Sie sind auf Seite 1von 16

guideline

Guidelines for the diagnosis and management of primary


central nervous system diffuse large B-cell lymphoma

Christopher P. Fox,1 Elizabeth H. Phillips,2 Jeffery Smith,3 Kim Linton,4 Eve Gallop-Evans,5 Claire Hemmaway,6
Dorothee P. Auer, Charlotte Fuller, Andrew J. Davies,9 Pamela McKay,10
7 8
Kate Cwynarski2 and on behalf of the British
Society for Haematology
1
Clinical Haematology, Nottingham University Hospitals NHS Trust, Nottingham, 2Department of Haematology, University College
Hospital, London, 3Department of Haematology, Division of Cancer Sciences, School of Medical Sciences, Aintree Hospital NHS Trust,
Liverpool, 4University of Manchester, Manchester, 5Velindre Cancer Centre, Cardiff, 6Department of Haematology, Barking, Havering
and Redbridge University Hospitals, Essex, 7Division of Clinical Neuroscience, Radiological Sciences, University of Nottingham, Not-
tingham, 8Department of Neuropsychology, Barking, Havering and Redbridge University Hospitals, Essex, 9Department of Medical
Oncology, Southampton General Hospital, Southampton, and 10Department of Haematology, Beatson West of Scotland Cancer Centre,
Glasgow, UK

Keywords: primary CNS lymphoma, diffuse large B- and Evaluation (GRADE) nomenclature was used to assess levels
cell lymphoma, intraocular lymphoma, diagnosis, of evidence and assess the strength of recommendations (http://
treatment, staging. www.gradeworkinggroups.org).
Review of the manuscript was performed by the BSH
Guidelines Committee Haemato-Oncology Task Force, the
BSH Guidelines Committee and the Haemato-Oncology
Scope sounding board of BSH. It was posted on the members sec-
This guideline is aimed at providing healthcare professionals tion of the BSH website for comment. It has also been
with clear guidance on the diagnosis and management of pri- reviewed by the patient-focused UK charity ‘Lymphoma
mary central nervous system lymphoma (PCNSL), defined as Action’; although these organisations do not necessarily
diffuse large B-cell lymphoma (DLBCL) solely confined to approve or endorse the contents.
the central nervous system (CNS): brain, spinal cord, cranial
nerves, eyes and meninges. Secondary CNS lymphoma, Context
immunodeficiency-associated lymphoma and rare forms of
non-DLBCL CNS lymphoma are outside the scope of this Primary central nervous system lymphoma accounts for 1%
guideline. It is not the intention of this guideline to provide of all non-Hodgkin lymphomas and 3% of all brain tumours
treatment recommendations for all situations and clinicians (Swerdlow et al, 2008; Rigau et al, 2011). It is essential to
are advised to take individual patient circumstances into minimise diagnostic delay, and clinical management requires
account when making management decisions. multidisciplinary team (MDT) support. This includes hae-
mato-oncology but, unique to haematological cancers, also
requires input from specialist neurological services.
Methodology Methotrexate-based protocols should only be delivered at
Recommendations included a systematic review of published centres experienced in intensive chemotherapy.
English language literature from publication of previous British
Society for Haematology (BSH) PCNSL guidance (1 January Diagnosis and imaging
2007) up to 29 May 2017. MEDLINE, EMBASE, Cochrane data-
bases and Web of Science were searched using the preliminary Primary central nervous system lymphoma presents with a
search terms ‘CNS lymphoma’ and ‘intraocular lymphoma’. range of symptoms and signs including behavioural change,
The Grading of Recommendations Assessment, Development memory and language impairment, focal motor deficits, sei-
zures, raised intracranial pressure, uveitis and neuropsychi-
atric symptoms (Zhang et al, 2010; Aki et al, 2013).
The diagnosis should, in all cases, be confirmed by special-
Correspondence: BSH Administrator, British Society for ist haematopathology review of sampled tumour tissue or
Haematology, 100 White Lion Street, London N1 9PF, UK. fluid according to the current World Health Organization
E-mail: bshguidelines@b-s-h.org.uk classification (Swerdlow et al, 2008, 2016). Stereotactic

First published online 23 November 2018 ª 2018 British Society for Haematology and John Wiley & Sons Ltd
doi: 10.1111/bjh.15661 British Journal of Haematology, 2019, 184, 348–363
Guideline

biopsy is recommended for intracerebral lesions with intra- dissemination occurs in around 16% of cases as judged by
operative rapid cytology and review of frozen sections to cytology (Korfel et al, 2012); it can be difficult to detect on
avoid unnecessary extensive surgery (Abrey et al, 2005). MRI (Tang et al, 2011).
Cerebrospinal fluid (CSF) cytology and flow cytometry may All patients should undergo cross-sectional imaging to
be used in cases where a biopsy is not possible or to investi- exclude systemic disease. 18FDG-PET combined with com-
gate for leptomeningeal involvement, although sensitivity is puted tomography (CT) imaging (PET-CT) is a sensitive
low (Schroers et al, 2010). At least 3–10 ml of CSF should screening tool for systemic involvement at the time of diagno-
be taken (Ferreri et al, 2004) and rapidly analysed (Patrick & sis (Mohile et al, 2008). A bone marrow biopsy is not essential
Mohile, 2015). The identification of clonal IGH rearrange- in the context of typical histology if PET-CT has excluded sys-
ments by polymerase chain reaction may improve diagnostic temic disease, full blood count parameters are normal and
yield (Ekstein et al, 2006; Langerak et al, 2012). there is no detectable serum monoclonal protein to suggest a
Corticosteroids should, wherever possible, be avoided concurrent low-grade lymphoma. There is insufficient evi-
prior to biopsy as they have a substantial negative impact on dence that PET-CT has sufficient sensitivity to exclude testicu-
diagnosis; non-diagnostic rates range from 33% after a short lar involvement therefore testicular ultrasound should be
course (<1 week) to 57% after a longer course of steroids performed in men. Serum lactate dehydrogenase, performance
(Manoj et al, 2014). If corticosteroids have been adminis- status and CSF protein levels form part of established prognos-
tered, but an enhancing lesion is still present, they should be tic scores (see Appendix S1) and should be measured, where
discontinued prior to urgent biopsy (Patrick & Mohile, feasible (Ferreri et al, 2003). The International PCNSL Collab-
2015). If a suspected PCNSL lesion resolves following steroid orative Group (IPCG) guidelines on standardised evaluation
administration, re-imaging by magnetic resonance imaging of patients with newly diagnosed or suspected PCNSL (Abrey
(MRI) should be performed after a short interval (e.g. 2– et al, 2005) are summarised in Table I.
4 weeks) following steroid cessation. Close clinical follow-up
and serial imaging thereafter is recommended, at a frequency
Recommendations
guided by MDT advice, with urgent biopsy at lesion
regrowth. 1 Patients with suspected PCNSL should be discussed
Up to 20% of PCNSL patients have intraocular involve- with a PCNSL specialist early in their pathway to min-
ment (Coupland et al, 2004), which may resemble chronic imise delays. (1C)
uveitis (Coupland et al, 2004; Chan & Sen, 2013). Diagnosis 2 A histological or cytological diagnosis is required to
can be difficult and imaging alone cannot reliably establish confirm PCNSL; MRI findings alone are insufficient.
ocular involvement (Haldorsen et al, 2011). It is recom- The diagnosis should always be confirmed by specialist
mended that slit lamp examination and ophthalmoscopy are haematopathology review. (1B)
followed, if necessary, by vitreous biopsy. Vitreous biopsy 3 Corticosteroids should be avoided prior to biopsy. (1A)
should be combined with a sub-retinal aspirate or chorioreti-
a Where steroids have been administered and an
nal biopsy, particularly for those with visible sub-retinal
enhancing lesion is still present, steroids should be
deposits, as vitrectomy specimens have diagnostic failure
discontinued prior to urgent biopsy to improve
rates up to 30% (Coupland et al, 2004).
diagnostic yield. (1B)
Contrast-enhanced MRI of the brain is the neuroimaging
b If a suspected PCNSL lesion resolves following ster-
modality of choice for both diagnosis and response assess-
oid administration, re-imaging with MRI should be
ment (Coulon et al, 2002; Abrey et al, 2005; Ferreri, 2011). A
performed after a short interval with a view to
diagnosis of PCNSL cannot be presumed on radiological
urgent biopsy at lesion regrowth. (1B)
appearance and/or clinical features (such as responsiveness to
corticosteroids) alone, as neurosarcoidosis, multiple sclerosis, 4 Stereotactic biopsy of a brain lesion is the recommended
glioblastoma and vasculitis can mimic disease features (Zaki approach for histological diagnosis. Intra-operative rapid
et al, 2004; Abrey et al, 2005). PCNSL can also manifest with diagnosis using cytology and frozen sections is recom-
atypical features, including variable contrast enhancement or mended to avoid unnecessary surgical resection. (1B)
diffusion characteristics, absence of focal masses or presence 5 Vitreous biopsy should ideally be combined with a sub-
of necrosis (Tang et al, 2011). Use of additional imaging retinal aspirate or chorioretinal biopsy to establish a
studies, such as 18F deoxyglucose positron emission tomogra- diagnosis of primary intraocular lymphoma (PIOL). (1B)
phy (18FDG-PET), or advanced MRI parameters, such as per- 6 Where biopsy is not possible, a diagnosis of PCNSL
fusion metrics, diffusion coefficients, MR spectroscopy and may be supported by the combined assessment of
novel contrasts, may have the potential to improve the dis- characteristic MRI findings, clinical features AND
tinction between PCNSL and glioblastoma but none has demonstration of large clonal B cells in the CSF or
shown sufficient evidence of specificity to allow use in rou- vitreous fluid by multi-parameter flow cytometry
tine practice (Barajas et al, 2009; Wieduwilt et al, 2012; and/or polymerase chain reaction for IGHV gene
Kawai et al, 2013; Valles et al, 2013). Leptomeningeal rearrangements. (1B)

ª 2018 British Society for Haematology and John Wiley & Sons Ltd 349
British Journal of Haematology, 2019, 184, 348–363
Guideline

Table I. Pre-treatment investigations and staging for primary central nervous system lymphoma.

Histology Imaging Clinical evaluation Laboratory evaluation CSF analysis*

Essential Tissue diagnosis: Gadolinium-enhanced MRI Physical examination including Renal and liver
brain and spine* full neurological assessment function
a) Stereotactic or surgical Systemic cross-sectional Full medical history and drug Serum LDH
biopsy imaging (PET-CT preferred history, including
b) Vitrectomy specimen but contrast-enhanced CT of corticosteroid use
(if PIOL suspected) neck to pelvis acceptable)
c) CSF (if no other diag- Testicular ultrasound Performance status Creatinine clearance
nostic material) Ophthalmological examination HIV, hepatitis B
with fundoscopy and slit-lamp (including core Ab)
examination and C serology
Baseline MMSE

Desirable Bone marrow trephine Whole body FDG-PET† Formal neuropsychological CSF protein
and aspirate† Assessment of LVEF if assessment (see Table II) Cytology
indicated assessment

Ab, antibody; CSF, cerebrospinal fluid; CT, computed tomography; FDG, fluorodeoxyglucose; HIV, human immunodeficiency virus; LDH, lactate
dehydrogenase; LVEF, left ventricular ejection fraction; MMSE, mini mental state evaluation; MRI, magnetic resonance imaging; PET, positron
emission tomography; PIOL: primary intraocular lymphoma.
*Unless contraindicated.
†Strongly encouraged unless likely to cause treatment delay.

7 Contrast-enhanced MRI (including diffusion sequences) Fitness for chemotherapy should be determined by physio-
is the recommended imaging modality pre-treatment logical fitness (organ function and comorbidities) rather than
and for response assessment. Neuroaxis imaging (brain chronological age (Zhu et al, 2009; Roth et al, 2012; Welch
and entire spinal cord) should be reviewed by a special- et al, 2012).
ist neuroradiologist. (1B) Performance status is frequently impaired at diagnosis of
8 Thorough ophthalmological assessment, including slit PCNSL and should not preclude intensive induction ther-
lamp examination, should be performed in all patients apy. Performance status often improves following initial
to exclude intraocular involvement. (1B) therapy which may allow subsequent intensification of ther-
9 All patients should undergo cross-sectional imaging to apy. Moreover, both intensive multi-agent induction and
exclude systemic disease. (1A) high dose therapy with autologous stem cell transplant
(HDT-ASCT) approaches are feasible in selected older
a F deoxyglucose positron emission tomography –
18
patients, including some aged >70 years (Schorb et al,
computed tomography (PET-CT) is recommended.
2017a). No validated comorbidity assessment exists to guide
Contrast-enhanced CT of neck/chest/abdomen/pelvis
treatment choices. High-dose methotrexate (HD-MTX)-
should be performed if PET-CT is not possible. (1B)
based protocols require adequate renal, hepatic and cardiac
b Men should undergo testicular ultrasonography. (1B)
function and may require dose adjustment in line with
10 All confirmed PCNSL cases should be discussed at a institutional and manufacturer’s guidance if impaired (e.g.,
lymphoma MDT. Patients should receive definitive left ventricular ejection fraction >45% and creatinine clear-
treatment as soon as possible, ideally within 14 days ance <50 ml/min).
of diagnosis, at an established centre with multidisci- In practical terms, PCNSL patients may be delineated into
plinary PCNSL expertise. (1B) three treatment fitness groups:
1 Eligible for intensive combination immuno-chemotherapy
incorporating HD-MTX
Treatment of primary CNS lymphoma 2 Eligible for HD-MTX-based immuno-chemotherapy but
ineligible for intensive combination chemotherapy
Optimal therapy of PCNSL incorporates two phases of treat-
3 Ineligible for HD-MTX-based therapy: palliative treatment
ment: remission induction followed by consolidation. This
(a minority of patients)
concept largely relates to challenges in delivering sufficient
dose intensity across the blood–brain barrier and the high Outcomes for PCNSL patients have improved significantly
rates of PCNSL relapse when consolidation therapy is not over the past decade, largely as a result of alterations in man-
delivered (Ferreri, 2011). agement based on evidence emerging from prospective

350 ª 2018 British Society for Haematology and John Wiley & Sons Ltd
British Journal of Haematology, 2019, 184, 348–363
Guideline

clinical trials. Patients should be offered entry into a clinical haematological toxicity. In a recent real world study of out-
trial wherever available. comes with MATRix chemotherapy (median age 61 years),
23 of 88 included patients would not have met IELSG32 eli-
gibility criteria due to age, performance status or co-morbid-
Remission induction
ities (Schorb et al, 2017b). Consequently, chemotherapy
modifications were more frequent (40–50%) but survival
Intensive methotrexate-based induction
rates were similar to those in IELSG32, with 2-year OS of
immunochemotherapy
64%. Severe infectious complications and intensive care sup-
Combination chemotherapy regimens incorporating HD- port were more common during cycle 1 (16%) than cycle 4
MTX are considered the standard of care for newly diag- (5%). Thus, for patients considered at risk of increased toxic-
nosed PCNSL, resulting in high rates of initial response when ity (any of: Eastern Cooperative Oncology Group perfor-
combined with other agents (Hoang-Xuan et al, 2015; Ferreri mance status ≥2, co-morbid conditions or age >65 years) we
et al, 2016). Penetration of MTX into the CNS is influenced recommend dose reducing the myelotoxic agents in MATRix
by the total dose and rate of infusion. Most studies employ (cytarabine and thiotepa) for the initial cycle, with dynamic
doses of between 3 and 8 g/m2 (Rubenstein et al, 2013; Fer- review cycle by cycle. In practice, a 25% reduction of cytara-
reri et al, 2016; Glass et al, 2016) although the optimal dose bine (achieved by omitting the 4th dose in the cycle) with or
has not been established. It is crucial that MTX is adminis- without a 25% dose reduction of thiotepa is a reasonable
tered as a rapid infusion (2–4 h) at a dose of at least 3 g/m2 strategy. Granulocyte colony-stimulating factor (G-CSF) and
to maximise therapeutic CSF concentrations, at an interval of anti-infection prophylaxis (against herpes simplex and Pneu-
10–21 days, as part of an established protocol. Modern pro- mocytis jirovecii as a minimum) are recommended.
tocols typically employ four to eight cycles of HD-MTX- A number of alternative remission-induction regimens
based therapy. The optimal number of treatment cycles is have been assessed in non-randomised trials, limiting reli-
likely to be influenced by partner chemotherapy agents, dose able comparisons. Promising results have been reported with
intensity of the regimen and intended consolidation HD-MTX, temozolomide and rituximab (MT-R), with a
approach, but comparative data are not available. variety of consolidation strategies (2-year PFS of 57–64%)
The International Extranodal Lymphoma Study Group (Rubenstein et al, 2013; Glass et al, 2016). The R-MPV regi-
(IELSG) study IELSG20 demonstrated the value of combin- men (HD-MTX, procarbazine, vincristine and rituximab)
ing HD-MTX with partner cytotoxic agents: the addition of has also been associated with encouraging results in a num-
cytarabine (4 doses of 2 g/m2) significantly improved rates of ber of small, non-comparative phase 2 studies (Morris et al,
complete response (CR) and progression-free survival (PFS) 2013; Omuro et al, 2015a,b). R-MBVP induction
as compared to HD-MTX alone (Ferreri et al, 2009). The chemotherapy (rituximab, HD-MTX, BCNU [carmustine],
randomised phase 2 IELSG32 trial subsequently demon- prednisolone, etoposide) followed by either WBRT or HDT-
strated that the addition of eight doses of rituximab ASCT resulted in a 2-year OS of 86% in the PRECIS trial
(375 mg/m2) to HD-MTX/cytarabine improved response (Houillier et al, 2016). A recent randomised study failed to
rates. Importantly, the addition of both thiotepa and ritux- demonstrate that the addition of rituximab to the same
imab to HD-MTX/cytarabine as a 4-drug regimen (MATRix; MBVP regimen improves outcomes (hazard ratio for PFS:
see Appendix S2) resulted in a clear improvement in overall 077, 95% CI 052–113, P = 018) (Bromberg et al, 2017).
survival (OS) over HD-MTX/cytarabine alone, with a 2-year However, a post hoc analysis showed evidence of a PFS ben-
OS rate of 69% (95% confidence interval [CI] 64–74) vs. efit in patients aged <60 years, and those aged ≥60 years
42% (95% CI 36–48), respectively. Patients with stable dis- did not receive consolidation therapy; the full study publica-
ease or better after four cycles of MATRix were subsequently tion is awaited.
randomised to receive consolidation with whole brain radio- Whilst rituximab and HD-MTX are internationally
therapy (WBRT) or HDT-ASCT. Peripheral blood stem cells accepted as standard of care (Hoang-Xuan et al, 2015), the
(PBSC) were successfully collected in 96% patients after two optimal induction regimen remains uncertain. Although a
cycles of MATRix (Ferreri et al, 2016). PBSC collection after number of published protocols are associated with promising
MATRix should be attempted but may be less successful if outcomes, MATRix is recommended given the higher level of
deferred beyond cycle 2 (C.P. Fox and K. Cwynarski, unpub- randomised evidence demonstrating clear survival advantages
lished data). over comparator arms.
Treatment-related mortality is around 4–7% with The additional value of concurrent intrathecal chemother-
MATRix, with most treatment-related deaths occurring dur- apy is unproven, with conflicting data from published series
ing the first treatment cycle (Ferreri et al, 2016; Schorb et al, (Ferreri et al, 2002a; Khan et al, 2002; Pels et al, 2009; Sierra
2017b). Within the IELSG32 study (median age 57 years), Del Rio et al, 2012). In view of the risks and potential morbid-
the relative dose intensity of cytarabine and thiotepa during ity associated with repeated lumbar punctures, low level of
remission induction was 78% and 76% respectively, with available evidence and improved efficacy of systemic therapy,
protocol-defined reductions predominantly for we do not advocate concurrent intrathecal chemotherapy.

ª 2018 British Society for Haematology and John Wiley & Sons Ltd 351
British Journal of Haematology, 2019, 184, 348–363
Guideline

Surgical intervention has a very limited role in PCNSL >60 years. HD-MTX-based therapy was associated with
therapy. Although a retrospective analysis of the German improved OS, although there was no discernible survival
PCNSL Study Group (G-PCNSL-SG)-1 trial suggested that benefit to using intensive intravenous treatment protocols
patients with subtotal or total resections had improved out- over HD-MTX combined with oral alkylating agents
comes (Weller et al, 2012), this finding is likely to be influ- (Kasenda et al, 2015).
enced by patient selection bias; resection was more
commonly performed for superficial lesions associated with a
Palliative therapy
better prognosis (Ferreri et al, 2003). These findings remain
controversial and have not been adopted by consensus For patients considered unfit for MTX-based therapy there
(Hoang-Xuan et al, 2015). Given that PCNSL is considered a remains a paucity of good quality data to inform treatment
whole brain disease (Lai et al, 2002), therapeutic resection of decisions. WBRT, corticosteroids and oral chemotherapy are
PCNSL lesions should be restricted to critical circumstances common approaches. A small retrospective study of single
where urgent surgical reduction of intracranial pressure is agent temozolomide in elderly patients with co-morbidities
essential. (n = 19) showed a CR rate of 47%, with prolonged responses
(>12 months) in 294%, a median PFS of 5 months and
median OS of 21 months (Kurzwelly et al, 2010). A number
Less intensive methotrexate-based remission induction
of novel agents have shown promise as single agent therapy
Historically, clinical studies for PCNSL have defined ‘elderly’ in relapsed/refractory PCNSL (see below) but there are cur-
patients as >60 years of age, although this cut-off has been rently insufficient data to recommend the use of these, as yet
empirically adopted without a modern evidence-base. It is unlicensed, agents for those unfit for MTX-based therapy. In
clear that chronological age is not a barrier to safe delivery patients aged ≥60 years, WBRT alone (40 Gy + 20 Gy boost)
of HD-MTX (>3 g/m2) if physiological fitness (particularly gave a median survival of only 76 months (Nelson et al,
cardiac and renal function) is deemed adequate (e.g. left ven- 1992). Lower doses and shorter treatment durations (20–
tricular ejection fraction >45% and creatinine clearance 30 Gy in 18–4 Gy fractions) may be a more pragmatic
>50 ml/min) (Zhu et al, 2009; Roth et al, 2012; Welch et al, approach as long term neurotoxicity is not the primary clini-
2012). cal concern. Fatigue is the most common side effect of pallia-
A number of phase 2 studies have focused on less tive WBRT and may take several months to improve.
myelotoxic induction regimens in patients aged ≥60 years, Performance status, life expectancy and quality of life are
typically combining HD-MTX with orally administered important factors to take into consideration when discussing
alkylating agents, without WBRT consolidation (Hoang- treatment options, including best supportive care. Key in this
Xuan et al, 2003; Illerhaus et al, 2009; Fritsch et al, 2011). context are good communication and input from palliative
Recently, the PRIMAIN study assessed HD-MTX with care specialists if required; expectations of patients and their
rituximab, procarbazine and lomustine, followed by procar- families should be carefully managed.
bazine maintenance in patients aged ≥65 years (median age
73 years). The study was amended to omit lomustine due
Response assessment
to excessive toxicity, without any clear loss of efficacy. The
2-year OS with and without lomustine was 479% (95% CI Response should be assessed by contrast-enhanced MRI with
304–653) and 46% (95% CI 341–578), respectively neuroradiology review according to international guidelines
(Fritsch et al, 2017). A Nordic study used a modified Bonn (Abrey et al, 2005), as summarised in Table II. The role of
protocol (Pels et al, 2003) in patients aged 65–75 years interim MRI has not been clearly defined, although early
(median age 70 years), incorporating reduced dose HD- achievement of CR after two cycles of chemotherapy (of 6)
MTX-based polychemotherapy with temozolomide, followed appears to be associated with improved OS (Pels et al, 2010).
by maintenance temozolomide for 1 year. Estimated 2-year Early response assessment after one to two cycles may also
OS was 556% (95% CI 353–718), which was similar to a facilitate decisions regarding PBSC harvest and allow detec-
younger cohort receiving a more intensive version of the tion of early disease progression in the 8–29% who progress
same regimen (2-year OS 607%, 95% CI 433–742) (Pul- during first-line chemotherapy (Ferreri et al, 2016; Langner-
czynski et al, 2015). The only randomised phase 2 study in Lemercier et al, 2016). Patients with non-progressive disease
patients aged >60 years (median age 72 years) compared (stable disease or better) following induction therapy should
HD-MTX, procarbazine, vincristine and cytarabine (MPV- be considered for consolidation therapy (Soussain et al,
A) to HD-MTX and temozolomide (MT), without ritux- 2008; Illerhaus et al, 2016).
imab. No significant difference between the arms could be
demonstrated with a 1-year PFS of 36% in both arms
Recommendations
(Omuro et al, 2015a).
A meta-analysis evaluated individual patient data from 20 1 Fitness for chemotherapy should be determined by
prospective and retrospective studies in PCNSL patients aged physiological fitness rather than chronological age. (1A)

352 ª 2018 British Society for Haematology and John Wiley & Sons Ltd
British Journal of Haematology, 2019, 184, 348–363
Guideline

Table II. Response criteria for primary central nervous system lymphoma.

Response Brain imaging Corticosteroid dose Eye examination CSF cytology

CR No contrast enhancement None Normal Negative


CRu No contrast enhancement Any Normal Negative
Minimal abnormality Any Minor RPE abnormality Negative
PR 50% decrease in enhancing tumour Irrelevant Minor RPE abnormality or normal Negative
No contrast enhancement Irrelevant Decrease in vitreous cells or retinal Persistent or suspicious
infiltrate
SD <50% decrease and <25% increase in lesion Irrelevant No new ocular disease Persistent
PD 25% increase in lesion Irrelevant Recurrent or new ocular disease Recurrent or new disease
Any new site of disease: CNS or systemic

From Abrey et al. (2005). Reprinted with permission. ©2005 American Society of Clinical Oncology. All rights reserved.
CNS, central nervous system; CR, complete response; CRu, unconfirmed complete response; CSF, cerebrospinal fluid; PR, partial response; PD,
progressive disease; RPE, retinal pigment epithelium; SD, stable disease.

2 Patients should be offered a clinical trial wherever pos- 5 Intrathecal chemotherapy is not recommended along-
sible. (1A) side systemic CNS-directed therapy (1A) but may be
3 For patients eligible for HD-MTX-based regimens: considered for symptomatic control of leptomeningeal
disease in patients unfit for systemic therapy. (2C)
a If fit for intensive therapy, offer treatment with
6 Response assessment should be performed with con-
four cycles of MATRix (HD-MTX, cytarabine, thio-
trast-enhanced MRI:
tepa, rituximab) immuno-chemotherapy. (1A)
a Consider performing after cycle 1 to inform timing
i Dose reductions should be considered for those
of PBSC collection. (2C)
with impaired performance status at presentation,
b Routinely perform every 2 cycles of HD-MTX-based
co-morbid conditions and/or who experience sig-
therapy and at the end of remission induction
nificant toxicity from MATRix. (2C)
therapy. (1B)
ii Granulocyte colony-stimulating factor and pro-
phylaxis against opportunistic infections should
be employed (2C)
iii For patients where high dose therapy with autol- Consolidation treatment
ogous stem cell transplant (HDT-ASCT) is
planned, PBSC collection should be attempted Timing of consolidation therapy
following cycle 2, if practicable. (1B)
Given the inherent difficulty in delivering optimal dose
b If unfit for intensive therapy, offer treatment incor- intensity across the blood–brain barrier, the risk of early
porating HD-MTX, rituximab and an orally admin- relapse, and the important role of consolidation in the man-
istered alkylating agent within an established agement of PCNSL, we recommend that clinicians plan to
protocol (e.g. R-MP [rituximab, MTX, procar- commence consolidation therapy within 6–8 weeks of the
bazine], see Appendix S3). (1B) first day of the final induction chemotherapy cycle. This is a
c HD-MTX should be delivered at doses of at least judgement for individual clinicians and assumes that the
3 g/m2 with an infusion time of 2–4 h, for a mini- patient’s performance status is reasonable and any significant
mum of four cycles at 2- to 3-week intervals. (1B) toxicities have sufficiently resolved.
d Rituximab should be delivered at 375 mg/m2 for
eight doses (i.e. 2 doses per cycle with MATRix) (1A)
Radiotherapy consolidation
4 For those patients ineligible for HD-MTX, consider
The diffuse multifocal nature of PCNSL necessitates WBRT
one, or a combination, of the following treatments.
rather than targeted radiotherapy. Most protocols empirically
(2C)
use a WBRT dose of 30–45 Gy although it should be empha-
a Oral chemotherapy (such as temozolomide) sised that the optimal total dose and role of a ‘boost’
b Whole-brain radiotherapy (WBRT; 20–30 Gy in 18– remains uncertain, particularly in the era of more intensive
4 Gy fractions according to performance status, remission induction protocols. WBRT has traditionally been
therapeutic aims and life expectancy) +/ orbital used as consolidation following response to first-line
radiotherapy if co-existing ocular involvement chemotherapy (DeAngelis et al, 2002), but its role in the
c Corticosteroids (dexamethasone is typically used) context of modern immuno-chemotherapy is less clear. A

ª 2018 British Society for Haematology and John Wiley & Sons Ltd 353
British Journal of Haematology, 2019, 184, 348–363
Guideline

randomised phase 3 trial compared WBRT (45 Gy) with no Importantly, results of the first randomised trials compar-
further treatment in patients achieving CR following MTX- ing WBRT consolidation with HDT-ASCT have recently
based chemotherapy (Thiel et al, 2010; Korfel et al, 2015). been reported. Interim analysis of the PRECIS trial reported
The design and conduct of this study have been criticised; a 2-year PFS of 632% (95% CI 495–805) for WBRT con-
only 58% patients received protocol treatment and the trial solidation versus 868% (95% CI 766–983) after HDT-
failed to meet its non-inferiority end-point. Therefore, it ASCT, with identical 2-year OS between the two arms
remains unclear whether WBRT consolidation can be safely (Houillier et al, 2016). The IELSG32 trial reported no signif-
omitted for patients in CR (Thiel et al, 2010). icant difference in PFS or OS between WBRT and HDT-
Irreversible, and sometimes disabling, neurocognitive ASCT consolidation on intention-to-treat analysis (2-year
dysfunction is a well-recognised consequence of WBRT, OS 80% [95% CI 70–90] vs. 69%, respectively [95% CI 59–
particularly in those aged >60 years. Indeed, a systematic 79]) (Ferreri et al, 2017). Although longer follow-up and
meta-analysis found no clear overall benefit, in terms of neurocognitive results are awaited, a reduced incidence of
quality-adjusted life years, for WBRT in first remission in delayed neurotoxicity is likely to favour HDT-ASCT as con-
patients >60 years of age (Prica et al, 2012). In an attempt to solidation therapy. Notably, feasibility of consolidation with
mitigate this risk, trial strategies have investigated hyper-frac- HDT-ASCT has also been demonstrated in selected older
tionated or lower radiation doses (Correa et al, 2009; Doolit- patients (>65 years old) by a recent European collaborative
tle et al, 2013; Ferreri et al, 2016; Glass et al, 2016). Use of study (Schorb et al, 2017a).
reduced dose radiotherapy (234 Gy) as consolidation for A non-myeloablative chemotherapy consolidation
patients in CR has been explored by Morris et al (2013) in a approach has been investigated as an alternative to HDT-
non-randomised phase 2 study. In this subset of patients ASCT. A single arm trial of intensive EA consolidation
(n = 12), survival and neurocognitive outcomes were encour- (etoposide/cytarabine) after MT-R induction (HD-MTX/
aging, but the number of evaluable patients was small and temozolomide/rituximab) reported a 4-year OS of 65%.
definitive conclusions cannot be drawn (Morris et al, 2013). Notwithstanding a relatively high rate of early disease pro-
Randomised studies are needed to validate this approach. gression, long-term disease control using this strategy appears
Any decision to employ WBRT as consolidation should be broadly comparable to chemo-radiation protocols (Ruben-
based on high quality MRI imaging reviewed by a neuroradi- stein et al, 2013). Randomised trials are ongoing to ascertain
ologist or, ideally, following neuroradiology consensus opin- whether non-myeloablative chemotherapy approaches, such
ion through MDT structures. as EA or DeVIC (dexamethasone, etoposide, ifosfamide and
carboplatin) (Motomura et al, 2011) are comparable to
HDT-ASCT consolidation (NCT01511562).
Chemotherapy consolidation
Encouraging results from early studies with HDT-ASCT con-
Recommendations
solidation in PCNSL have challenged the role of WBRT as the
favoured first-line consolidation strategy (Soussain et al, 2001; 1 Consolidation therapy should be considered for all
Illerhaus et al, 2006). On an intention-to-treat basis, prospec- patients with non-progressive disease following induc-
tive trials of thiotepa/carmustine-conditioned ASCT after tion chemotherapy. This decision should be informed
intensive induction chemotherapy have reported 3- to 5-year by comorbidities, performance status, neurocognitive
OS rates of 70–81%, where 79–92% received planned HDT- function and patient wishes. (1B)
ASCT (Kasenda et al, 2012; Illerhaus et al, 2016). In the study 2 High-dose thiotepa-based chemotherapy with ASCT as
by Illerhaus et al (2016) only a minority (14%) received addi- first-line consolidation should be considered for all eli-
tional WBRT. Broadly similar results have also been achieved gible patients. (1B)
with thiotepa, busulfan and cyclophosphamide conditioning
a Patients with PCNSL achieving at least stable disease
(Soussain et al, 2012; Omuro et al, 2015b; Houillier et al,
following HD-MTX-based first-line therapy should
2016), although no formal comparison of conditioning regi-
be considered for HDT-ASCT. (1B)
mens has been conducted. Historical results with the BEAM
b BEAM (carmustine, etoposide, cytarabine, melpha-
regimen (carmustine, etoposide, cytarabine, melphalan) were
lan) should not be used as HDT-ASCT conditioning
disappointing (Abrey et al, 2003). In the IELSG32 study, 24 of
for PCNSL. (1A)
28 patients with partial response or stable disease after induc-
tion therapy achieved CR following HDT-ASCT (Ferreri et al, 3 WBRT consolidation +/ boost should be considered
2017). Long-term survival rates, however, are lower in for:
chemorefractory patients who fail to achieve partial remission
a Patients ineligible for HDT-ASCT with residual dis-
to induction therapy (Soussain et al, 2012; Schorb et al,
ease following induction immunochemotherapy. (1B)
2013). Survival outcomes from the prospective studies have
b Patients with residual disease after thiotepa-based
been mirrored by a recently published ‘real world’ UK retro-
ASCT. (1B)
spective study (Kassam et al, 2017).

354 ª 2018 British Society for Haematology and John Wiley & Sons Ltd
British Journal of Haematology, 2019, 184, 348–363
Guideline

4 Patients with concurrent ocular involvement should following completion of therapy. Further surveillance
also be considered for bilateral ocular radiotherapy (see MRI imaging should be judged on an individual patient
PIOL section) if ineligible for HDT-ASCT, or not in basis. (2B)
complete response (CR) following thiotepa-based ASCT.
(2B)
5 For HDT-ASCT-ineligible patients in CR after HD Primary intraocular lymphoma
MTX-regimens, WBRT consolidation is contentious:
Primary intraocular lymphoma is classified as a variant of
a Potential improvement in progression-free survival PCNSL with its own therapeutic considerations. The optimal
should be carefully balanced against risks of neu- treatment for intraocular disease remains controversial as evi-
rocognitive toxicity for individual patients. (1B) dence is largely limited to retrospective and small prospective
b For patients aged ≥60 years in CR after HD MTX, case series.
either WBRT should be omitted, or lower dose Given that CNS lymphoma is the principal cause of death
WBRT consolidation may be considered given the in PIOL (Grimm et al, 2007), the potential coexistence of
higher risk of neurocognitive toxicity. (2B) CNS disease should be proactively addressed within PIOL
6 Where WBRT is offered, the following dosing schedules treatment algorithms. Chemotherapy agents that penetrate
are recommended depending on age, comorbidities and the blood-brain barrier, particularly HD-MTX and cytara-
induction treatment received. bine, also cross the blood-ocular barrier (Baumann et al,
1986; Siegel et al, 1989; Batchelor et al, 2003), resulting in
a 36 Gy in 20 fractions (1B). ocular responses and persistent remissions (Grimm et al,
b Reduced dose (234 Gy in 18 or 2 Gy fractions) for 2007; Riemens et al, 2015). Single agent ifosfamide and tro-
selected cases at higher risk of neurotoxicity. (2C) fosfamide have also been shown to have some efficacy in a
c Consider a 9 Gy boost with a 1–2 cm margin (total small prospective study (n = 10) (Jahnke et al, 2009). The
dose 45 Gy/25 fractions) to residual enhancing optimal combination of these drugs is yet to be determined.
lesion(s) at the time of WBRT. Intravitreal chemotherapy alone, usually MTX +/ ritux-
d Orbits should be shielded after 30 Gy (36 Gy if imab (Frenkel et al, 2008; Larkin et al, 2014) can achieve
previously documented intraocular disease). remission in a proportion of patients. However, CNS relapse
occurs in 33–58% patients, and ocular toxicity is reported in
26–36% (Grimm et al, 2007; Riemens et al, 2015), hence,
Follow-up systemic PCNSL regimens should be employed. A limited
Around 6–25% of all relapses are asymptomatic and detected number of small retrospective and single arm studies have
on follow-up imaging (Langner-Lemercier et al, 2016; Fos- reported promising outcomes with a combined intravitreal
sard et al, 2017; Mylam et al, 2017). In a large population- and systemic approach (Ma et al, 2016). However, ran-
based retrospective study, asymptomatic patients had better domised data are absent and, given the potential for addi-
performance status, which may have facilitated delivery of tional ocular toxicity, this approach cannot be routinely
intensive salvage therapy, and was associated with improved recommended.
outcomes. On multivariable analysis, performance status at Irrespective of treatment modality used, relapse rates
relapse was one of the factors most strongly associated with remain high (Grimm et al, 2007; Riemens et al, 2015;
OS (Langner-Lemercier et al, 2016). Smaller studies have not Nguyen et al, 2016) therefore consolidation therapy is rec-
shown a clear benefit for surveillance imaging, but this may ommended. Both ocular radiotherapy (usually incorporating
relate to the frequency and timing of surveillance (Fossard both globes +/ WBRT) (Ferreri et al, 2002b) and HDT-
et al, 2017; Mylam et al, 2017). Prospective data are lacking. ASCT (Soussain et al, 2001) can induce durable remissions
A recommended schedule for disease monitoring has been in PIOL. At present there are insufficient data to recommend
outlined in international consensus guidelines (Abrey et al, one consolidation strategy over another.
2005).
Recommendations
Recommendations
1 PIOL should be treated with systemic HD-MTX-based
1 Response assessment with contrast-enhanced MRI combination chemotherapy with rituximab. For fit
should be performed 1–2 months after completion of patients, consider using evidence-based PCNSL induc-
consolidation therapy. (1B) tion protocols, such as the MATRix regimen. (1C)
2 Follow-up imaging with contrast-enhanced MRI may be 2 Intravitreal MTX (administered by a specialist ophthal-
considered, particularly in the first 2 years following mologist) can be considered for elderly patients with
treatment, in patients eligible for salvage therapies. A isolated PIOL who are unfit for systemic therapy. (2C)
suggested schedule is every 3–4 months for 2 years

ª 2018 British Society for Haematology and John Wiley & Sons Ltd 355
British Journal of Haematology, 2019, 184, 348–363
Guideline

3 Concurrent intravitreal therapy, for patients receiving a appears to have activity in PCNSL when combined with
systemic HD-MTX regimen, cannot be routinely recom- rituximab (Ghesquieres et al, 2016; Rubenstein et al, 2016).
mended. (2C) However, these agents are not yet licensed for this indication
4 For PIOL patients who have responded to intensive sys- and survival data are immature.
temic chemotherapy, the following consolidation For eligible, chemosensitive patients, available data sup-
options should be considered: port the use of thiotepa-based HDT-ASCT in R/R PCNSL,
with a median PFS of 24–41 months reported (Soussain
a For eligible patients, high-dose thiotepa-based
et al, 2008, 2012; Kasenda et al, 2017). Either HDT-ASCT or
chemotherapy with ASCT
WBRT should be considered in second remission if not
b Bilateral orbital radiotherapy (up to 36 Gy in
undertaken as first-line consolidation therapy. WBRT alone
18–2 Gy fractions). Concurrent WBRT (234–30 Gy
(median dose 36–40 Gy) can result in CR rates of 37–58%
in 18–2 Gy fractions) should be considered but
and a median survival of 10–16 months in radiotherapy-
needs to be carefully balanced against risks of
na€ıve patients who are refractory to, or relapse after, HD-
neurocognitive toxicity for individual patients. (2B)
MTX therapy (Nguyen et al, 2005; Hottinger et al, 2007;
Khimani et al, 2011).

Relapsed and refractory PCNSL


Recommendations
Treatment of relapsed and refractory (R/R) PCNSL remains
a major area of unmet clinical need. The prognosis of R/R 1 All patients with suspected PCNSL relapse should be
PCNSL is very poor with a median OS of 35 months reviewed urgently within regional MDT meetings. The
(Langner-Lemercier et al, 2016). Published studies of salvage primary treating Haemato-oncology team should be
therapy for R/R PCNSL comprise small, non-randomised promptly informed. (1C)
and often retrospective analyses, with low quality evidence 2 Re-biopsy at relapse is recommended in the context of
and thus no established standard of care. Studies of non- atypical MRI appearances, or for new brain lesions
myeloablative chemotherapy, either single-agent or combina- occurring beyond 2 years from initial therapy, particu-
tion approaches, typically report dismal outcomes with larly if intensive salvage therapy is planned. (2C)
response rates of 30–55% and median PFS of 2–11 months 3 Patients with a confirmed diagnosis of relapsed PCNSL
(Grommes & DeAngelis, 2017). Consequently, patients should undergo complete re-staging if further therapy
should be offered clinical trial entry wherever available. is planned. Re-staging is not usually necessary for
Ifosfamide-based salvage regimens, usually in combination PCNSL refractory to first-line therapy. (1C)
with etoposide +/ carboplatin and rituximab (R-IE and R- 4 Wherever possible, patients should be offered participa-
ICE), have resulted in overall response rates of 41–95% in tion in a clinical trial. (1C)
predominantly chemorefractory or heavily treated patient 5 Outside of clinical trials, potential treatment options
cohorts (Motomura et al, 2011; Choi et al, 2013; Mappa should be individualised (1C), taking account of:
et al, 2013; Choquet et al, 2015; Langner-Lemercier et al,
a Physiological fitness, performance status and neu-
2016). For patients who experience durable first remissions
rocognitive function
following HD-MTX-based protocols, re-treatment with HD-
b Previous therapy and duration of response
MTX-based regimens may be effective, although the effective-
c Patient choice
ness of this approach is much more uncertain in the mod-
ern-era of intensified MTX-containing regimens. Two 6 For patients eligible for intensive treatment: (see Fig 1)
retrospective studies described a median PFS of 16 and
a Consider ifosfamide-based immunochemotherapy,
258 months after re-treatment with HD-MTX at relapse,
particularly for refractory disease or early relapse
usually as part of multi-agent salvage regimens, in patients
after MTX-based immunochemotherapy. (2B)
who had experienced as median duration of response to
b Consider HD-MTX-based immunochemotherapy if
first-line HD-MTX of 244 and 26 months, respectively
the duration of first remission to HD-MTX-based
(Plotkin et al, 2004; Pentsova et al, 2014). This may be par-
therapy was >2 years. (2B)
ticularly relevant for MTX-experienced, but rituximab- and
thiotepa-na€ıve, patients for whom the more intensive 7 Consolidation following salvage chemotherapy:
MATRix protocol may be an option, although it should be
a Patients who have not previously undergone HDT-
noted that currently no published data exist for MATRix in
ASCT should be considered for thiotepa-based
R/R PCNSL. Encouraging results have been achieved with a
HDT-ASCT in second or subsequent response. (1B)
number of novel agents, with response rates >50% observed
b WBRT-naive patients who are ineligible for, or
with single agent ibrutinib (Chamoun et al, 2017; Grommes
have previously undergone, HDT-ASCT should be
et al, 2017; Lionakis et al, 2017) and nivolumab (Nayak et al,
considered for WBRT (234–36 Gy in 18–2 Gy
2017). Lenalidomide also crosses the blood–brain barrier and

356 ª 2018 British Society for Haematology and John Wiley & Sons Ltd
British Journal of Haematology, 2019, 184, 348–363
Guideline

Fit for intensive Consider WBRT and/or


chemotherapy? oral temozolomide +/-
NO rituximab
YES
Refractory to, or relapse within
2 years of MTX-based
combination therapy?
YES NO

Consider rituximab and Consider rituximab plus either


ifosfamide-based salvage MTX-based therapy OR
chemotherapy ifosfamide-based salvage

Assess objective response and clinical benefit after


2 cycles of salvage therapy

CR, PR or SD with clinical PD or SD without clinical


benefit and good PS benefit, or poor PS

ASCT eligible ASCT ineligible Consider palliative WBRT


and/or best supportive care

Consolidate with Consider WBRT


Fig 1. Algorithm for treating patients with thiotepa-based and/or oral
ASCT alkylator
relapsed/refractory primary central nervous sys- consolidation
tem lymphoma. ASCT, autologous stem cell
transplant; CR, complete response; MTX, high Consider WBRT
dose methotrexate; PD, progressive disease; PR, consolidation for
partial response; PS, performance status; SD, residual disease
stable disease; WBRT, whole brain after ASCT if
radiotherapy. WBRT-naïve

fractions), either alone or following salvage studies, cognitive impairment was found in most PCNSL
chemotherapy. (2C) patients treated with WBRT plus chemotherapy whereas
patients treated with chemotherapy alone had either stable or
8 Patients ineligible for intensive treatment:
improved cognitive performance (Correa et al, 2007). In the
a Palliative treatment should be offered, which may longest observational study of 80 PCNSL survivors in CR,
include WBRT (234–36 Gy in 18–2 Gy fractions), patients receiving WBRT had lower mean scores in attention,
corticosteroids and/or oral temozolomide. (2C) executive function, motor skills and overall neuropsychological
b Patients should receive best supportive care and, composite score compared with those treated without WBRT
where appropriate, palliative care input. (1B) (Doolittle et al, 2013). Treatment-related cognitive morbidity
is also associated with a lower quality of life and poor progno-
sis (Prica et al, 2012; Doolittle et al, 2013).
Neuropsychological assessments
There is no consensus on the optimal neurocognitive test
Cognitive impairment is a feature of PCNSL, with inevita- battery for PCNSL patients, or on the interval at which fol-
ble heterogeneity in neuropsychological deficits being asso- low-up neurocognitive assessments should be performed.
ciated with different tumour locations. In some patients Baseline and serial mini mental state examination have been
there may also be unilateral weakness affecting motor func- suggested as a minimum requirement (Abrey et al, 2005).
tion and/or behavioural syndromes, such as disinhibition or Table III shows the proposed minimum core battery for the
apathy, which may influence test selection or approach assessment of neuropsychological functions and quality of life
during assessments. in PCNSL patients recommended by Correa et al (2007) and
Elucidating the cognitive effects of disease versus treatment proposed alternative measures for UK patients. The battery
is difficult due to methodological limitations of published can be completed within 30–40 min and has been incorpo-
studies, including lack of baseline cognitive assessments and rated into prospective trials (Correa et al, 2007).
control groups, and variable disease status. The type of treat- Given the cognitive, psychological and physical effects of
ment and age are important considerations. In a review of 17 both CNS lymphoma and its treatment, it is important that

ª 2018 British Society for Haematology and John Wiley & Sons Ltd 357
British Journal of Haematology, 2019, 184, 348–363
Guideline

Table III. Proposed baseline and follow-up neuropsychological evaluations in primary central nervous system lymphoma.

Domain Tests recommended by Correa et al (2007) Tests recommended for use with UK patients

Premorbid IQ Barona Index (Barona et al, 1984) National Adult Reading Test-Revised (Nelson & Willison,
estimation 1991) or Test of Premorbid Functioning (Wechsler, 2011)
Attention/executive WAIS-III (Wechsler, 1997) Digits Forward and WAIS-IV Digits Forward, Backward Span and Sequencing Span
Backward Span; Trail Making Test (Parts A and B) (Wechsler, 2008); Trail Making Test (Parts A and B)
(Reitan & Wolfson, 1985)
Verbal memory Hopkins Verbal Learning Test-Revised (Benedict et al, Hopkins Verbal Learning Test-Revised or California Verbal
1998) Learning Test Standard Form (Delis et al, 2000)
Motor Grooved Pegboard Test (Russell & Starkey, 1993) Grooved Pegboard Test
Quality of life EORTC-QLQ-C30 (Aaronson et al, 1993) EORTC-QLQ-C30
BCM 20 (Osoba et al, 1996) BCM 20

BCM 20, Brain Cancer Module 20; EORTC-QLQ-C30, The European Organisation for Research and Treatment of Cancer Quality of Life Ques-
tionnaire C-30; IQ, intelligence quotient; WAIS-III, Wechsler Adult Intelligence Scale Third Edition; WAIS-IV, Wechsler Adult Intelligence Scale
Fourth Edition.

holistic needs are addressed during treatment and through- interests to the BSH and Task Force Chairs which may be
out the recovery period. Early referral to support services viewed on request. CPF, KL and KC have received advi-
and specialist therapies should be considered according to sory board and/or speakers’ fees from Adienne. CPF, EHP,
individual patient need. EGE, CH, PM, AD and KC have received financial reim-
bursement, advisory board fees and/or research funding
from F. Hoffman-La Roche. The following members of the
Recommendations
writing have no conflicts of interest to declare: JS, DPA
1 Cognitive and quality of life outcomes should be and CFu.
assessed in all patients with PCNSL before and after
treatment, with ongoing long-term monitoring.
2 As a minimum, all patients receiving treatment for Review process
PCNSL should be assessed on cognitive domains of
Members of the writing group will inform the writing group
attention, processing speed, motor speed, executive
Chair if any new pertinent evidence becomes available that
function and memory before and after treatment.
would alter the strength of the recommendations made in
this document or render it obsolete. The document will be
archived and removed from the BSH current guidelines web-
Disclaimer
site if it becomes obsolete. If new recommendations are
While the advice and information in this guidance is believed made an addendum will be published on the BSH guidelines
to be true and accurate at the time of going to press, neither website (https://b-s-h.org.uk/guidelines/).
the authors, the BSH nor the publishers accept any legal
responsibility for the content of this guidance.

Author contributions
Acknowledgments
All authors reviewed the literature and contributed to the
The BSH Haemato-oncology task force members at the time drafting and editing of this manuscript.
of writing this guidance were Dr Gail Jones (Chair), Dr Guy
Pratt (Secretary), Dr Simon Stern, Dr Jonathan Lambert, Dr
Nilima Parry-Jones, Dr Pam McKay and Dr Alastair White- Supporting Information
way. The authors would like to thank them, the BSH sound-
Additional supporting information may be found online in
ing board and the BSH guidelines committee for their
the Supporting Information section at the end of the article.
support in preparing this guideline. The authors are also
Appendix S1. Prognostic scores in primary CNS lymphoma.
grateful to Lymphoma Action for their review and input.

Appendix S2. MATRix regimen-methotrexate, cytarabine,


Declaration of interests thiotepa and rituximab.
Appendix S3. R-MP regimen-rituximab, methotrexate and
The BSH paid the expenses incurred during the writing of procarbazine.
this guidance. All authors have made a declaration of

358 ª 2018 British Society for Haematology and John Wiley & Sons Ltd
British Journal of Haematology, 2019, 184, 348–363
Guideline

References Nijland, M., Mason, K., Beeker, A., van den Delis, D.C., Kramer, J.H., Kaplan, E. & Ober, B.A.
Bent, M., Jong, D.D., Gonzales, M.A. & Door- (2000) California Verbal Learning Test, 2nd edn.
Aaronson, N.K., Ahmedzai, S., Bergman, B., Bul- duijn, J.K. (2017) Effect of rituximab in primary The Psychological Corporation, San Antonio.
linger, M., Cull, A., Duez, N.J., Filiberti, A., central nervous system lymphoma – results of Doolittle, N.D., Korfel, A., Lubow, M.A., Schorb,
Flechtner, H., Fleishman, S.B., Dehaes, J.C.J.M., the randomized phase III HOVON 105/ALLG E., Schlegel, U., Rogowski, S., Fu, R., Dosa, E.,
Kaasa, S., Klee, M., Osoba, D., Razavi, D., Rofe, NHL 24 study. Blood, 130, 582. Illerhaus, G., Kraemer, D.F., Muldoon, L.L., Cal-
P.B., Schraub, S., Sneeuw, K., Sullivan, M. & Chamoun, K., Choquet, S., Boyle, E., Houillier, C., abrese, P., Hedrick, N., Tyson, R.M., Jahnke, K.,
Takeda, F. (1993) The European-Organization- Larrieu-Ciron, D., Al Jijakli, A., Delrieu, V., Del- Maron, L.M., Butler, R.W. & Neuwelt, E.A.
for-Research-and-Treatment-of-Cancer QLQ- wail, V., Morschhauser, F., Hoang-Xuan, K. & (2013) Long-term cognitive function, neu-
C30 – a quality-of-life instrument for use in Soussain, C. (2017) Ibrutinib monotherapy in roimaging, and quality of life in primary CNS
international clinical-trials in oncology. Journal relapsed/refractory CNS lymphoma: a retrospec- lymphoma. Neurology, 81, 84–92.
of the National Cancer Institute, 85, 365–376. tive case series. Neurology, 88, 101–102. Ekstein, D., Ben-Yehuda, D., Slyusarevsky, E., Los-
Abrey, L.E., Moskowitz, C.H., Mason, W.P., Chan, C.C. & Sen, H.N. (2013) Current concepts sos, A., Linetsky, E. & Siegal, T. (2006) CSF
Crump, M., Stewart, D., Forsyth, P., Paleologos, in diagnosing and managing primary vitreoreti- analysis of IgH gene rearrangement in CNS lym-
N., Correa, D.D., Anderson, N.D., Caron, D., nal (intraocular) lymphoma. Discovery Medicine, phoma: relationship to the disease course. Jour-
Zelenetz, A., Nimer, S.D. & DeAngelis, L.M. 15, 93–100. nal of the Neurological Sciences, 247, 39–46.
(2003) Intensive methotrexate and cytarabine Choi, M.K., Kang, E.S., Kim, D.W., Ko, Y.H., Ferreri, A.J.M. (2011) How I treat primary CNS
followed by high-dose chemotherapy with autol- Seok, H., Park, J.H., Pyo, D.H., Hoon Lim, D., lymphoma. Blood, 118, 510–522.
ogous stem-cell rescue in patients with newly Kim, S.J. & Kim, W.S. (2013) Treatment out- Ferreri, A.J., Reni, M., Pasini, F., Calderoni, A.,
diagnosed primary CNS lymphoma: an intent- come of relapsed/refractory primary central ner- Tirelli, U., Pivnik, A., Aondio, G.M., Ferrarese,
to-treat analysis. Journal of Clinical Oncology, 21, vous system diffuse large B-cell lymphoma: a F., Gomez, H., Ponzoni, M., Borisch, B., Berger,
4151–4156. single-center experience of autologous stem cell F., Chassagne, C., Iuzzolino, P., Carbone, A.,
Abrey, L.E., Batchelor, T.T., Ferreri, A.J., Gospo- transplantation. International Journal of Hema- Weis, J., Pedrinis, E., Motta, T., Jouvet, A., Bar-
darowicz, M., Pulczynski, E.J., Zucca, E., Smith, tology, 98, 346–354. bui, T., Cavalli, F. & Blay, J.Y. (2002a) A multi-
J.R., Korfel, A., Soussain, C., DeAngelis, L.M., Choquet, S., Grenier, A., Houillier, C., Soussain, center study of treatment of primary CNS
Neuwelt, E.A., O’Neill, B.P., Thiel, E., Shenkier, C., Moles, M.P., Gastinne, T., Cassoux, N., lymphoma. Neurology, 58, 1513–1520.
T., Graus, F., van den Bent, M., Seymour, J.F., Merle Beral, H., Roos Weil, D., Leblond, V. & Ferreri, A.J., Blay, J.Y., Reni, M., Pasini, F., Gub-
Poortmans, P., Armitage, J.O. & Cavalli, F. Hoang Xuan, K. (2015) Very high efficiency of kin, A., Tirelli, U., Calderoni, A., Zucca, E.,
(2005) Report of an international workshop to ICE (Ifosfamide-Carboplatin-Etoposide) in Cortelazzo, S., Chassagne, C., Tinguely, M., Bor-
standardize baseline evaluation and response cri- relapse/refractory (R/R) primary central nervous isch, B., Berger, F., Ponzoni, M., Cavalli, F. &
teria for primary CNS lymphoma. Journal of system (PCNSL) and vitreo-retinal (VRL) non International Extranodal Lymphoma Study
Clinical Oncology, 23, 5034–5043. Hodgkin lymphoma. A LOC Network Multicen- Group (IELSG). (2002b) Relevance of intraocu-
Aki, H., Uzunaslan, D., Saygin, C., Batur, S., Tuzu- ter Retrospective Study on 58 Cases. Blood, 126, lar involvement in the management of primary
ner, N., Kafadar, A., Ongoren, S. & Oz, B. 1524. central nervous system lymphomas. Annals of
(2013) Primary central nervous system lym- Correa, D.D., Maron, L., Harder, H., Klein, M., Oncology, 13, 531–538.
phoma in immunocompetent individuals: a sin- Armstrong, C.L., Calabrese, P., Bromberg, J.E., Ferreri, A.J., Blay, J.Y., Reni, M., Pasini, F., Spina,
gle center experience. International Journal of Abrey, L.E., Batchelor, T.T. & Schiff, D. (2007) M., Ambrosetti, A., Calderoni, A., Rossi, A.,
Clinical and Experimental Pathology, 6, 1068– Cognitive functions in primary central nervous Vavassori, V., Conconi, A., Devizzi, L., Berger,
1075. system lymphoma: literature review and assess- F., Ponzoni, M., Borisch, B., Tinguely, M., Cer-
Barajas, R.F., Chang, J.S., Segal, M.R., Parsa, A.T., ment guidelines. Annals of Oncology, 18, 1145– ati, M., Milani, M., Orvieto, E., Sanchez, J.,
McDermott, M.W., Berger, M.S. & Cha, S. 1151. Chevreau, C., Dell’Oro, S., Zucca, E. & Cavalli,
(2009) Differentiation of recurrent glioblastoma Correa, D.D., Rocco-Donovan, M., DeAngelis, F. (2003) Prognostic scoring system for primary
multiforme from radiation necrosis after exter- L.M., Dolgoff-Kaspar, R., Iwamoto, F., Yahalom, CNS lymphomas: the International Extranodal
nal beam radiation therapy with dynamic sus- J. & Abrey, L.E. (2009) Prospective cognitive fol- Lymphoma Study Group experience. Journal of
ceptibility-weighted contrast-enhanced perfusion low-up in primary CNS lymphoma patients Clinical Oncology, 21, 266–272.
MR imaging. Radiology, 253, 486–496. treated with chemotherapy and reduced-dose Ferreri, A.J., Guerra, E., Regazzi, M., Pasini, F.,
Barona, A., Reynolds, C.R. & Chastain, R. (1984) radiotherapy. Journal of Neurooncology, 91, 315– Ambrosetti, A., Pivnik, A., Gubkin, A., Calder-
A demographically based index of premorbid 321. oni, A., Spina, M., Brandes, A., Ferrarese, F.,
intelligence for the WAIS-R. Journal of Consult- Coulon, A., Lafitte, F., Hoang-Xuan, K., Martin- Rognone, A., Govi, S., Dell’Oro, S., Locatelli,
ing and Clinical Psychology, 52, 885–887. Duverneuil, N., Mokhtari, K., Blustajn, J. & M., Villa, E. & Reni, M. (2004) Area under the
Batchelor, T.T., Kolak, G., Ciordia, R., Foster, C.S. Chiras, J. (2002) Radiographic findings in 37 curve of methotrexate and creatinine clearance
& Henson, J.W. (2003) High-dose methotrexate cases of primary CNS lymphoma in immuno- are outcome-determining factors in primary
for intraocular lymphoma. Clinical Cancer competent patients. European Radiology, 12, CNS lymphomas. British Journal of Cancer, 90,
Research, 9, 711–715. 329–340. 353–358.
Baumann, M.A., Ritch, P.S., Hande, K.R., Wil- Coupland, S.E., Heimann, H. & Bechrakis, N.E. Ferreri, A.J., Reni, M., Foppoli, M., Martelli, M.,
liams, G.A., Topping, T.M. & Anderson, T. (2004) Primary intraocular lymphoma: a review Pangalis, G.A., Frezzato, M., Cabras, M.G., Fab-
(1986) Treatment of intraocular lymphoma with of the clinical, histopathological and molecular bri, A., Corazzelli, G., Ilariucci, F., Rossi, G.,
high-dose Ara-C. Cancer, 57, 1273–1275. biological features. Graefes Archive for Clinical Soffietti, R., Stelitano, C., Vallisa, D., Zaja, F.,
Benedict, R.H.B., Schretlen, D., Groninger, L. & and Experimental Ophthalmology, 242, 901–913. Zoppegno, L., Aondio, G.M., Avvisati, G., Bal-
Brandt, J. (1998) Hopkins Verbal Learning Test DeAngelis, L.M., Seiferheld, W., Schold, S.C., zarotti, M., Brandes, A.A., Fajardo, J., Gomez,
– Revised: normative data and analysis of inter- Fisher, B. & Schultz, C.J. (2002) Combination H., Guarini, A., Pinotti, G., Rigacci, L., Uhl-
form and test-retest reliability. The Clinical Neu- chemotherapy and radiotherapy for primary mann, C., Picozzi, P., Vezzulli, P., Ponzoni, M.,
ropsychologist, 12, 43–55. central nervous system lymphoma: Radiation Zucca, E., Caligaris-Cappio, F. & Cavalli, F.
Bromberg, J., Issa, S., Bukanina, K., Minnema, Therapy Oncology Group Study 93-10. Journal (2009) High-dose cytarabine plus high-dose
M.C., Seute, T., Durian, M., Cull, G., Schouten, of Clinical Oncology, 20, 4643–4648. methotrexate versus high-dose methotrexate
H.C., Stevens, W.B., Zijlstra, J., Baars, J.W.,

ª 2018 British Society for Haematology and John Wiley & Sons Ltd 359
British Journal of Haematology, 2019, 184, 348–363
Guideline

alone in patients with primary CNS lymphoma: Ihorst, G., Finke, J. & Illerhaus, G. (2017) High- Treatment of Cancer Brain Tumor Group. Jour-
a randomised phase 2 trial. Lancet, 374, 1512– dose methotrexate-based immuno-chemotherapy nal of Clinical Oncology, 21, 2726–2731.
1520. for elderly primary CNS lymphoma patients Hoang-Xuan, K., Bessell, E., Bromberg, J., Hot-
Ferreri, A.J., Cwynarski, K., Pulczynski, E., Pon- (PRIMAIN study). Leukemia, 31, 846–852. tinger, A.F., Preusser, M., Ruda, R., Schlegel, U.,
zoni, M., Deckert, M., Politi, L.S., Torri, V., Ghesquieres, H., Houillier, C., Chinot, O., Cho- Siegal, T., Soussain, C., Abacioglu, U., Cassoux,
Fox, C.P., Rosee, P.L., Schorb, E., Ambrosetti, quet, S., Molucon-Chabrot, C., Beauchene, P., N., Deckert, M., Dirven, C.M., Ferreri, A.J.,
A., Roth, A., Hemmaway, C., Ferrari, A., Linton, Gressin, R., Morschhauser, F., Schmitt, A., Graus, F., Henriksson, R., Herrlinger, U.,
K.M., Ruda, R., Binder, M., Pukrop, T., Balzar- Gyan, E., Hoang-Xuan, K., Nicolas-Virelizier, E., Taphoorn, M., Soffietti, R., Weller, M. & for the
otti, M., Fabbri, A., Johnson, P., Gorlov, J.S., Chevrier, M., Savignoni, A., Turbiez, I., Veillas, European Association for Neuro-Oncology Task
Hess, G., Panse, J., Pisani, F., Tucci, A., Stilgen- F., Soumelis, V. & Soussain, C. (2016) Ritux- Force on Primary CNSL. (2015) Diagnosis and
bauer, S., Hertenstein, B., Keller, U., Krause, imab-Lenalidomide (REVRI) in relapse or treatment of primary CNS lymphoma in
S.W., Levis, A., Schmoll, H.J., Cavalli, F., Finke, refractory primary central nervous system immunocompetent patients: guidelines from the
J., Reni, M., Zucca, E. & Illerhaus, G. (2016) (PCNSL) or vitreo retinal lymphoma (PVRL): European Association for Neuro-Oncology. Lan-
Chemoimmunotherapy with methotrexate, results of a “Proof of Concept” Phase II Study cet Oncology, 16, e322–e332.
cytarabine, thiotepa, and rituximab (MATRix of the French LOC Network. Blood, 128, 785. Hottinger, A.F., DeAngelis, L.M., Yahalom, J. &
regimen) in patients with primary CNS lym- Glass, J., Won, M., Schultz, C.J., Brat, D., Bartlett, Abrey, L.E. (2007) Salvage whole brain radio-
phoma: results of the first randomisation of the N.L., Suh, J.H., Werner-Wasik, M., Fisher, B.J., therapy for recurrent or refractory primary CNS
International Extranodal Lymphoma Study Liepman, M.K., Augspurger, M., Bokstein, F., lymphoma. Neurology, 69, 1178–1182.
Group-32 (IELSG32) phase 2 trial. Lancet Hae- Bovi, J.A., Solhjem, M.C. & Mehta, M.P. (2016) Houillier, C., Taillandier, L., Lamy, T., Chinot, O.,
matology, 3, e217–e227. Phase I and II study of induction chemotherapy Molucon-Chabrot, C., Soubeyran, P., Gressin,
Ferreri, A.J.M., Cwynarski, K., Pulczynski, E., Fox, with methotrexate, rituximab, and temozolo- R., Choquet, S., Damaj, G., Thyss, A., Jaccard,
C.P., Schorb, E., La Rosee, P., Binder, M., Fab- mide, followed by whole-brain radiotherapy and A., Delwail, V., Gyan, E., Sanhes, L., Cornillon,
bri, A., Torri, V., Minacapelli, E., Falautano, M., postirradiation temozolomide for primary CNS J., Garidi, R., Delmer, A., Savignoni, A., Jijakli,
Ilariucci, F., Ambrosetti, A., Roth, A., Hemm- lymphoma: NRG Oncology RTOG 0227. Journal A.L.A., Morel, P., Bourquard, P., Moles, M.-P.,
away, C., Johnson, P., Linton, K.M., Pukrop, T., of Clinical Oncology, 34, 1620–1625. Deconinck, E., Morel, P., Gastinne, T., Con-
Sønderskov Gørløv, J., Balzarotti, M., Hess, G., Grimm, S.A., Pulido, J.S., Jahnke, K., Schiff, D., stans, J.-M., Langer, A., Lacomblez, L., Del-
Keller, U., Stilgenbauer, S., Panse, J., Tucci, A., Hall, A.J., Shenkier, T.N., Siegal, T., Doolittle, gadillo, D., Dureau, S., Turbiez, I., Plissonnier,
Orsucci, L., Pisani, F., Levis, A., Krause, S.W., N.D., Batchelor, T., Herrlinger, U., Neuwelt, A.-S., Cassoux, N., Touitou, V., Ricard, D.,
Schmoll, H.J., Hertenstein, B., Rummel, M., E.A., Laperriere, N., Chamberlain, M.C., Blay, Hoang-Xuan, K. & Soussain, C. (2016) Whole
Smith, J., Pfreundschuh, M., Cabras, G., J.Y., Ferreri, A.J., Omuro, A.M., Thiel, E. & brain radiotherapy (WBRT) versus intensive
Angrilli, F., Ponzoni, M., Deckert, M., Politi, Abrey, L.E. (2007) Primary intraocular lym- chemotherapy with haematopoietic stem cell
L.S., Finke, J., Reni, M., Cavalli, F., Zucca, E. & phoma: an International Primary Central Ner- rescue (IC + HCR) for primary central nervous
Illerhaus, G. (2017) Whole-brain radiotherapy vous System Lymphoma Collaborative Group system lymphoma (PCNSL) in young patients:
or autologous stem-cell transplantation as con- Report. Annals of Oncology, 18, 1851–1855. an Intergroup Anocef-Goelams Randomized
solidation strategies after high-dose methotrex- Grommes, C. & DeAngelis, L.M. (2017) Primary Phase II Trial (PRECIS). Blood, 128, 782.
ate-based chemoimmunotherapy in patients CNS lymphoma. Journal of Clinical Oncology, Illerhaus, G., Marks, R., Ihorst, G., Guttenberger,
with primary CNS lymphoma: results of the sec- 35, 2410–2418. R., Ostertag, C., Derigs, G., Frickhofen, N.,
ond randomisation of the International Extran- Grommes, C., Pastore, A., Palaskas, N., Tang, S.S., Feuerhake, F., Volk, B. & Finke, J. (2006) High-
odal Lymphoma Study Group-32 phase 2 trial. Campos, C., Schartz, D., Codega, P., Nichol, D., dose chemotherapy with autologous stem-cell
Lancet Haematology, 4, e510–e523. Clark, O., Hsieh, W.Y., Rohle, D., Rosenblum, transplantation and hyperfractionated radiother-
Fossard, G., Ferlay, C., Nicolas-Virelizier, E., Rey, M., Viale, A., Tabar, V.S., Brennan, C.W., Gavri- apy as first-line treatment of primary CNS lym-
P., Ducray, F., Jouanneau, E., Faurie, P., Bel- lovic, I.T., Kaley, T.J., Nolan, C.P., Omuro, A., phoma. Journal of Clinical Oncology, 24, 3865–
habri, A., Sunyack, M.P., Chassagne-Clement, Pentsova, E., Thomas, A.A., Tsyvkin, E., Noy, A., 3870.
C., Thiesse, P., Sebban, C., Biron, P., Blay, J.Y. Palomba, M.L., Hamlin, P., Sauter, C.S., Illerhaus, G., Marks, R., Muller, F., Ihorst, G.,
& Ghesquieres, H. (2017) Utility of post-therapy Moskowitz, C.H., Wolfe, J., Dogan, A., Won, M., Feuerhake, F., Deckert, M., Ostertag, C. & Finke,
brain surveillance imaging in the detection of Glass, J., Peak, S., Lallana, E.C., Hatzoglou, V., J. (2009) High-dose methotrexate combined
primary central nervous system lymphoma Reiner, A.S., Gutin, P.H., Huse, J.T., Panageas, with procarbazine and CCNU for primary CNS
relapse. European Journal of Cancer, 72, 12–19. K.S., Graeber, T.G., Schultz, N., DeAngelis, L.M. lymphoma in the elderly: results of a prospective
Frenkel, S., Hendler, K., Siegal, T., Shalom, E. & & Mellinghoff, I.K. (2017) Ibrutinib unmasks pilot and phase II study. Annals of Oncology, 20,
Pe’er, J. (2008) Intravitreal methotrexate for critical role of bruton tyrosine kinase in primary 319–325.
treating vitreoretinal lymphoma: 10 years of CNS lymphoma. Cancer Discovery, 7, 1018–1029. Illerhaus, G., Kasenda, B., Ihorst, G., Egerer, G.,
experience. British Journal of Ophthalmology, 92, Haldorsen, I.S., Espeland, A. & Larsson, E.M. Lamprecht, M., Keller, U., Wolf, H.H., Hirt, C.,
383–388. (2011) Central nervous system lymphoma: char- Stilgenbauer, S., Binder, M., Hau, P., Edinger,
Fritsch, K., Kasenda, B., Hader, C., Nikkhah, G., acteristic findings on traditional and advanced M., Frickhofen, N., Bentz, M., Mohle, R., Roth,
Prinz, M., Haug, V., Haug, S., Ihorst, G., Finke, J. imaging. American Journal of Neuroradiology, 32, A., Pfreundschuh, M., von Baumgarten, L.,
& Illerhaus, G. (2011) Immunochemotherapy 984–992. Deckert, M., Hader, C., Fricker, H., Valk, E.,
with rituximab, methotrexate, procarbazine, and Hoang-Xuan, K., Taillandier, L., Chinot, O., Sou- Schorb, E., Fritsch, K. & Finke, J. (2016)
lomustine for primary CNS lymphoma (PCNSL) beyran, P., Bogdhan, U., Hildebrand, J., Frenay, High-dose chemotherapy with autologous
in the elderly. Annals of Oncology, 22, 2080–2085. M., De Beule, N., Delattre, J.Y., Baron, B. & haemopoietic stem cell transplantation for newly
Fritsch, K., Kasenda, B., Schorb, E., Hau, P., Bloe- European Organization for Research and Treat- diagnosed primary CNS lymphoma: a prospec-
hdorn, J., Mohle, R., Low, S., Binder, M., Atta, J., ment of Cancer Brain Tumor Group. (2003) tive, single-arm, phase 2 trial. Lancet Haematol-
Keller, U., Wolf, H.H., Krause, S.W., Hess, G., Chemotherapy alone as initial treatment for pri- ogy, 3, e388–e397.
Naumann, R., Sasse, S., Hirt, C., Lamprecht, M., mary CNS lymphoma in patients older than Jahnke, K., Thiel, E., Bechrakis, N.E., Willerding,
Martens, U., Morgner, A., Panse, J., Frickhofen, 60 years: a multicenter phase II study (26952) of G., Kraemer, D.F., Fischer, L. & Korfel, A.
N., Roth, A., Hader, C., Deckert, M., Fricker, H., the European Organization for Research and (2009) Ifosfamide or trofosfamide in patients

360 ª 2018 British Society for Haematology and John Wiley & Sons Ltd
British Journal of Haematology, 2019, 184, 348–363
Guideline

with intraocular lymphoma. Journal of Neuroon- G-PCNSL-SG1 trial. Annals of Oncology, 23, Ma, W.L., Hou, H.A., Hsu, Y.J., Chen, Y.K., Tang,
cology, 93, 213–217. 2374–2380. J.L., Tsay, W., Yeh, P.T., Yang, C.M., Lin, C.P.
Kasenda, B., Schorb, E., Fritsch, K., Finke, J. & Korfel, A., Thiel, E., Martus, P., Mohle, R., Grie- & Tien, H.F. (2016) Clinical outcomes of pri-
Illerhaus, G. (2012) Prognosis after high-dose singer, F., Rauch, M., Roth, A., Hertenstein, B., mary intraocular lymphoma patients treated
chemotherapy followed by autologous stem-cell Fischer, T., Hundsberger, T., Mergenthaler, with front-line systemic high-dose methotrexate
transplantation as first-line treatment in primary H.G., Junghanss, C., Birnbaum, T., Fischer, L., and intravitreal methotrexate injection. Annals
CNS lymphoma–a long-term follow-up study. Jahnke, K., Herrlinger, U., Roth, P., Bamberg, of Hematology, 95, 593–601.
Annals of Oncology, 23, 2670–2675. M., Pietsch, T. & Weller, M. (2015) Randomized Manoj, N., Arivazhagan, A., Mahadevan, A., Bhat,
Kasenda, B., Ferreri, A.J., Marturano, E., Forst, D., phase III study of whole-brain radiotherapy for D.I., Arvinda, H.R., Devi, B.I., Sampath, S. &
Bromberg, J., Ghesquieres, H., Ferlay, C., Blay, primary CNS lymphoma. Neurology, 84, 1242– Chandramouli, B.A. (2014) Central nervous sys-
J.Y., Hoang-Xuan, K., Pulczynski, E.J., Fossa, A., 1248. tem lymphoma: patterns of incidence in Indian
Okoshi, Y., Chiba, S., Fritsch, K., Omuro, A., Kurzwelly, D., Glas, M., Roth, P., Weimann, E., population and effect of steroids on stereotactic
O’Neill, B.P., Bairey, O., Schandelmaier, S., Lohner, H., Waha, A., Schabet, M., Reifenberger, biopsy yield. Neurology India, 62, 19–25.
Gloy, V., Bhatnagar, N., Haug, S., Rahner, S., G., Weller, M. & Herrlinger, U. (2010) Primary Mappa, S., Marturano, E., Licata, G., Frezzato, M.,
Batchelor, T.T., Illerhaus, G. & Briel, M. (2015) CNS lymphoma in the elderly: temozolomide Frungillo, N., Ilariucci, F., Stelitano, C., Ferrari,
First-line treatment and outcome of elderly therapy and MGMT status. Journal of Neuroon- A., Soraru, M., Vianello, F., Baldini, L., Proser-
patients with primary central nervous system cology, 97, 389–392. pio, I., Foppoli, M., Assanelli, A., Reni, M., Cali-
lymphoma (PCNSL) – a systematic review and Lai, R., Rosenblum, M.K. & DeAngelis, L.M. garis-Cappio, F. & Ferreri, A.J. (2013) Salvage
individual patient data meta-analysis. Annals of (2002) Primary CNS lymphoma: a whole-brain chemoimmunotherapy with rituximab, ifos-
Oncology, 26, 1305–1313. disease? Neurology, 59, 1557–1562. famide and etoposide (R-IE regimen) in patients
Kasenda, B., Ihorst, G., Schroers, R., Korfel, A., Langerak, A.W., Groenen, P.J., Bruggemann, M., with primary CNS lymphoma relapsed or refrac-
Schmidt-Wolf, I., Egerer, G., von Baumgarten, Beldjord, K., Bellan, C., Bonello, L., Boone, E., tory to high-dose methotrexate-based
L., Roth, A., Bloehdorn, J., Mohle, R., Binder, Carter, G.I., Catherwood, M., Davi, F., Delfau- chemotherapy. Hematological Oncology, 31, 143–
M., Keller, U., Lamprecht, M., Pfreundschuh, Larue, M.H., Diss, T., Evans, P.A., Gameiro, P., 150.
M., Valk, E., Fricker, H., Schorb, E., Fritsch, K., Garcia Sanz, R., Gonzalez, D., Grand, D., Mohile, N.A., Deangelis, L.M. & Abrey, L.E.
Finke, J. & Illerhaus, G. (2017) High-dose Hakansson, A., Hummel, M., Liu, H., Lombar- (2008) The utility of body FDG PET in staging
chemotherapy with autologous haematopoietic dia, L., Macintyre, E.A., Milner, B.J., Montes- primary central nervous system lymphoma.
stem cell support for relapsed or refractory pri- Moreno, S., Schuuring, E., Spaargaren, M., Neuro Oncology, 10, 223–228.
mary CNS lymphoma: a prospective multicentre Hodges, E. & van Dongen, J.J. (2012) EuroClon- Morris, P.G., Correa, D.D., Yahalom, J., Raizer,
trial by the German Cooperative PCNSL study ality/BIOMED-2 guidelines for interpretation J.J., Schiff, D., Grant, B., Grimm, S., Lai, R.K.,
group. Leukemia, 31, 2623–2629. and reporting of Ig/TCR clonality testing in sus- Reiner, A.S., Panageas, K., Karimi, S., Curry, R.,
Kassam, S., Chernucha, E., O’Neill, A., Hemm- pected lymphoproliferations. Leukemia, 26, Shah, G., Abrey, L.E., DeAngelis, L.M. &
away, C., Cummins, T., Montoto, S., Lennard, 2159–2171. Omuro, A. (2013) Rituximab, methotrexate,
A., Adams, G., Linton, K., McKay, P., Davies, Langner-Lemercier, S., Houillier, C., Soussain, C., procarbazine, and vincristine followed by con-
D., Rowntree, C., Easdale, S., Eyre, T.A., Mar- Ghesquieres, H., Chinot, O., Taillandier, L., solidation reduced-dose whole-brain radiother-
cus, R., Cwynarski, K. & Fox, C.P. (2017) High- Soubeyran, P., Lamy, T., Morschhauser, F., apy and cytarabine in newly diagnosed primary
dose chemotherapy and autologous stem cell Benouaich-Amiel, A., Ahle, G., Moles-Moreau, CNS lymphoma: final results and long-term out-
transplantation for primary central nervous sys- M.P., Molucon-Chabrot, C., Bourquard, P., come. Journal of Clinical Oncology, 31, 3971–
tem lymphoma: a multi-centre retrospective Damaj, G., Jardin, F., Larrieu, D., Gyan, E., 3979.
analysis from the United Kingdom. Bone Mar- Gressin, R., Jaccard, A., Choquet, S., Brion, A., Motomura, K., Natsume, A., Fujii, M., Ito, M.,
row Transplantation, 52, 1268–1272. Casasnovas, O., Colin, P., Reman, O., Tempes- Momota, H. & Wakabayashi, T. (2011) Long-
Kawai, N., Miyake, K., Yamamoto, Y., Nishiyama, cul, A., Marolleau, J.P., Fabbro, M., Naudet, F., term survival in patients with newly diagnosed
Y. & Tamiya, T. (2013) 18F-FDG PET in the Hoang-Xuan, K. & Houot, R. (2016) Primary primary central nervous system lymphoma trea-
diagnosis and treatment of primary central ner- CNS lymphoma at first relapse/progression: ted with dexamethasone, etoposide, ifosfamide
vous system lymphoma. BioMed Research Inter- characteristics, management, and outcome of and carboplatin chemotherapy and whole-brain
national, 2013, 247152. 256 patients from the French LOC network. radiation therapy. Leukemia & Lymphoma, 52,
Khan, R.B., Shi, W., Thaler, H.T., DeAngelis, Neuro Oncology, 18, 1297–1303. 2069–2075.
L.M. & Abrey, L.E. (2002) Is intrathecal Larkin, K.L., Saboo, U.S., Comer, G.M., Foroo- Mylam, K.J., Michaelsen, T.Y., Hutchings, M.,
methotrexate necessary in the treatment of ghian, F., Mackensen, F., Merrill, P., Sen, H.N., Jacobsen Pulczynski, E., Pedersen, L.M.,
primary CNS lymphoma? Journal of Neuroon- Singh, A., Essex, R.W., Lake, S., Lim, L.L., Brændstrup, P., Gade, I.L., Eberlein, T.R., Gang,
cology, 58, 175–178. Vasconcelos-Santos, D.V., Foster, C.S., Wilson, A.O., Bøgsted, M., Brown, P.D.N. & El-Galaly,
Khimani, N.B., Ng, A.K., Chen, Y.H., Catalano, P., D.J. & Smith, J.R. (2014) Use of intravitreal T.C. (2017) Little value of surveillance magnetic
Silver, B. & Mauch, P.M. (2011) Salvage radio- rituximab for treatment of vitreoretinal lym- resonance imaging for primary CNS lymphomas
therapy in patients with recurrent or refractory phoma. British Journal of Ophthalmology, 98, in first remission: results from a Danish Multi-
primary or secondary central nervous system 99–103. centre Study. British Journal of Haematology,
lymphoma after methotrexate-based chemother- Lionakis, M.S., Dunleavy, K., Roschewski, M., 176, 671–673.
apy. Annals of Oncology, 22, 979–984. Widemann, B.C., Butman, J.A., Schmitz, R., Nayak, L., Iwamoto, F.M., LaCasce, A., Mukundan,
Korfel, A., Weller, M., Martus, P., Roth, P., Klasen, Yang, Y., Cole, D.E., Melani, C., Higham, C.S., S., Roemer, M.G.M., Chapuy, B., Armand, P.,
H.A., Roeth, A., Rauch, M., Hertenstein, B., Fis- Desai, J.V., Ceribelli, M., Chen, L., Thomas, Rodig, S.J. & Shipp, M.A. (2017) PD-1 blockade
cher, T., Hundsberger, T., Leithauser, M., Birn- C.J., Little, R.F., Gea-Banacloche, J., Bhaumik, with nivolumab in relapsed/refractory primary
baum, T., Kirchen, H., Mergenthaler, H.G., S., Stetler-Stevenson, M., Pittaluga, S., Jaffe, E.S., central nervous system and testicular lymphoma.
Schubert, J., Berdel, W., Birkmann, J., Hummel, Heiss, J., Lucas, N., Steinberg, S.M., Staudt, Blood, 129, 3071–3073.
M., Thiel, E. & Fischer, L. (2012) Prognostic L.M. & Wilson, W.H. (2017) Inhibition of B cell Nelson, H.E. & Willison, J. (1991) National Adult
impact of meningeal dissemination in primary receptor signaling by ibrutinib in primary CNS Reading Test (NART): Test Manual, 2nd edn.
CNS lymphoma (PCNSL): experience from the lymphoma. Cancer Cell, 31, e835. NFER Nelson, Windsor.

ª 2018 British Society for Haematology and John Wiley & Sons Ltd 361
British Journal of Haematology, 2019, 184, 348–363
Guideline

Nelson, D.F., Martz, K.L., Bonner, H., Nelson, J.S., Pels, H., Juergens, A., Glasmacher, A., Schulz, H., Figarella-Branger, D., Costes, V. & Bauchet, L.
Newall, J., Kerman, H.D., Thomson, J.W. & Mur- Engert, A., Linnebank, M., Schackert, G., Reich- (2011) French brain tumor database: 5-year his-
ray, K.J. (1992) Non-Hodgkin’s lymphoma of the mann, H., Kroschinsky, F., Vogt-Schaden, M., tological results on 25 756 cases. Brain Pathol-
brain: can high dose, large volume radiation ther- Egerer, G., Bode, U., Schaller, C., Lamprecht, ogy, 21, 633–644.
apy improve survival? Report on a prospective trial M., Hau, P., Deckert, M., Fimmers, R., Bangard, Roth, P., Martus, P., Kiewe, P., Mohle, R., Klasen,
by the Radiation Therapy Oncology Group C., Schmidt-Wolf, I.G. & Schlegel, U. (2009) H., Rauch, M., Roth, A., Kaun, S., Thiel, E., Kor-
(RTOG): RTOG 8315. International Journal of Early relapses in primary CNS lymphoma after fel, A. & Weller, M. (2012) Outcome of elderly
Radiation Oncology Biology Physics, 23, 9–17. response to polychemotherapy without intraven- patients with primary CNS lymphoma in the G-
Nguyen, P.L., Chakravarti, A., Finkelstein, D.M., tricular treatment: results of a phase II study. PCNSL-SG-1 trial. Neurology, 79, 890–896.
Hochberg, F.H., Batchelor, T.T. & Loeffler, J.S. Journal of Neurooncology, 91, 299–305. Rubenstein, J.L., Hsi, E.D., Johnson, J.L., Jung,
(2005) Results of whole-brain radiation as Pels, H., Juergens, A., Schirgens, I., Glasmacher, S.H., Nakashima, M.O., Grant, B., Cheson, B.D.
salvage of methotrexate failure for immuno- A., Schulz, H., Engert, A., Schackert, G., Reich- & Kaplan, L.D. (2013) Intensive chemotherapy
competent patients with primary CNS mann, H., Kroschinsky, F., Vogt-Schaden, M., and immunotherapy in patients with newly
lymphoma. Journal of Clinical Oncology, 23, Egerer, G., Bode, U., Deckert, M., Fimmers, R., diagnosed primary CNS lymphoma: CALGB
1507–1513. Urbach, H., Schmidt-Wolf, I.G. & Schlegel, U. 50202 (Alliance 50202). Journal of Clinical
Nguyen, D.T., Houillier, C., Choquet, S., Cassoux, (2010) Early complete response during Oncology, 31, 3061–3068.
N., Soussain, C., Le Cossec, C., Legarf-Tavernier, chemotherapy predicts favorable outcome in Rubenstein, J.L., Fraser, E., Formaker, P., Lee, J.C.-
M., Costopoulos, M., LeHoang, P., Bodaghi, B., patients with primary CNS lymphoma. Neuro C., Chen, N., Kock, M., Cheung, W., Wang, X.,
Omuro, A., Hoang-Xuan, K. & Touitou, V. Oncology, 12, 720–724. Munster, P.N. & Damato, B. (2016) Phase I
(2016) Primary oculocerebral lymphoma: MTX Pentsova, E., Deangelis, L.M. & Omuro, A. (2014) investigation of lenalidomide plus rituximab and
polychemotherapy alone on intraocular disease Methotrexate re-challenge for recurrent primary outcomes of lenalidomide maintenance in recur-
control. Ophthalmology, 123, 2047–2050. central nervous system lymphoma. Journal of rent CNS lymphoma. Journal of Clinical Oncol-
Omuro, A., Chinot, O., Taillandier, L., Ghes- Neurooncology, 117, 161–165. ogy, 34, 7502.
quieres, H., Soussain, C., Delwail, V., Lamy, T., Plotkin, S.R., Betensky, R.A., Hochberg, F.H., Russell, E.W. & Starkey, R.I. (1993) Halstead Rus-
Gressin, R., Choquet, S., Soubeyran, P., Huchet, Grossman, S.A., Lesser, G.J., Nabors, L.B., Chon, sell Neuropsychological Evaluation System
A., Benouaich-Amiel, A., Lebouvier-Sadot, S., B. & Batchelor, T.T. (2004) Treatment of (HNRES). Western Psychological Services, Los
Gyan, E., Touitou, V., Barrie, M., del Rio, M.S., relapsed central nervous system lymphoma with Angeles.
Gonzalez-Aguilar, A., Houillier, C., Delgadillo, high-dose methotrexate. Clinical Cancer Schorb, E., Kasenda, B., Atta, J., Kaun, S.,
D., Lacomblez, L., Tanguy, M.L. & Hoang-Xuan, Research, 10, 5643–5646. Morgner, A., Hess, G., Elter, T., von Bubnoff,
K. (2015a) Methotrexate and temozolomide ver- Prica, A., Chan, K. & Cheung, M.C. (2012) Com- N., Dreyling, M., Ringhoffer, M., Krause, S.W.,
sus methotrexate, procarbazine, vincristine, and bined modality therapy versus chemotherapy Derigs, G., Klimm, B., Niemann, D., Fritsch, K.,
cytarabine for primary CNS lymphoma in an alone as an induction regimen for primary cen- Finke, J. & Illerhaus, G. (2013) Prognosis of
elderly population: an intergroup ANOCEF- tral nervous system lymphoma: a decision analy- patients with primary central nervous system
GOELAMS randomised phase 2 trial. Lancet sis. British Journal of Haematology, 158, 600– lymphoma after high-dose chemotherapy fol-
Haematology, 2, e251–e259. 607. lowed by autologous stem cell transplantation.
Omuro, A., Correa, D.D., DeAngelis, L.M., Pulczynski, E.J., Kuittinen, O., Erlanson, M., Hag- Haematologica, 98, 765–770.
Moskowitz, C.H., Matasar, M.J., Kaley, T.J., berg, H., Fossa, A., Eriksson, M., Nordstrom, Schorb, E., Fox, C.P., Fritsch, K., Isbell, L., Neu-
Gavrilovic, I.T., Nolan, C., Pentsova, E., M., Ostenstad, B., Fluge, O., Leppa, S., Fiir- bauer, A., Tzalavras, A., Witherall, R., Choquet,
Grommes, C.C., Panageas, K.S., Baser, R.E., gaard, B., Bersvendsen, H. & Fagerli, U.M. S., Kuittinen, O., De-Silva, D., Cwynarski, K.,
Faivre, G., Abrey, L.E. & Sauter, C.S. (2015b) R- (2015) Successful change of treatment strategy Houillier, C., Hoang-Xuan, K., Touitou, V.,
MPV followed by high-dose chemotherapy with in elderly patients with primary central nervous Cassoux, N., Marolleau, J.P., Tamburini, J.,
TBC and autologous stem-cell transplant for system lymphoma by de-escalating induction Houot, R., Delwail, V., Illerhaus, G., Soussain,
newly diagnosed primary CNS lymphoma. and introducing temozolomide maintenance: C. & Kasenda, B. (2017a) High-dose thiotepa-
Blood, 125, 1403–1410. results from a phase II study by the Nordic based chemotherapy with autologous stem cell
Osoba, D., Aaronson, N.K., Muller, M., Sneeuw, Lymphoma Group. Haematologica, 100, 534– support in elderly patients with primary central
K., Hsu, M.A., Yung, W.K.A., Brada, M. & New- 540. nervous system lymphoma: a European retro-
lands, E. (1996) The development and Reitan, R.M. & Wolfson, D. (1985) The Halstead- spective study. Bone Marrow Transplantation,
psychometric validation of a brain cancer qual- Reitan Neuropsychological Test Battery. Neu- 52, 1113–1119.
ity-of-life questionnaire for use in combination ropsychology Press, Tucson. Schorb, E., Fox, C.P., Kasenda, B., Calimeri, T.,
with general cancer-specific questionnaires. Riemens, A., Bromberg, J., Touitou, V., Sobolewska, Linton, K., Smith, J., Ninkovic, S., Yallop, D.,
Quality of Life Research, 5, 139–150. B., Missotten, T., Baarsma, S., Hoyng, C., Cor- Holl, H.-G., Fabbri, A., Cummin, T.E.C., Iller-
Patrick, L.B. & Mohile, N.A. (2015) Advances in dero-Coma, M., Tomkins-Netzer, O., Rozalski, haus, G., Cwynarski, K. & Ferreri, A.J.M.
primary central nervous system lymphoma. Cur- A., Tugal-Tutkun, I., Guex-Crosier, Y., Los, L.I., (2017b) Induction chemo-immunotherapy with
rent Oncology Reports, 17, 60. Bollemeijer, J.G., Nolan, A., Pawade, J., Willer- the matrix regimen in patients with newly diag-
Pels, H., Schmidt-Wolf, I.G., Glasmacher, A., main, F., Bodaghi, B., ten Dam-van Loon, N., nosed PCNSL – a multicenter retrospective anal-
Schulz, H., Engert, A., Diehl, V., Zellner, A., Dick, A., Zierhut, M., Lightman, S., Mackensen, ysis on feasibility and effectiveness in routine
Schackert, G., Reichmann, H., Kroschinsky, F., F., Moulin, A., Erckens, R., Wensing, B., le clinical practice. Blood, 130, 376.
Vogt-Schaden, M., Egerer, G., Bode, U., Schaller, Hoang, P., Lokhorst, H. & Rothova, A. (2015) Schroers, R., Baraniskin, A., Heute, C., Vorgerd,
C., Deckert, M., Fimmers, R., Helmstaedter, C., Treatment strategies in primary vitreoretinal lym- M., Brunn, A., Kuhnhenn, J., Kowoll, A., Alek-
Atasoy, A., Klockgether, T. & Schlegel, U. phoma: a 17-center European collaborative study. seyev, A., Schmiegel, W., Schlegel, U., Deckert,
(2003) Primary central nervous system lym- JAMA Ophthalmology, 133, 191–197. M. & Pels, H. (2010) Diagnosis of lep-
phoma: results of a pilot and phase II study of Rigau, V., Zouaoui, S., Mathieu-Daude, H., Darlix, tomeningeal disease in diffuse large B-cell lym-
systemic and intraventricular chemotherapy with A., Maran, A., Tretarre, B., Bessaoud, F., Bau- phomas of the central nervous system by flow
deferred radiotherapy. Journal of Clinical Oncol- chet, F., Attaoua, R., Fabbro-Peray, P., Fabbro, cytometry and cytopathology. European Journal
ogy, 21, 4489–4495. M., Kerr, C., Taillandier, L., Duffau, H., of Haematology, 85, 520–528.

362 ª 2018 British Society for Haematology and John Wiley & Sons Ltd
British Journal of Haematology, 2019, 184, 348–363
Guideline

Siegel, M.J., Dalton, J., Friedman, A.H., Strauchen, mary central nervous system lymphoma and Wechsler, D. (2008) Wechsler Adult Intelligence
J. & Watson, C. (1989) Ten-year experience with intraocular lymphoma: a retrospective study of Scale (WAIS-IV). Pearson, San Antonio.
primary ocular ‘reticulum cell sarcoma’ (large 79 cases. Haematologica, 97, 1751–1756. Wechsler, D. (2011) Test of Premorbid Function-
cell non-Hodgkin’s lymphoma). British Journal Swerdlow, S.H., Campo, E., Harris, N.L., Jaffe, ing, UK version (TOPF UK). Pearson Assess-
of Ophthalmology, 73, 342–346. E.S., Pileri, S.A., Stein, H., Thiele, J. & Vardi- ment, London.
Sierra Del Rio, M., Ricard, D., Houillier, C., Navarro, man, J.W. (2008) WHO Classification of Welch, M.R., Omuro, A. & Deangelis, L.M. (2012)
S., Gonzalez-Aguilar, A., Idbaih, A., Kaloshi, G., Tumours of Haematopoietic and Lymphoid Tis- Outcomes of the oldest patients with primary
Elhallani, S., Omuro, A., Choquet, S., Soussain, C. sues. IARC, Lyon. CNS lymphoma treated at Memorial Sloan-
& Hoang-Xuan, K. (2012) Prophylactic intrathecal Swerdlow, S.H., Campo, E., Pileri, S.A., Harris, Kettering Cancer Center. Neuro Oncology, 14,
chemotherapy in primary CNS lymphoma. Journal N.L., Stein, H., Siebert, R., Advani, R., Ghiel- 1304–1311.
of Neurooncology, 106, 143–146. mini, M., Salles, G.A., Zelenetz, A.D. & Jaffe, Weller, M., Martus, P., Roth, P., Thiel, E. & Kor-
Soussain, C., Suzan, F., Hoang-Xuan, K., Cassoux, E.S. (2016) The 2016 revision of the World fel, A. (2012) Surgery for primary CNS lym-
N., Levy, V., Azar, N., Belanger, C., Achour, E., Health Organization classification of lymphoid phoma? Challenging a paradigm. Neuro
Ribrag, V., Gerber, S., Delattre, J.Y. & Leblond, neoplasms. Blood, 127, 2375–2390. Oncology, 14, 1481–1484.
V. (2001) Results of intensive chemotherapy fol- Tang, Y.Z., Booth, T.C., Bhogal, P., Malhotra, A. Wieduwilt, M.J., Valles, F., Issa, S., Behler, C.M.,
lowed by hematopoietic stem-cell rescue in 22 & Wilhelm, T. (2011) Imaging of primary cen- Hwang, J., McDermott, M., Treseler, P.,
patients with refractory or recurrent primary tral nervous system lymphoma. Clinical Radiol- O’Brien, J., Shuman, M.A., Cha, S., Damon, L.E.
CNS lymphoma or intraocular lymphoma. Jour- ogy, 66, 768–777. & Rubenstein, J.L. (2012) Immunochemotherapy
nal of Clinical Oncology, 19, 742–749. Thiel, E., Korfel, A., Martus, P., Kanz, L., Grie- with intensive consolidation for primary CNS
Soussain, C., Hoang-Xuan, K., Taillandier, L., singer, F., Rauch, M., Roth, A., Hertenstein, B., lymphoma: a pilot study and prognostic assess-
Fourme, E., Choquet, S., Witz, F., Casasnovas, von Toll, T., Hundsberger, T., Mergenthaler, ment by diffusion-weighted MRI. Clinical Can-
O., Dupriez, B., Souleau, B., Taksin, A.L., Gis- H.G., Leithauser, M., Birnbaum, T., Fischer, L., cer Research, 18, 1146–1155.
selbrecht, C., Jaccard, A., Omuro, A., Sanson, Jahnke, K., Herrlinger, U., Plasswilm, L., Nagele, Zaki, H.S., Jenkinson, M.D., Du Plessis, D.G.,
M., Janvier, M., Kolb, B., Zini, J.M., Leblond, T., Pietsch, T., Bamberg, M. & Weller, M. Smith, T. & Rainov, N.G. (2004) Vanishing con-
V. & Societe Francßaise de Greffe de Mo€elle (2010) High-dose methotrexate with or without trast enhancement in malignant glioma after
Osseuse-Therapie Cellulaire. (2008) Intensive whole brain radiotherapy for primary CNS lym- corticosteroid treatment. Acta Neurochir (Wien),
chemotherapy followed by hematopoietic stem- phoma (G-PCNSL-SG-1): a phase 3, ran- 146, 841–845.
cell rescue for refractory and recurrent primary domised, non-inferiority trial. Lancet Oncology, Zhang, D., Hu, L.B., Henning, T.D., Ravarani,
CNS and intraocular lymphoma: Societe Fran- 11, 1036–1047. E.M., Zou, L.G., Feng, X.Y., Wang, W.X. &
caise de Greffe de Moelle Osseuse-Therapie Valles, F.E., Perez-Valles, C.L., Regalado, S., Bara- Wen, L. (2010) MRI findings of primary CNS
Cellulaire. Journal of Clinical Oncology, 26, jas, R.F., Rubenstein, J.L. & Cha, S. (2013) lymphoma in 26 immunocompetent patients.
2512–2518. Combined diffusion and perfusion MR imaging Korean Journal of Radiology, 11, 269–277.
Soussain, C., Choquet, S., Fourme, E., Delgadillo, as biomarkers of prognosis in immunocompe- Zhu, J.J., Gerstner, E.R., Engler, D.A., Mrugala,
D., Bouabdallah, K., Ghesquieres, H., Damaj, G., tent patients with primary central nervous sys- M.M., Nugent, W., Nierenberg, K., Hochberg,
Dupriez, B., Vargaftig, J., Gonzalez, A., Houillier, tem lymphoma. American Journal of F.H., Betensky, R.A. & Batchelor, T.T. (2009)
C., Taillandier, L., Hoang-Xuan, K. & Leblond, V. Neuroradiology, 34, 35–40. High-dose methotrexate for elderly patients with
(2012) Intensive chemotherapy with thiotepa, Wechsler, D. (1997) Wechsler Adult Intelligence primary CNS lymphoma. Neuro Oncology, 11,
busulfan and cyclophosphamide and hematopoi- Scale, 3rd edn. The Psychological Corporation, 211–215.
etic stem cell rescue in relapsed or refractory pri- San Antonio.

ª 2018 British Society for Haematology and John Wiley & Sons Ltd 363
British Journal of Haematology, 2019, 184, 348–363

Das könnte Ihnen auch gefallen