW
,
Johnston
PB
,
DeAngelis
LM
, et al
.
Phase 1 study of pomalidomide and dexamethasone for relapsed/refractory primary CNS or vitreoretinal lymphoma
.
Blood
.
2018
;
132
(
21
):
2240
-
2248
.
113.
Frigault
MJ
,
Dietrich
J
,
Gallagher
K
, et al
.
Safety and efficacy of tisagenlecleucel in primary CNS lymphoma: a phase 1/2 clinical trial
.
Blood
.
2022
;
139
(
15
):
2306
-
2315
.
114.
Jacobson
CA
,
Falvey
C
,
Bouvier
R
, et al
.
A pilot study of axicabtagene ciloleucel (axi-cel) for the treatment of relapsed/refractory primary and secondary central nervous system lymphoma (CNSL) [abstract]
.
Blood
.
2022
;
140
(
suppl 1
):
1060
-
1061
.
115.
Cook
MR
,
Dorris
CS
,
Makambi
KH
, et al
.
Toxicity and efficacy of CAR T-cell therapy in PCNSL and SCNSL: a meta-analysis of 128 patients
.
Blood Adv
.
2023
;
7
(
1
):
32
-
39
.
Sign in via your Institution