ArticlePDF Available

R-MPV followed by high-dose chemotherapy with TBC and autologous stem-cell transplant for newly diagnosed primary CNS lymphoma

Authors:

Abstract and Figures

High-dose methotrexate-based chemotherapy is the mainstay of treatment for primary central nervous system lymphoma (PCNSL), but relapses remain frequent. High-dose chemotherapy (HDC) with autologous stem-cell transplant (ASCT) may provide an alternative to address chemoresistance and overcome the blood-brain barrier. In this single-center phase II study, newly-diagnosed PCNSL patients received 5-7 cycles of chemotherapy with rituximab, methotrexate (3.5g/m(2)), procarbazine and vincristine (R-MPV). Those with a complete or partial response proceeded with consolidation HDC with thiotepa, cyclophosphamide and busulfan (TBC), followed by ASCT and no radiotherapy. Primary endpoint was 1-year progression-free survival (PFS), N=32. The median age was 57; median KPS was 80. Following R-MPV, objective response rate was 97% and 26 (81%) patients proceeded with HDC-ASCT. Among all patients, median PFS and overall survival (OS) were not reached (median follow-up: 45 months). The 2-year PFS was 79% (95%CI 58-90), with no events observed beyond two years. The 2y-OS was 81% (95%CI 63-91). In transplanted patients, the 2-year PFS and OS were 81%. There were three treatment-related deaths. Prospective neuropsychological evaluations suggested relatively stable cognitive functions post-transplant. In conclusion, this treatment was associated with excellent disease control and survival, with an acceptable toxicity profile and no evidence of neurotoxicity thus far. This trial is registered to www.clinicaltrials.gov as NCT00596154. Copyright © 2015 American Society of Hematology.
Content may be subject to copyright.
Regular Article
CLINICAL TRIALS AND OBSERVATIONS
R-MPV followed by high-dose chemotherapy with TBC and autologous
stem-cell transplant for newly diagnosed primary CNS lymphoma
Antonio Omuro,
1
Denise D. Correa,
1
Lisa M. DeAngelis,
1
Craig H. Moskowitz,
2
Matthew J. Matasar,
2
Thomas J. Kaley,
1
Igor T. Gavrilovic,
1
Craig Nolan,
1
Elena Pentsova,
1
Christian C. Grommes,
1
Katherine S. Panageas,
3
Raymond E. Baser,
3
Geraldine Faivre,
1
Lauren E. Abrey,
1
and Craig S. Sauter
2
1
Department of Neurology,
2
Department of Medicine, and
3
Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center,
New York, NY
Key Points
We conducted a phase-2
study in newly diagnosed
PCNSL utilizing R-MPV and
HDC with ASCT.
Excellent disease control and
OS (2-year PFS: 79%) were
observed, with an acceptable
toxicity profile and minimal
neurotoxicity.
High-dose methotrexate-based chemotherapy is the mainstay of treatment of primary
central nervous system lymphoma (PCNSL), but relapses remain frequent. High-dose
chemotherapy (HDC) with autologous stem-cell transplant (ASCT) may provide an
alternative to address chemoresistance and overcome the blood-brainbarrier. In this
single-center phase-2 study, newly diagnosed PCNSL patients received 5 to 7 cycles
of chemotherapy with rituximab, methotrexate (3.5 g/m
2
), procarbazine, and vincristine
(R-MPV). Those with a complete or partial response proceeded with consolidation HDC
with thiotepa, cyclophosphamide, and busulfan, followed by ASCT and no radiotherapy.
Primary end point was 1-year progression-free survival (PFS), N 532. Median age was 57,
and median Karnofsky performance status 80. Following R-MPV, objective response rate
was 97%, and 26 (81%) patients proceeded with HDC-ASCT. Among all patients, median
PFS and overall survival (OS) were not reached (median follow-up: 45 months). Two-year
PFS was 79% (95% confidence interval [CI], 58-90), with no events observed beyond
2 years. Two-year OS was 81% (95% CI, 63-91). In transplanted patients, 2-year PFS and OS were 81%. There were 3 treatment-related
deaths. Prospective neuropsychological evaluations suggested relatively stable cognitive functions posttransplant. In conclusion,
this treatment was associated with excellent disease control and survival, an acceptable toxicity profile, and no evidence of
neurotoxicity thus far. This trial was registered at www.clinicaltrials.gov as NCT00596154. (Blood. 2015;125(9):1403-1410)
Introduction
More than 90% of patients with primary central nervous system
lymphoma (PCNSL) display a diffuse large B-cell lymphoma (DLBCL)
phenotypic subtype, but standard DLBCL regimens such as cyclo-
phosphamide, doxorubicin, vincristine and prednisone and variations
are ineffective in this disease.
1,2
This has been explained by poor
penetration of these agents across the blood-brain barrier (BBB), a
problem that has been partially addressed with the development of
high-dose methotrexate-based regimens (HD-MTX) that result in
therapeutic central nervous system (CNS) and cerebrospinal uid
(CSF) levels after rapid infusions of 1.5 to 8 g/m
2
.
3-5
Such high
methotrexate doses are made possible with the concomitant use of
leucovorin, which prevents bone marrow and systemic organ damage,
while limiting rescue of lymphoma cells in the CNS because it has poor
BBB penetration. This clever strategy, used with or without whole-
brain radiotherapy (WBRT), has resulted in remarkable survival im-
provements, with recent studies reporting median overall survival (OS)
of 31 to 79 months,
6-13
as compared with 12 months observed with
WBRT alone.
14
In spite of these improvements, early and late relapses
remain frequent, and the majority of patients still die of disease.
15
High-dose chemotherapy (HDC) followed by autologous stem-
cell transplant (ASCT) has been proposed as an alternative con-
solidation therapy in PCNSL.
16
In addition to overcoming intrinsic
chemoresistance of lymphoma cells, HDC-ASCT may improve
disease control by affording higher CNS drug concentrations,
circumventing chemoresistance mediated by the BBB, a similar
high-dose/ rescueparadigm proved successful in the development
of HD-MTX. We previously conducted a phase-2 study in newly
diagnosed PCNSL testing an induction chemotherapy with HD-
MTX and cytarabine, followed by consolidation HDC-ASCT
utilizing carmustine (BCNU), etoposide, cytarabine, and melphalan
(BEAM).
17
Unfortunately, that treatment resulted in suboptimal
disease control, with an intent-to-treat (ITT) 3-year event-free sur-
vival (EFS) of only 25%, and a 3-yearOS of 60%. In the present study,
we sought to optimize this strategy by utilizing an enhanced induc-
tion regimen consisting of rituximab, methotrexate, procarba-
zine, and vincristine (R-MPV),
6
as well as a more aggressive, CNS
penetrant HDC regimen consisting of thiotepa, busulfan, and cyclo-
phosphamide (TBC).
16
Submitted October 9, 2014; accepted December 22, 2014. Prepublished
online as Blood First Edition paper, January 7, 2015; DOI 10.1182/blood-2014-
10-604561.
The online version of this article contains a data supplement.
There is an Inside Blood Commentary on this article in this issue.
The publication costs of this article were defrayed in part by page charge
payment. Therefore, and solely to indicate this fact, this article is hereby
marked “advertisement” in accordance with 18 USC section 1734.
©2015 by The American Society of Hematology
BLOOD, 26 FEBRUARY 2015 xVOLUME 125, NUMBER 9 1403
For personal use only.on November 4, 2015. by guest www.bloodjournal.orgFrom
Patients and methods
Patients
Immunocompetent adult patients with histologically conrmed newly diagnosed
PCNSL with brain involvement were enrolled in this prospective, single-arm,
phase-2 study between June 2005 and September 2011. The supplemental
Appendix (available on the Blood Web site) details inclusion/exclusion criteria
and baseline evaluations. Patients were eligible if they had non-Hodgkin lym-
phoma involving the brain, as demonstrated by magnetic resonance imaging
(MRI) and histologic conrmation by (1) positive CSF cytology for lym-
phoma ora monoclonal lymphocyte populationdened by cell surfacemarkers;
(2) vitreous or uvea biopsydemonstratingnon-Hodgkinlymphoma; or (3) brain
biopsy. Other inclusion criteria consisted of age 18 to 72 years; negative HIV
testing; left ventricular ejection fraction $50%; absence of systemic lym-
phoma on the chest, abdomen, and pelvis CT and bone marrow biopsy;
leukocytes .3000/mm
3
; platelets .100 000/mm
3
; bilirubin ,2mg/dL;
serum creatinine,1.5 mg/dL; or creatinine clearance .50cc/min per 1.73 m
2
.
Exclusion criteria consisted of prior cranial irradiation or chemotherapy for
PCNSL, other active malignancy (exception: skin basal cell carcinoma and
cervical carcinoma in situ), or immunodeciency. Patients were eligible
regardless of performance status. Pretreatment evaluations also included
CSF sampling, slit lamp exam, electrolytes, liver enzymes, Epstein-Barr virus,
cytomegalovirus, herpes simplex virus and hepatitis B/C serologies, urinalysis,
24-hour creatinine clearance, and chest radiograph.
The trial and informed consent were approved by the Institutional Review
Board (www.clinicaltrials.gov: NCT00596154). Written informed consent
was obtained from all patients or guardians in accordance with the Dec-
laration of Helsinki. Data were analyzed by the authors at Memorial Sloan
Kettering Cancer Center, and all authors had access to primary clinical trial
data.
Induction chemotherapy
Treatment started with an induction R-MPV chemotherapy
6
(1 cycle 514 days),
as follows: day 1, rituximab 500 mg/m
2
IV; day 2, methotrexate 3.5 mg/m
2
IV
(over 2 hours) and vincristine 1.4 mg/m
2
(capped at 2.8 mg). Procarbazine
100 mg/m
2
per day was given on days 2 to 8 during odd cycles. Standard
hydration and leucovorin rescue were given per institutional guidelines.
To prevent febrile neutropenia and toxic deaths previously observed with
R-MPV, prophylactic lgrastim was given to all patients.
An MRI of the brain was performed after 5 cycles. Patients with pro-
gressive disease (PD) were taken off-study. Patients with complete response
(CR) proceeded directly to HDC-ASCT. Patients in partial response (PR) or
stable disease (SD) received 2 additional cycles and proceeded with HDC-
ASCT if PR/CR was observed on a repeat MRI or were taken off-study if SD
or PD. After transplant, patients were followed radiographically, with no
WBRT or further treatments offered until progression. Responses were
assessed utilizing previously described criteria
18
; in addition to CR, PR, SD
and PD, those criteria also characterize unconrmed CR (CRu), dened
by absence of contrast-enhancing disease in the setting of corticosteroids use,
or minimal enhancing abnormalities of uncertain signicance, typically cor-
responding to postoperative changes following biopsy.
Harvesting and HDC-ASCT
Peripheral blood stem cell harvesting was performed after the rst or second
R-MPV cycle, or at the discretion of treating physician, following insti-
tutional standard procedures. Cytapheresis was performed after bone marrow
recovery and repeated daily up to day 7 until .5310
6
CD34
1
cells were
collected (minimum acceptable total yield: 2 310
6
CD34
1
cells per kg).
Patients in CR/PR as described previously underwent pretransplant
evaluation (supplemental Appendix); those deemed clinically eligible pro-
ceeded with HDC-ASCT with thiotepa 250 mg/m
2
IV on days 29, 28, and 27;
busulfan 3.2 mg/kg IV on days 26, 25, and 24; and cyclophosphamide
60 mg/kg IV on days 23 and 22. Stem cell reinfusion occurred on day 0.
Supportive therapy followed institutional guidelines, including granulocyte
CSF, seizure and infection prophylaxis, hydration, antiemesis, vitamin K, and
irradiated blood products.
Response was assessed after R-MPV and prior to transplant, and every
3 months thereafter (rst 2 years), then every 4 months (third and fourth years)
and every 6 months thereafter. Repeat CSF and/or ocular exam were done at
those time points if CSF or ocular involvement was present at diagnosis.
Exploratory neuropsychological testing, quality of life, and
radiographic evaluation of neurotoxicity
In addition to investigatorsclinical assessment of neurotoxicity,
19
potential
neurotoxic effects of disease, R-MPV, and HDC-ASCT were further char-
acterized through prospective neuropsychological and quality of life (QoL)
evaluations, as well as radiographic assessment of white matter abnormal-
ities. Evaluations were conducted in progression-free patients at the following
time points: pretreatment (baseline), after induction chemotherapy (before
transplant), and at 6-month intervals following transplant. The present analy-
sis reports on evaluations up to 24 months after transplant.
Neuropsychological evaluations were performed in 60-minute sessions.
Raw test scores were compared with published normative values according to
age and education, and converted into zscores to characterize presence and
severity of cognitive difculties. A zscore #21.5 represents impairment.
The following tests were performed: Trail Making Test (TMT) Part A and
TMT Part B; Brief Test of Attention; Controlled Oral Word Association Test;
Hopkins Verbal Learning Test-Revised (HVLT-R)Total Learning, HVLT-
RDelayed Recall, and HVLT-RDiscrimination Index; and Grooved
Pegboard Test (GPT)Dominant Hand and GPTNon-Dominant Hand.
Self-reported QoL and mood were evaluated utilizing the Functional
Assessment of Cancer TherapyBrain Cancer (FACT-BR) and the Beck
Depression Inventory (BDI), respectively.
Fluid-attenuated inversion recovery (FLAIR) MRI sequences obtained at
the same time points were scored for white matter abnormalities utilizing the
modied Fazekas scale.
20
Statistics
The primary end point was 1-year progression-free survival (PFS) among
all patients, dened as time from registration to tumor relapse, progression, or
death of any cause, whichever comes rst. For the purposes of PFS estimates,
patients who withdrew consent or were removed from study because of reasons
other than progression, toxicity, or death were censored at the date of last ra-
diographic assessment conrming a progression-free status. With a sample size
of 33 patients, an exact binomial test with a nominal 0.05 1-sided signicance
level has 90% power to detect the difference between the null hypothesis
(0.5 observed in HD-MTX without radiotherapy
13
) and the alternative propor-
tion of 0.75. The study was to be stopped if more than 2 toxic deaths occurred
among the rst15patients,or4toxicdeathsatanytime.Ifthetrueriskoftoxic
death is $16%, then the probability of seeing 4 toxic deaths (and stopping the
trial) was at least 80%. Secondary end points consisted of OS, acute and chronic
toxicities (CTCAEv3), and response rate after R-MPV and after HDC-ASCT.
Explorator y end points consiste d of evaluation of neuro psychological, QoL ,
and white matt er abnormalities, as described previously.
Kaplan-Meier methodology was used for analysis of OS and PFS. Neuropsy-
chological test scores were summarized using descriptive statistics, and longitu-
dinal trajectories evaluated using linear mixed models (LMMs) controlling for age
and estimated interquartile range (IQR). Exact follow-up assessment times from
baseline were calculated, and both linear and quadratic terms were estimated by
the LMMs. McNemarsx
2
test was used to test for signicant changes in MRI
white matter abnormality scores between assessments.
Results
Patient characteristics
A total of 33 patients were enrolled and 32 analyzed; 1 registered
patient was excluded because of presence of systemic lymphoma at
1404 OMURO et al BLOOD, 26 FEBRUARY 2015 xVOLUME 125, NUMBER 9
For personal use only.on November 4, 2015. by guest www.bloodjournal.orgFrom
diagnosis, discovered upon post hoc radiology review, deeming the
patient ineligible. The median age was 57 (range: 23-67) and median
Karnofsky performance status (KPS) was 80 (40-100). Table 1 shows
detailed patient characteristics.
Induction treatment
Induction R-MPV was well tolerated, with no treatment-related
deaths and no treatment discontinuation because of toxicity; grades
3 and 4 toxicities are summarized in Table 2. Following 5 R-MPV
cycles, 14 patients were in CR/CRu, 16 in PR, and 1 progressed
(Table 3 and patient owchart [supplemental Appendix]). One ad-
ditional patient had undergone complete resection prior to enroll-
ment and had no measurable disease at baseline; that patient was
stable after 5 cycles of R-MPV. All patients in less than CR, in ad-
dition to 2 patients in CR/CRu with incomplete resolution of symp-
toms after 5 cycles, received 2 additional R-MPV cycles (N 519).
The objective response rate after R-MPV, dened as CR, CRu, or PR
after 5 or 7 cycles in eligible patients with measurable disease (N 531
evaluable) was 97% (95% condence interval [CI], 83-100).
HDC-ASCT
Thirty-one (97%) patients were deemed eligible for transplant based on
response, which includes the completely resected patient who remained
stable following 7 R-MPV cycles. A total of N 526 (81%) patients
eventually received HDC-ASCT. The other 5 patients did not undergo
transplant because of refusal/consent withdrawal (N 52) or physicians
decision (N 53) and were removed from study. There were no har-
vesting failures.
Table 4 summarizes toxicities observed with TBC and median times
to hematologic recovery. There was no venoocclusive disease. Two
patients died of transplant-related acute complications: a 57-year-old
patient developed a Stevens-Johnson syndrome, and a 59-year-old
patient developed septic shock. Another patient aged 61 developed
a fatal chronic colitis of undetermined etiology, potentially representing
graft-versus-host disease. In that patient, symptoms started 3 weeks
following transplant and responded poorly to corticosteroids. To date,
no patient has developed secondary malignancies.
PFS and OS
Among all patients (N 532), the median PFS has not been reached
(Figure 1A), and the 1-year PFS estimate is 82% (95% CI, 62-92).
There have been no events after 2 years, and the 2-year, 3-year, and
5-year PFS estimates are 79% (95% CI, 58-90). Aside from the toxic
deaths, there was no treatment discontinuation because of toxicities,
and EFS and PFS are identical. The events were toxic deaths (N 53),
progression during R-MPV (N 51), and progression after transplant
(N 52).
The median OS (Figure 1B) has not been reached, and the 1-year
OS is 88% (95% CI, 70-95). No deaths were observed beyond
2 years, with a 2-year, 3-year, and 5-year OS of 81% (95% CI,
63-91). The median follow-up of survivors is 45 months (range:
27-86).
Among the 26 transplanted patients, the 1-year PFS is 85%
(95% CI, 64-94), and the 2-year, 3-year, and 5-year PFS est imates are
81% (95% CI, 60-92); the median PFS was not reached (Figure 1C).
The 1-year OS is 88% (95% CI, 68-96), and the 2-year, 3-year, and
5-year OS estimates are 81% (60% to 92%); the median OS was
not reached (Figure 1C).
The effects of age were analyzed according the MSK RPA class
cutoff of 50.
21
Patients age #50 tended to achieve superior PFS
(P5.05, Figure 1D) and OS (P5.06), with no observed progression
or death of any cause.
The outcomes of patients removed from study in spite of being
eligible for transplant (N 55) were as follows: 2 patients chose to
receive high-dose cytarabine and no further treatment; 1 of those
relapsed and received salvage chemotherapy and HDC-ASCT with
TBC. The remainder 3 patients chose to receive WBRT (2 patients:
23.4 Gy; 1 patient: 45 Gy) and have never relapsed. All 5 patients
remain alive, but as described previously, they were censored at the
time of last on-study radiographic assessment for the purposes of
PFS calculation, and therefore the reported PFS end points do not
reect these additional treatments.
Neuropsychological testing, QoL, and radiographic evaluation
of neurotoxicity
No clinical neurotoxicity, dened as neurologic deterioration in the
absence of disease progression, was reported by treating physicians.
In addition, 16 progression-free patients participated in the neuro-
psychological and QoL evaluations (Figure 2 and supplemental
Appendix).
Table 1. Patient characteristics (N 532)
Characteristic Value
Median age (range) 57 (23-67)
Age ,60 21 (66%)
Age ,50 11 (34%)
Median KPS (range) 80 (40-100)
KPS ,70 6 (19%)
KPS ,50 1 (3%)
Women 15 (47%)
Men 17 (53%)
MSK RPA
Class I 11 (34%)
Class II 15 (47%)
Class III 6 (19%)
DLBCL 32 (100%)
CSF cytology*
Positive 1 (3%)
Suspicious 2 (6%)
Not performed 1 (3%)
Ocular involvement 3 (9%)
Median product of tumor diameters (range) 6 cm
2
(0-20 cm
2
)
MSK RPA, Memorial Sloan-Kettering prognostic score determined by recursive
partitioning analysis (I, age ,50; II, age $50 and KPS $70; III, age $50 and
KPS ,70).
*Conventional cytology; flow cytometry not performed.
Table 2. Grades 3 and 4 toxicities reported during R-MPV (N 532)
Toxicity Grade 3 Grade 4
Hemoglobin 6 18%
Neutrophils 6 18% 1 3%
ALT and/or AST 14 42% 3 9%
Creatinine 5 15%
Infection 4 12%
Peripheral neuropathy 2 6%
Fatigue 2 6%
Thrombosis/embolism 1 3% 2 6%
Constipation 1 3%
Syncope 1 3%
Encephalopathy 1 3%
ALT, alanine aminotransferase; AST, aspartate aminotransferase.
BLOOD, 26 FEBRUARY 2015 xVOLUME 125, NUMBER 9 R-MPV AND TRANSPLANT FOR PRIMARY CNS LYMPHOMA 1405
For personal use only.on November 4, 2015. by guest www.bloodjournal.orgFrom
As shown in Figure 2A, at baseline, there was evidence of
impairment in several cognitive domains, characterizing baseline
disease burden. Results of LMM analysis showed signicant posi-
tive linear time components (months since baseline) for the HVLT-
RDelayed Recall and HVLT-RDiscrimination Index (P,.05),
indicating continuous improvement in scores from baseline over
time. Likewise, all of the other tests had signicant positive linear
components up to 12 to 18 months posttransplant. However, a
signicant (TMT Part A, TMT Part B, Brief Test of Attention,
HVLT-RTotal Learning; P,.05) or marginally signicant (Con-
trolled Word Association Test, GPTDominant Hand, GPTNon-
Dominant Hand; P,.10) quadratic time component was observed,
suggesting that the rate of cognitive improvement slowed by 12 to
18 months posttransplant.
Self-reported QoL largely mirrored the improvement of cognitive
function in the LMM analysis (Figure 2B). FACT-BR scores sig-
nicantly improved from baseline, with slowed improvement by 12
to 18 months posttransplant; BDI scores signicantly and linearly
decreased over time.
Analysis of white matter abnormalities (Figure 2C) showed an im-
provement after R-MPV, with 81% of patients displaying scores
2 to 3 at baseline, compared with 19% after R-MPV (McNemarsx
2
P5.002). Following transplant, there was an increase in white matter
abnormalities, with 44% of patients with scores 2 to 3 (McNemarsx
2
P5.046), which then remained stable over time. No scores above
3wereseenatanytime.
Discussion
In this phase-2 study, patients with newly diagnosed PCNSL were
treated with R-MPV chemotherapy, followed by consolidation
HDC-ASCT with TBC in responding patients and no further treat-
ment until progression. Favorable disease control was observed
(2-year PFS, 75%; 2-year OS, 81%), far exceeding results of our
previous experience with HDC-ASCT in PCNSL utilizing HD-
MTX/cytarabine and BEAM.
17
Importantly, no clinically signicant
neurotoxicity has developed to date, and QoL continuously improved
over time.
A number of factors contributed to the favorable results observed
in this trial, in comparison with our previous study (Table 5).
17
The
induction chemotherapy performed better, with a higher response
rate (97%) that increased the number of patients undergoing HDC-
ASCT (81%) and minimized disease burden prior to transplant (69%
transplanted in CR). This excellent performance conrms our pre-
vious experience with R-MPV,
6
which seems superior to results with
MPV, although it remains difcult to determine the contribution of
rituximab, currently being investigated in an ongoing randomized
trial. Moreover, the TBC regimen, although more toxic, resulted in
improved, durable disease control, likely reecting higher CNS drug
levels in comparison with BEAM.
The TBC regimen was chosen based on favorable results ob-
served in recurrent PCNSL.
16,22-25
In a multicenter phase-2 study
(N 543),
16
TBC was used following a cytarabine/etoposide (CYVE)
salvage chemotherapy, achieving PFS of 12 months and OS of
18 months. Among transplanted patients (N 527), PFS was 41 months.
Three patients died of CYVE, but there were no transplant-related
deaths, which is in line with other studies on TBC in CNS lym-
phomas
24,25
and other malignancies,
26-28
all reporting transplant-
related mortality under 5%. In our study, treatment-related mortality
appeared higher, but it is difcult to determine if this could be
because of a higher susceptibility specic to PCNSL patients, or
if it could reect a less selected patient population, as compared
with recurrent disease patients receiving TBC after surviving highly
toxic salvage chemotherapies, such as CYVE. Also of interest is
the fact that some patients in Soussains study
16
were transplanted
in spite of no response to CYVE and still survived longer (OS:
9 months) than CYVE-refractory patients who were not transplanted
(OS: 5 months). This raises the intriguing question of whether
the chemoresistance observed in recurrent CNS lymphoma after
induction chemotherapies can be at least in part a consequence of the
BBB reducing drug access, which could be overcome by the high
chemotherapy doses afforded by transplant-based strategies. This
question was not addressed in our study, given that only 1 patient
progressed on R-MPV, and as per protocol design, she did not
receive HDC-ASCT.
In addition to our previous study with BEAM, a number of trials
have examined the use of HDC-ASCT in newly diagnosed PCNSL
(Table 5). However, interpreting results is difcult because, unlike
our trials, a signicant proportion of patients in those studies also
received WBRT, given either as adjuvant treatment post-ASCT to
all patients or to patients who did not achieve a CR to induction.
In a study of 30 patients, methotrexate, cytarabine, and thiotepa
induction was followed by HDC-ASCT with BCNU/thiotepa, and
hyperfractionated WBRT (45 Gy for CR and 50 Gy for PR).
29,30
The 3-year OS was 69% in all patients, and 87% in transplanted
patients. There was no neuropsychological evaluation, but 5 patients
developed clinically dened neurotoxicity. That same group
subsequently reported a small series (N 513) using a similar
Table 3. Response status after R-MPV and following transplant
CR/CRu PR SD PD
Response after 5 R-MPV cycles (N 532) 14 (44%) 16 (50%) 1* (3%) 1 (3%)
Best response to R-MPV induction
chemotherapy (5 or 7 cycles) (N 532)
21 (66%) 9 (28%) 1* (3%) 1 (3%)
Pretransplant response status in the
transplanted patients (N 526)
18 (69%) 7 (27%) 1* (4%) 0 (0)
Best response after transplant (N 526) 21 (81%) 3 (11%) 1* (4%) 1 (4%)
*This 1 patient had no measurable disease at start of R-MPV because of
complete resection, remained stable after 7 cycles of R-MPV and underwent
HDC-ASCT, and was considered nonevaluable for objective response rate
assessment.
Table 4. Number of patients experiencing grades 3 to 5 toxicities
from transplant, and time to hematologic recovery (N 526)
Nonhematologic Grade 3 Grade 4 Grade 5
Febrile neutropenia 11 42%
Infection 4 15% 2 8% 1 4%
Skin rash 1 4% 1 4% 1 4%
Chronic colitis 1 4%
Encephalopathy 1 4%
Dehydration 1 4%
Cardiac failure 1 4%
Weight loss 1 4%
Nausea 1 4%
Diarrhea 1 4%
Mucositis 1 4%
Engraftment
Median time to neutrophil recovery (.500/mm
3
): 9 d (range: 7-15)
Median time to platelet recovery (.25 000/mm
3
): 15 d (range: 10-124)
Median duration of hospital admission for HDC-ASCT: 26 d (range 20-90)
1406 OMURO et al BLOOD, 26 FEBRUARY 2015 xVOLUME 125, NUMBER 9
For personal use only.on November 4, 2015. by guest www.bloodjournal.orgFrom
strategy, but without WBRT if a CR was achieved, and the 3-year OS
was 77%
31
; that study has been expanded,
32
and nal results are
awaited. In another phase-2 study (N 525), induction with MTX 3 g/m
2
,
BCNU, etoposide, and methylprednisolone followed by HDC-
ASCT (BEAM) and WBRT (30 Gy) achieved 3-year EFS of
58% and 3-year OS of 64%. There was no neuropsychological
follow-up, but at least 1 patient developed neurotoxicity.
33
A study in
28 patients used 2 doses of single-agent HD-MTX, followed by
HDC-ASCT (busulfan/thiotepa), and WBRT 45 Gy if less than CR.
Three of the 9 irradiated patients died of neurotoxicity, for a 2-year
OS of 48%.
34
Another study focusing on patients with primary and
secondary CNS lymphoma of various histologies and previous treat-
ments selected for transplant used rituximab combined with TBC as
myeloablative regimen and found a favorable toxicity prole, with
a 2-year PFS of 81%.
35
Additional retrospective studies have been
reported, adding to anecdotal experience with HDC-ASCT in CNS
lymphoma.
36-39
The lack of clinically detectable neurotoxicity is a favorable
aspect of our regimen, but comprehensive neuropsychological eval-
uations are essential to fully characterize neurotoxic effects that
may impair QoL in long-term survivors. In line with previous reports
of R-MPV in PCNSL,
6
results showed marked improvements in
cognitive function and QoL following induction MTX chemother-
apy, reecting a decrease in disease burden. Following transplant,
neuropsychological test scores remained overall stable, with self-
reported QoL continuously improving. However, we found some
indications of acute neurotoxicity following TBC, as exemplied by
the increase in white matter abnormalities, and a mild, transient
decrease in some neurocognitive scores at the rst posttransplant
evaluation (Figure 2A and supplemental Appendix). Moreover, the
rate of cognitive improvement slowed after 12 to 18 months, and
long-term follow-up is warranted to determine whether this could
represent a trend toward development of late-delayed neurotoxicity.
Our study has some limitations. Inclusion criteria allowed for
enrollment of patients regardless of performance status, and up to
72 years old, but our oldest patient was 67, and median age was 57,
which is younger than the typical PCNSL population enrolled in
non-ASCT trials; only 19% of patients had an MSK RPA class III.
Therefore, results must be compared against other PCNSL transplant
trials (Table 5), rather than nontransplant treatment strategies that are
inclusive of a wider population of patients. Moreover, the R-MPV
regimen has not been formally tested without a consolidation strat-
egy such as WBRT or HDC-ASCT. Therefore, it is difcult to assess
whether such patients with a more favorable prognosis
40
could have
achieved similar results with R-MPV alone and without any con-
solidation treatment.It is noteworthy that salvage treatments including
TBC-based HDC-ASCT
16,24
or even WBRT
41,42
are also effective in
this population and will require further investigation. Ongoing study
Radiation Therapy Oncology Group 1114 is investigating R-MPV
with and without reduced-dose WBRT and willprovide further dataon
the relevance of consolidation treatments innewly diagnosed PCNSL.
Finally, this was a single-institution trial, and even though we are
Figure 1. PFS and O S. (A) PFS, all pa tients (N 532). (B) OS, all patients (N 532). (C ) PFS and OS in transp lanted patients. (D) PFS accord ing to age (above 50 v s 50
and under). P5.05.
BLOOD, 26 FEBRUARY 2015 xVOLUME 125, NUMBER 9 R-MPV AND TRANSPLANT FOR PRIMARY CNS LYMPHOMA 1407
For personal use only.on November 4, 2015. by guest www.bloodjournal.orgFrom
reporting on the largest number of transplanted patients published to
date (Table 5), the sample size remains small, and results need veri-
cation in a multicenter, randomized environment.
In summary, we report a prospective trial of HDC-ASCT in
newly diagnosed PCNSL that was associated with excellent disease
control, reecting an optimized induction chemotherapy and a more
Figure 2. Neuropsychol ogical testi ng, mood/QoL scor es, and evaluat ion of white matte r changes on MRI ove r time. Time points: 1, baseline; 2, after R-MPV and
before transplant; 3, 4, 5, and 6, every 6 months after transplant. (A) Neuropsychological testing (zscores) boxplot. BL, baseline; BTA, Brief Test of Attention; COWA,
Controlled Word Association Test; GPT-D, Grooved Pegboard Test–Dominant Hand; GPT-ND, Grooved Pegboard Test–Non-Dominant Hand; HVLT-R-DEL, Hopkins Verbal
Learning Test-Revised–Delayed Recall; HVLT-R-DI, Hopkins Verbal Learning Test-Revised–Discrimination Index; HVLT-R-TL, Hopkins Verbal Learning Test-Revised–Total
Learning; R-MPV, after induction chemotherapy; TMTA, Trail Making Test Part A; TMTB, Trail Making Test Part B. The zscores #1.5 represent impairment. The lower and
upper boundaries of each box represent the first and third quartiles (ie, the 25th and 75th percentiles), respectively, at the indicated time point, and the IQR is the distance
between the lower and upper boundaries. The error bars (“whiskers”) extend from the first and third quartiles to the lowest and highest scores that are within 1.5 3IQR of their
respective quartile. Any scores beyond the error bars are considered outliers and are represented as individual points. The asterisks represent the median scores. The lines
connecting the medians over time for each test have no statistical interpretation and are intended to serve as visual aids. (B) BDI and FACT-BR (raw scores) boxplot. See
panel A for boxplot explanation. (C) White matter abnormality scores (modified Fazekas scale) over time (N 516). Fazekas scores: 0, no white matter abnormality; 1, minimal
patchy white matter foci; 2, start of confluence of white matter disease; 3, large confluent areas; 4, confluence of white matter abnormalities with cortical and subcortical
involvement; 5, diffuse leukoencephalopathy. No patient developed white matter abnormality scores of 4 or 5.
1408 OMURO et al BLOOD, 26 FEBRUARY 2015 xVOLUME 125, NUMBER 9
For personal use only.on November 4, 2015. by guest www.bloodjournal.orgFrom
effective myeloablative regimen. However, acute toxicities, in-
cluding 3 toxic deaths, were observed, and longer neurocognitive
follow-up is warranted for evaluation of late-delayed neurotoxic-
ity. This treatment should still be considered experimental, and
enrollment in randomized trials
43
encouraged to establish the ulti-
mate role and optimal timing of HDC-ASCT in the care of PCNSL
patients.
Acknowledgments
The authors thank participants of the American Association for Cancer
Research (AACR)/American Society of Clinical Oncology (ASCO)
Vail Workshop on Methods in Clinical Cancer Research where the
protocol was developed. Presented at the ASCO Annual Meeting 2012
(platform presentation).
This work was an investigator-initiated clinical trial sponsored by
Memorial Sloan Kettering Cancer Center Department of Neurology
Research and Development Funds and was conducted with no
industry support. K.S.P. received research funding from National
Institutes of Health, National Cancer Institute (grant P30 CA008748).
Authorship
Contribution: A.O., C.H.M., D.D.C., K.S.P., and L.E.A. con-
ceived of and designed the trial; A.O. and G.F. performed
collection and assembling of data; all authors performed analysis
and interpretation of data; A.O. and G.F. wrote the manuscript;
and all authors performed nal approval of the manuscript.
Conict-of-interest disclosure: L.E.A. is currently an employee at
Roche. The remaining authors declare no competing nancial interests.
The current afliation for L.E.A. is Roche, Basel, Switzerland.
Correspondence: Antonio Omuro, Department of Neurology,
Memorial Sloan Kettering Cancer Center, 1275York Ave, New York,
NY 10065; e-mail: omuroa@mskcc.org.
References
1. Schultz C, Scott C, Sherman W, et al.
Preirradiation chemotherapy with
cyclophosphamide, doxorubicin, vincristine, and
dexamethasone for primary CNS lymphomas:
initial report of radiation therapy oncology group
protocol 88-06. J Clin Oncol. 1996;14(2):556-564.
2. O’Neill BP, O’Fallon JR, Earle JD, Colgan JP,
Brown LD, Krigel RL. Primary central nervous
system non-Hodgkin’s lymphoma: survival
advantages with combined initial therapy? Int J
Radiat Oncol Biol Phys. 1995;33(3):663-673.
3. Gabbai AA, Hochberg FH, Linggood RM, Bashir
R, Hotleman K. High-dose methotrexate for non-
AIDS primary central nervous system lymphoma.
Report of 13 cases. J Neurosurg. 1989;70(2):
190-194.
4. DeAngelis LM, Yahalom J, Thaler HT, Kher U.
Combined modality therapy for primary CNS
lymphoma. J Clin Oncol. 1992;10(4):635-643.
5. Glass J, Gruber ML, Cher L, Hochberg FH.
Preirradiation methotrexate chemotherapy of
primary central nervous system lymphoma: long-
term outcome. J Neurosurg. 1994;81(2):188-195.
6. Morris PG, Correa DD, Yahalom J, et al.
Rituximab, methotrexate, procarbazine, and
vincristine followed by consolidation reduced-dose
whole-brain radiotherapy and cytarabine in newly
diagnosed primary CNS lymphoma: final results
and long-term outcome. J Clin Oncol. 2013;
31(31):3971-3979.
7. Rubenstein JL, Hsi ED, Johnson JL, et al.
Intensive chemotherapy and immunotherapy in
patients with newly diagnosed primary CNS
lymphoma: CALGB 50202 (Alliance 50202). J Clin
Oncol. 2013;31(25):3061-3068.
8. Ferreri AJ, Reni M, Foppoli M, et al; International
Extranodal Lymphoma Study Group (IELSG).
High-dose cytarabine plus high-dose
methotrexate versus high-dose methotrexate
alone in patients with primary CNS lymphoma:
a randomised phase 2 trial. Lancet. 2009;
374(9700):1512-1520.
9. Thiel E, Korfel A, Martus P, et al. High-dose
methotrexate with or without whole brain
radiotherapy for primary CNS lymphoma
(G-PCNSL-SG-1): a phase 3, randomised,
non-inferiority trial. Lancet Oncol. 2010;11(11):
1036-1047.
10. Poortmans PM, Kluin-Nelemans HC, Haaxma-
Reiche H, et al; European Organization for
Research and Treatment of Cancer Lymphoma
Group. High-dose methotrexate-based
chemotherapy followed by consolidating
radiotherapy in non-AIDS-related primary central
nervous system lymphoma: European
Organization for Research and Treatment of
Cancer Lymphoma Group Phase II Trial 20962.
J Clin Oncol. 2003;21(24):4483-4488.
11. Pels H, Schmidt-Wolf IGH, Glasmacher A, et al.
Primary central nervous system lymphoma:
results of a pilot and phase II study of systemic
and intraventricular chemotherapy with deferred
radiotherapy. J Clin Oncol. 2003;21(24):
4489-4495.
12. Abrey LE, Yahalom J, DeAngelis LM. Treatment
for primary CNS lymphoma: the next step. J Clin
Oncol. 2000;18(17):3144-3150.
13. Batchelor T, Carson K, O’Neill A, et al. Treatment
of primary CNS lymphoma with methotrexate and
deferred radiotherapy: a report of NABTT 96-07.
J Clin Oncol. 2003;21(6):1044-1049.
14. Nelson DF, Martz KL, Bonner H, et al. Non-
Hodgkin’s lymphoma of the brain: can high dose,
large volume radiation therapy improve survival?
Report on a prospective trial by the Radiation
Therapy Oncology Group (RTOG): RTOG 8315.
Int J Radiat Oncol Biol Phys. 1992;23(1):9-17.
15. Graber JJ, Omuro A. Primary central nervous
system lymphoma: is there still a role for
radiotherapy? Curr Opin Neurol. 2011;24(6):
633-640.
Table 5. Trials or case series (20 or more patients) reporting outcomes in newly diagnosed PCNSL patients treated with consolidation
HDC-ASCT after methotrexate-based initial treatment
Reference N
Induction
regimen
ORR to
induction
HDC-ASCT
regimen
Received
consolidation
HDC-ASCT
Received
consolidation
WBRT*
ORR to
HDC-
ASCT
Med F/u
(mo)
ITT 3-y
PFS
ITT 3-y
OS
Regimens including WBRT to all patients
29 30 MTX 8 g/m
2
, Ara-C, thiotepa 80% BCNU, thiotepa 1WBRT 23 (77%) 21 (70%) 100% 63 na 69%
33 25 MTX 3 g/m
2
, BCNU, VP16,
methylpred
84% BEAM 1WBRT 17 (68%) 24 (96%) 100% 34 58% 64%
Regimens with WBRT to patients achieving less than CR
34 28 MTX 8 g/m
2
61% Busulfan, thiotepa 1WBRT
if no CR
16 (70%) 8 (29%) 69% 15 45% 48% (2 y)
Regimens without WBRT
17 28 MTX 3.5 g/m
2
, Ara-C 57% BEAM 14 (50%) 0 77% 28 25% 60%
This study 32 MTX 3.5 g/m
2
, rituximab,
procarbazine, vincristine
97% Thiotepa, busulfan, CTX 26 (81%) 0 92% 45 79% 81%
*Number of patients who received WBRT as part of initial treatment, in the absence of tumor progression.
Ara-C, cytarabine; CTX, cyclophosphamide; Med F/u, median follow-up; methylpred, methylprednisolone; MTX, methotrexate; na, not available; ORR, objective response
rate; VP-16, etoposide.
BLOOD, 26 FEBRUARY 2015 xVOLUME 125, NUMBER 9 R-MPV AND TRANSPLANT FOR PRIMARY CNS LYMPHOMA 1409
For personal use only.on November 4, 2015. by guest www.bloodjournal.orgFrom
16. Soussain C, Hoang-Xuan K, Taillandier L, et al;
Soci´et ´e Franc¸ aise de Greffe de Mo¨elle Osseuse-
Th´erapie Cellulaire. Intensive chemotherapy
followed by hematopoietic stem-cell rescue for
refractory and recurrent primary CNS and
intraocular lymphoma: Soci ´et ´e Franc¸ aise de
Greffe de Mo¨elle Osseuse-Th ´erapie Cellulaire.
J Clin Oncol. 2008;26(15):2512-2518.
17. Abrey LE, Moskowitz CH, Mason WP, et al.
Intensive methotrexate and cytarabine followed
by high-dose chemotherapy with autologous
stem-cell rescue in patients with newly diagnosed
primary CNS lymphoma: an intent-to-treat
analysis. J Clin Oncol. 2003;21(22):4151-4156.
18. Abrey LE, Batchelor TT, Ferreri AJM, et al;
International Primary CNS Lymphoma
Collaborative Group. Report of an international
workshop to standardize baseline evaluation and
response criteria for primary CNS lymphoma.
J Clin Oncol. 2005;23(22):5034-5043.
19. Omuro AM, Ben-Porat LS, Panageas KS, et al.
Delayed neurotoxicity in primary central nervous
system lymphoma. Arch Neurol. 2005;62(10):
1595-1600.
20. Fazekas F, Chawluk JB, Alavi A, Hurtig HI,
Zimmerman RA. MR signal abnormalities at 1.5 T
in Alzheimer’s dementia and normal aging. AJR
Am J Roentgenol. 1987;149(2):351-356.
21. Abrey LE, Ben-Porat L, Panageas KS, et al.
Primary central nervous system lymphoma:
the Memorial Sloan-Kettering Cancer Center
prognostic model. J Clin Oncol. 2006;24(36):
5711-5715.
22. Soussain C, Choquet S, Fourme E, et al.
Intensive chemotherapy with thiotepa, busulfan
and cyclophosphamide and hematopoietic stem
cell rescue in relapsed or refractory primary
central nervous system lymphoma and intraocular
lymphoma: a retrospective study of 79 cases.
Haematologica. 2012;97(11):1751-1756.
23. Soussain C, Suzan F, Hoang-Xuan K, et al.
Results of intensive chemotherapy followed by
hematopoietic stem-cell rescue in 22 patients with
refractory or recurrent primary CNS lymphoma or
intraocular lymphoma. J Clin Oncol. 2001;19(3):
742-749.
24. Welch MR, Sauter CS, Matasar MJ, et al.
Autologous stem cell transplant in recurrent or
refractory primary or secondary central nervous
system lymphoma using thiotepa, busulfan and
cyclophosphamide [published online ahead of
print June 27, 2014]. Leuk Lymphoma. doi:
10.3109/10428194.2014.916800.
25. Cote GM, Hochberg EP, Muzikansky A, et al.
Autologous stem cell transplantation with
thiotepa, busulfan, and cyclophosphamide (TBC)
conditioning in patients with CNS involvement by
non-Hodgkin lymphoma. Biol Blood Marrow
Transplant. 2012;18(1):76-83.
26. Przepiorka D, Nath R, Ippoliti C, et al. A phase I-II
study of high-dose thiotepa, busulfan and
cyclophosphamide as a preparative regimen for
autologous transplantation for malignant
lymphoma. Leuk Lymphoma. 1995;17(5-6):
427-433.
27. Shimoni A, Smith TL, Aleman A, et al. Thiotepa,
busulfan, cyclophosphamide (TBC) and
autologous hematopoietic transplantation: an
intensive regimen for the treatment of multiple
myeloma. Bone Marrow Transplant. 2001;27(8):
821-828.
28. Anagnostopoulos A, Aleman A, Ayers G, et al.
Comparison of high-dose melphalan with a more
intensive regimen of thiotepa, busulfan, and
cyclophosphamide for patients with multiple
myeloma. Cancer. 2004;100(12):2607-2612.
29. Illerhaus G, Marks R, Ihorst G, et al. High-dose
chemotherapy with autologous stem-cell
transplantation and hyperfractionated
radiotherapy as first-line treatment of primary
CNS lymphoma. J Clin Oncol. 2006;24(24):
3865-3870.
30. Schorb E, Kasenda B, Atta J, et al. Prognosis of
patients with primary central nervous system
lymphoma after high-dose chemotherapy followed
by autologous stem cell transplantation.
Haematologica. 2013;98(5):765-770.
31. Illerhaus G, M¨uller F, Feuerhake F, Sch ¨afer AO,
Ostertag C, Finke J. High-dose chemotherapy
and autologous stem-cell transplantation without
consolidating radiotherapy as first-line treatment
for primary lymphoma of the central nervous
system. Haematologica. 2008;93(1):147-148.
32. Illerhaus G, Fritsch K, Egerer G, et al. Sequential
high dose immuno-chemotherapy followed
by autologous peripheral blood stem cell
transplantation for patients with untreated primary
central nervous system lymphoma - a multicentre
study by the Collaborative PCNSL Study Group
Freiburg. Blood. 2012;120(21). Abstract 302.
33. Colombat P, Lemevel A, Bertrand P, et al. High-
dose chemotherapy with autologous stem cell
transplantation as first-line therapy for primary
CNS lymphoma in patients younger than
60 years: a multicenter phase II study of the
GOELAMS group. Bone Marrow Transplant.
2006;38(6):417-420.
34. Montemurro M, Kiefer T, Sch¨uler F, et al. Primary
central nervous system lymphoma treated with
high-dose methotrexate, high-dose busulfan/
thiotepa, autologous stem-cell transplantation and
response-adapted whole-brain radiotherapy:
results of the multicenter Ostdeutsche
Studiengruppe Hamato-Onkologie OSHO-53
phase II study. Ann Oncol. 2007;18(4):665-671.
35. Chen YB, Batchelor T, Li S, et al. Phase 2 trial of
high-dose rituximab with high-dose cytarabine
mobilization therapy and high-dose thiotepa,
busulfan, and cyclophosphamide autologous
stem cell transplantation in patients with central
nervous system involvement by non-Hodgkin
lymphoma. Cancer. 2015;121(2):226-233.
36. Yoon DH, Lee DH, Choi DR, et al. Feasibility of
BU, CY and etoposide (BUCYE), and auto-SCT in
patients with newly diagnosed primary CNS
lymphoma: a single-center experience. Bone
Marrow Transplant. 2011;46(1):105-109.
37. Brevet M, Garidi R, Gruson B, Royer B, Vaida I,
Damaj G. First-line autologous stem cell
transplantation in primary CNS lymphoma. Eur J
Haematol. 2005;75(4):288-292.
38. Bromberg JE, Doorduijn JK, Illerhaus G,
et al. Central nervous system recurrence of
systemic lymphoma in the era of stem cell
transplantation—an International Primary Central
Nervous System Lymphoma Study Group project.
Haematologica. 2013;98(5):808-813.
39. Alimohamed N, Daly A, Owen C, Duggan P,
Stewart DA. Upfront thiotepa, busulfan,
cyclophosphamide, and autologous stem cell
transplantation for primary CNS lymphoma:
a single centre experience. Leuk Lymphoma.
2012;53(5):862-867.
40. Omuro A, Taillandier L, Chinot O, et al; ANOCEF
Group (French Neuro-Oncology Association).
Primary CNS lymphoma in patients younger than
60: can whole-brain radiotherapy be deferred?
J Neurooncol. 2011;104(1):323-330.
41. Hottinger AF, DeAngelis LM, Yahalom J, Abrey
LE. Salvage whole brain radiotherapy for
recurrent or refractory primary CNS lymphoma.
Neurology. 2007;69(11):1178-1182.
42. Nguyen PL, Chakravarti A, Finkelstein DM,
Hochberg FH, Batchelor TT, Loeffler JS. Results
of whole-brain radiation as salvage of
methotrexate failure for immunocompetent
patients with primary CNS lymphoma. J Clin
Oncol. 2005;23(7):1507-1513.
43. ClinicalTrials.gov, US National Institutes of
Health. Available at: https://clinicaltrials.gov/.
1410 OMURO et al BLOOD, 26 FEBRUARY 2015 xVOLUME 125, NUMBER 9
For personal use only.on November 4, 2015. by guest www.bloodjournal.orgFrom
online January 7, 2015 originally publisheddoi:10.1182/blood-2014-10-604561
2015 125: 1403-1410
S. Panageas, Raymond E. Baser, Geraldine Faivre, Lauren E. Abrey and Craig S. Sauter
Thomas J. Kaley, Igor T. Gavrilovic, Craig Nolan, Elena Pentsova, Christian C. Grommes, Katherine
Antonio Omuro, Denise D. Correa, Lisa M. DeAngelis, Craig H. Moskowitz, Matthew J. Matasar,
stem-cell transplant for newly diagnosed primary CNS lymphoma
R-MPV followed by high-dose chemotherapy with TBC and autologous
http://www.bloodjournal.org/content/125/9/1403.full.html
Updated information and services can be found at:
(2147 articles)Lymphoid Neoplasia (3438 articles)Free Research Articles (4194 articles)Clinical Trials and Observations
Articles on similar topics can be found in the following Blood collections
http://www.bloodjournal.org/site/misc/rights.xhtml#repub_requests
Information about reproducing this article in parts or in its entirety may be found online at:
http://www.bloodjournal.org/site/misc/rights.xhtml#reprints
Information about ordering reprints may be found online at:
http://www.bloodjournal.org/site/subscriptions/index.xhtml
Information about subscriptions and ASH membership may be found online at:
Copyright 2011 by The American Society of Hematology; all rights reserved.
of Hematology, 2021 L St, NW, Suite 900, Washington DC 20036.
Blood (print ISSN 0006-4971, online ISSN 1528-0020), is published weekly by the American Society
For personal use only.on November 4, 2015. by guest www.bloodjournal.orgFrom
... Although case reports and small case series have inherent publication bias and data heterogeneity, a meta-analysis of existing literature is still the only means of a comprehensive analysis of this disorder. Our literature analysis of 559 NL patients from 286 articles revealed that they had a shorter survival (median 12 months) compared to other extranodal lymphomas involving the nervous system, most notably primary central nervous system lymphoma (median 25 to >71 months) and intravascular lymphomatosis (median 19 months) [12][13][14][15]. This is most likely due to difficulties in establishing timely diagnosis so that appropriate treatment can be applied. ...
Article
Full-text available
Background Neurolymphomatosis is a lymphoid malignancy of the peripheral nervous system and its natural history is poorly understood. Methods We performed PubMed search and extracted clinical data for Kaplan-Meier statistics to determine outcome parameters over time. Kruskal-Wallis test was performed to compare prognostic factors. Results Our search identified 559 patients and their median age was 61 years. Median overall survival (OS) was 12.0 (range 10.0–15.0) months. Diffuse large B-cell lymphoma was the most frequent histology, involving the brachial plexus, cranial nerves, and sciatic nerve. None had molecular profiling. There was a progressive lengthening of OS in successive decades, from 0.5 (95% CI 0.0–0.8) to 26.4 (95% CI 18.0–34.8) months between 1951 and 2022 (r ² = 0.0528, p < 0.00001). Time from first treatment (treatment 1) to progression increased from 2.0 to 36.0 (95% CI 6.5–50.7) months (r ² = 0.0961, p = 0.00236). Time from symptom onset to diagnosis remained unchanged (r ² = 0.0000556, p = 0.939). Patients were most frequently treated with methotrexate, rituximab, and/or radiation either alone or in combination. Primary neurolymphomatosis had a better prognosis than secondary neurolymphomatosis. No OS difference was noted between B- and T-cell disease, but low-grade B-cell performed better than Burkitt’s lymphoma. Discussion Better outcome for patients with neurolymphomatosis is noted over time. But timely diagnosis remains a major problem that needs improvement.
... 26 patients with PCNSL were qualified for R-MPV chemotherapy (rituximab, methotrexate, procarbazine, vincristine), 11 patients received treatment according to the MATRix protocol (methotrexate, cytarabine, thiotepa, rituximab), six patients received R-HD-MTX-ARA-C (rituximab, methotrexate, cytarabine), and one received R-HD-MTX (rituximab, methotrexate) [7,8]. ...
... Birnbaum et al. [26] have suggested that the addition of rituximab significantly improves the clinical efficacy of chemotherapy. High-dose chemotherapy with autologous stem cell transplantation (HDC + ASCT) enhances the bioavailability of drugs and achieves better therapeutic effects through high-dose chemotherapy supported by hematopoietic stem cells [27]. HDC-ASCT can be considered for patients with newly diagnosed or relapsed/refractory PCNSL, particularly in younger patients (age <65 years old) [25,28]. ...
Article
Full-text available
Purpose The purpose of this retrospective study was to analyze the imaging and pathological features, treatment, and prognosis of patients with primary intraventricular lymphomas (PIL) in order to enhance physicians' understanding of the diagnosis and treatment of PIL. Methods A retrospective analysis was conducted on 13 cases of PIL that were hospitalized in our institution. Clinical and imaging data of the patients were collected and compared with the pathology data to summarize and analyze the qualitative diagnostic value of magnetic resonance (MR) features. Results Among the enrolled patients, there were nine males and four females, with an average age of (56 ± 9.0) years. The major clinical features observed in PIL patients were headache and dizziness. All 13 patients underwent plain and contrast-enhanced MR scans, revealing multiple foci in 7 cases and single foci in 6 cases. The lesions were located in the lateral ventricle in 10 cases, the third ventricle in 4 cases, and the fourth ventricle in 4 cases. Plain MR scans demonstrated an isointense or slightly hypointense signal on T1-weighted imaging (T1WI) and an isointense or slightly hyperintense signal on T2-weighted imaging (T2WI). Contrast-enhanced scans showed uniform and consistent enhancement of the tumors. Surgical treatment was performed in all patients, and postoperative pathology confirmed the presence of diffuse large B-cell lymphoma. Conclusions PIL exhibits specific imaging and pathological features, with diffuse large B-cell lymphoma being the main pathological type. Pathological examination and immunophenotype analysis serve as the gold standards for PIL diagnosis.
... Our study showed better rate of 2 year OS (67%) than that of 2 year PFS (31%), because patient with relapse lived long due to the response on the second line of the treatment. Our results follow those reported earlier, even though one of the study showed better 2 year OS (81%) in patients who were treated with combination of chemotherapy followed by autologous stem cell transplantation (15). Published prospective studies are showing encouraging results; patients who were treated with protocols similar to RTOG 93-10 (excluding radiotherapy due its toxicity) and in conjunction with Rituximab, resulted in PFS rate of 63% (11), while other study with similar regimen showed 2 year PFS of 0.57 (16). ...
Article
Background: Primary central nervous system lymphoma(PCNSL) is an aggressive, rare form of Non-Hodgkin lymphoma, characterized by the absence of systemic disease. There are limited data and no strictly defined guidelines for management of PCNSL. Objective: The aim of this study was to report a 10 year experience of PCNSL treatment, to evaluate treatment outcomes and asses Progression Free and Overall Survival of these patients. Methods: Study was conducted on the Haematology Clinic, Clinical center University of Sarajevo, BH, in the period from January 2012.-December 2022. Total sample of 24 patients were enrolled. All have undergone diagnostic surgery. Patients were treated with regimens based on High dose Methotrexate, with/without whole brain radiotherapy as consolidation. Treatment response was captured by imaging techniques. Patients who have relapsed were evaluated with imaging techniques and treated according to Methotrexate-based treatment protocols. Results: We have captured equal gender distribution. The median age of patients was 59.5 years (range 20-79). Pathohistological analysis confirmed DLBCL diagnosis in 22 patients, T cell lymphoma and anaplastic large cell lymphoma, each in 1 patient. Chemotherapy, chemotherapy combined with WBRT and radiotherapy were given to 5, 18 and 1 patients, respectively. The overall complete response rate (CR) was 87,15%. Those receiving combined modality-treatment had higher CR than those receiving chemotherapy (94,4% versus 60%). Out of 24 patients, 11 of them relapsed. The median time to relapse was 29 months (from 1 to 105). After second line of the treatment, CR was 54,5%, while 45,45% of patients died during the treatment. 4 patients relapsed for the second time with median time to relapse of 9 months (from 2 to 77). 2 year OS rate was 67%, and the median OS rate was 45,9 months. 2 year PFS rate was 31%. Conclusion: The OS and PFS rates indicate the usage of new drugs and consolidation with autologous stem cell transplantation in patients with PCNSL in order to achieve better treatment outcomes.
... Whole brain radiation therapy (WBRT) is an alternative option for patients who cannot tolerate chemotherapy. After achieving remission, high dose thiotepa followed by ASCT can be used as consolidation therapy [64]. WBRT can also be considered for consolidation in ASCT-intolerant patients. ...
Article
Full-text available
Purpose Lymphoma has become a major threat to human health. Fortunately, the diagnosis and treatment of lymphoma have developed rapidly, and research progress has emerged in an endless stream, with new drugs emerging one after another. These results are constantly rewriting guidelines changing clinical practice, need to be popularized and applied more widely. Methods This guideline has integrated consensuses reached by the Lymphoma Committee of China Anti-Cancer Association (CACA), based on China's practice, tracking previous results of the most advanced clinical researches, absorbing the latest clinical evidence, and referring to domestic and international lymphoma guidelines. Results This holistic integrative guideline of lymphoma introduces the latest progress in the diagnosis and treatment of different subtypes of lymphoma, guide the clinical application of new drugs, standardized and precise management for lymphoma patients. Conclusions CACA guidelines for holistic integrative management of lymphoma (version 2022) enhance standardization and precision of the management for lymphoma patients in China.
Preprint
Full-text available
Introduction: Primary central nervous system lymphoma (PCNSL) is a rare form of non-Hodgkin's lymphoma that primarily affects the brain, spinal cord, and/or eyes. Despite advances in treatment, PCNSL remains a formidable challenge due to its aggressive nature and limited understanding of disease progression. Recurrence of PCNSL within the central nervous system (CNS) is not uncommon, but patterns of intracranial recurrence following induction chemotherapy have not been extensively studied. This study aims to elucidate the patterns of recurrence in PCNSL, providing insight into the disease’s behavior. Methods: Patient charts and diagnostic imaging from those treated for PCNSL from 2 institutions and ≥ 18 years old from the years 2011-2023 were retrospectively reviewed. Relevant data from the charts to identify disease progression, patterns of recurrence, and survival was collected from the patient charts. The pattern of recurrence was reviewed by comparing the initial diagnostic MRI to the recurrence MRI (T1 post-contrast and T2/FLAIR sequences). The pattern of recurrence was classified as either local or distant depending on the anatomical location of the recurrent lesion. Distant recurrences were classified as those separate from the previous enhancing lesion and located outside of the initial T2/FLAIR abnormality appreciated on imaging. Local recurrences were classified as new lesions that overlapped with the primary enhancing lesion/T2/FLAIR abnormality. Results: 29 out of 47 patients met inclusion criteria with PCNSL and progressed through initial definitive treatment with high-dose methotrexate (HDMTX) or HDMTX and rituximab; with only 1 patient receiving initial management with radiation therapy. The median age at time of diagnosis was 73.0 years (range: 44.0-78.0), with 16 (55.2%) patients being male and 13 (44.8%) female. Eighteen of the 29 patients were found to have multifocal disease. 14/29 (48.3%) patients had distant recurrence only, 11/29 (37.9%) had only local recurrence, whereas 4/29 (13.8%) displayed both local and distant recurrence pattern. The median time until disease progression was 7.1 months (range: 1.3-69.0). Twelve of the 29 patients had a complete response (CR) to initial systemic therapy, 7 had a partial response (PR), and 10 patients had disease progression through HDMTX. Of the 18 patients who had multifocal disease at the time of diagnosis, 6 had disease progression through HDMTX, with 5 experiencing PR and only 7 achieved CR. Conclusion: Patterns of disease progression in patients diagnosed with PCNSL are not very well understood. In this retrospective review, we identified that the majority of disease recurrence in patients diagnosed with PCNSL who underwent HDMTX was outside or distant to the primary disease site vs solely local disease progression intracranially (62.1% vs 37.9%). Such findings need to be illustrated in a larger cohort to confirm such patterns; however, this insight raises awareness of the need for further advances in achieving higher and more durable rates of control for this rare disease.
Article
The incidence of primary central nervous system lymphoma (PCNSL) has grown recently, and neurosurgeons in Japan frequently have the opportunity to treat it. PCNSL causes cognitive impairment and has several atypical imaging findings, making it challenging to diagnose. In cases where biopsy is challenging, certain biomarkers in cerebrospinal fluid and deoxyribonucleic acid and ribonucleic acid assays via liquid biopsy have recently been reported to be significant. Rituximab, methotrexate (MTX), procarbazine, and vincristine are increasing accepted as routine therapies in Japan, and MTX-based multidrug chemotherapy is recommended as induction therapy. High-dose cytarabine is employed for subsequent consolidation therapy. High-dose whole-brain radiation therapy should be avoided as much as possible during initial therapy due to neurotoxicity, such as cognitive impairment. MTX re-challenge is effective at recurrence ; however, the time until recurrence should be considered. Recently developed Bruton's tyrosine kinase inhibitors have attracted attention and show high response rates. However, there are issues with persistence and adverse events ; therefore, further research is required its use.
Article
Objectives Treatment intensification (including consolidative high‐dose chemotherapy with autologous stem cell transplantation [HDT‐ASCT]) significantly improved outcome in primary central nervous system lymphoma (PCNSL) patients. Methods We conducted a multicenter, retrospective analysis of newly diagnosed PCNSL patients, treated with intensified treatment regimens. The following scores were evaluated in terms of overall survival (OS) and progression‐free survival (PFS): Memorial Sloan‐Kettering Cancer Center (MSKCC), International Extranodal Lymphoma Study Group (IELSG), and three‐factor (3F) prognostic score. Further, all scores were comparatively investigated for model quality and concordance. Results Altogether, 174 PCNSL patients were included. One hundred and five patients (60.3%) underwent HDT‐ASCT. Two‐year OS and 2‐year PFS for the entire population were 73.3% and 48.5%, respectively. The MSKCC ( p = .003) and 3F score ( p < .001), but not the IELSG score ( p = .06), had the discriminatory power to identify different risk groups for OS. In regard to concordance, the 3F score (C‐index [0.71]) outperformed both the MSKCC (C‐index [0.64]) and IELSG (C‐index [0.53]) score. Moreover, the superiority of the 3F score was shown for PFS, successfully stratifying patients in three risk groups, which also resulted in the highest C‐index (0.66). Conclusion The comparative analysis of established PCNSL risk scores affirm the clinical utility of the 3F score stratifying the widest prognostic spectrum among PCNSL patients treated with intensified treatment approaches.
Article
Central nervous system lymphoma (CNSL) is a rare and aggressive malignancy that primarily affects the brain, spinal cord, and meninges. This article provides a comprehensive overview of the current understanding of CNSL encompassing its epidemiology, pathophysiology, clinical presentation, diagnosis, treatment modalities, and prognosis. Although the main focus is on primary CNS lymphoma (PCNSL), ocular lymphoma, primary leptomeningeal lymphoma, and secondary CNS lymphoma are also discussed. The pathobiology of CNSL involves the infiltration of malignant lymphocytes within the CNS parenchyma or leptomeninges. Various risk factors and immunological mechanisms contribute to its development, including immunodeficiency states, chronic inflammation, and genomic alterations. Accurate diagnosis is crucial for appropriate management, given the heterogeneous clinical presentation. The neuroimaging, systemic imaging, and other modalities for diagnosis and evaluation for extent of disease involvement will be discussed. Additionally, the importance of histopathological examination, cerebrospinal fluid (CSF) analysis, and molecular testing in confirming the diagnosis and guiding treatment decisions are highlighted. The treatment landscape for CNSL has evolved significantly. Therapeutic approaches encompass a multimodal strategy combining high-dose methotrexate-based chemotherapy, consolidation with whole-brain radiation therapy, and high-dose chemotherapy with stem cell rescue. Recent advancements in targeted therapies and immunomodulatory agents offer promising avenues for future treatment options. We review the clinical outcomes and prognostic factors influencing the survival of CNSL patients, including age, performance status, disease stage, and genetic abnormalities.
Article
Introduction. Relapses of primary mediastinal large B-cell lymphoma (PMBСL) with involvement of the central nervous system (CNS) are usually incurable. Therapy tactics in patients with PMBCL with relapse in the CNS have not been defined. Aim: to present clinical observations of the treatment of relapses of PMBCL with CNS involvement using the “R-PML-CNS-2022” protocol. Main findings. Two clinical observations of patients with relapses of PMBCL with involvement of the CNS are presented. In both cases, the “R-PML—CNS-2022” protocol was used as a second-line treatment, and a complete remission of the disease was achieved. In the first case, within 6 months after auto-HCT, maintenance therapy with pembrolizumab was performed. Complete remission of the disease continues for 15 months. In the second observation, maintenance therapy with pembrolizumab was also started, however, due to the development of autoimmune complications after 2 cycles, treatment was discontinued. Complete remission of the disease continues for 13 months. Conclusion. The development of a new strategy for the treatment of relapses of PMLCL with CNS lesion according to the “R-PML—CNS-2022” protocol is a potentially curative approach in this group of patients.
Article
Full-text available
PURPOSEConcerns regarding neurocognitive toxicity of whole-brain radiotherapy (WBRT) have motivated development of alternative, dose-intensive chemotherapeutic strategies as consolidation in primary CNS lymphoma (PCNSL). We performed a multicenter study of high-dose consolidation, without WBRT, in PCNSL. Objectives were to determine: one, rate of complete response (CR) after remission induction therapy with methotrexate, temozolomide, and rituximab (MT-R); two, feasibility of a two-step approach using high-dose consolidation with etoposide plus cytarabine (EA); three, progression-free survival (PFS); and four, correlation between clinical and molecular prognostic factors and outcome. PATIENTS AND METHODS Forty-four patients with newly diagnosed PCNSL were treated with induction MT-R, and patients who achieved CR received EA consolidation. We performed a prospective analysis of molecular prognostic biomarkers in PCNSL in the setting of a clinical trial.ResultsThe rate of CR to MT-R was 66%. The overall 2-year PFS was 0.57, with median follow-up of 4.9 years. The 2-year time to progression was 0.59, and for patients who completed consolidation, it was 0.77. Patients age > 60 years did as well as younger patients, and the most significant clinical prognostic variable was treatment delay. High BCL6 expression correlated with shorter survival. CONCLUSIONCALGB 50202 demonstrates for the first time to our knowledge that dose-intensive consolidation for PCNSL is feasible in the multicenter setting and yields rates of PFS and OS at least comparable to those of regimens involving WBRT. On the basis of these encouraging results, an intergroup study has been activated comparing EA consolidation with myeloablative chemotherapy in this randomized trial in PCNSL, in which neither arm involves WBRT.
Article
302 Background Primary central nervous system lymphoma (PCNSL) is an aggressive non-Hodgkin′s lymphoma with poor prognosis. Addition of methotrexate (MTX) to whole brain radiotherapy (WBRT) has improved the prognosis of patients (pts) with PCNSL, but a significant proportion are still not cured. Preliminary reports suggested that dose-intensified chemotherapy (HDT) and autologous stem-cell transplantation (ASCT) are highly effective in the treatment of newly-diagnosed PCNSL in younger pts. To strengthen the evidence of this approach, we initiated a prospective multicenter phase II study with early HDT and ASCT to investigate efficacy, safety and survival. This trial is registered at ClinicalTrials.gov (NCT 00647049). Methods Immunocompetent pts <65 years with untreated biopsy proven PCNSL were eligible. Induction treatment consisted of 4 repetitive cycles of MTX (8g/m2) and 2 cycles cytarabine (2×3g/m2) and thiotepa (TT, 40mg/m2). Rituximab (375mg/m2) was added by amendmend after 2 included patients; it was given on day −7 before induction treatment and before each chemotherapy cycle. After the 2nd cycle cytarabine/TT stem-cells were collected after mobilisation with rG-CSF. The HDT regimen included carmustine (400mg/m2) and TT (4×5mg/kgBW) prior ASCT. Primary endpoint was complete remission (CR) 30 days after ASCT. Secondary end-points were overall-survival (OS), duration of response and toxicity. Patients not in complete remission after HDT and ASCT received WBRT. Results From 2007 to 2011 79 pts (44 female, 35 male) were enrolled from 18 German centers and evaluable for analysis (median age 55 years, range 20–66). Seventy eight pts had aggressive B-cell lymphomas and one T-cell-lymphoma. Median Karnofsky performance status at diagnosis was 90% (range 30–100). After induction treatment, 73 of 76 (96%) evaluable pts responded, (26,9% CR, 55,7 PR). Seventy-three pts (96%) received HDT and ASCT according to protocol. Six pts were treated off-protocol due to low performance status (n=1), progressive disease (n=1) and infectious complications (n=4). Regarding the primary endpoint, CR was achieved in 77% and partial remission (PR) in 14% of patients (overall response rate 91%) after HDT and ASCT. Ten pts in PR after HDT and ASCT received consolidating WBRT. After a median follow-up of 28.8 months (range 1–63 mo) 1 and 2 years OS was 92% and 87%, respectively. Myelotoxicity was the most frequent CTC grade 3–4 toxicity with grade 3–4 infections in 41/73 pts (56,2%) during the transplant-phase. Two patients had lethal infectious complications during induction treatment with cytarabine/TT, three further pts died after HDT and ASCT due to severe infection (n=1), renal failure (n=1) and pneumonitis (n=1). Further results will be presented. Conclusion Sequential MTX-based immuno-chemotherapy followed by carmustine/TT containing HDT and ASCT is highly effective and feasible in younger patients. Treatment related toxicity is of concern and comparable to non-high-dose protocols. Further randomised trials to compare HDT with conventional CT are needed. Disclosures Illerhaus: Riemser: Honoraria.
Article
PURPOSE: The use of preradiotherapy (RT) methotrexate (MTX) has improved disease control and survival in patients with primary CNS lymphoma (PCNSL). The reported protocol was designed to optimize and enhance the chemotherapeutic component of treatment. PATIENTS AND METHODS: Fifty-two patients were treated with five cycles of high-dose MTX 3.5 g/m², procarbazine 100 mg/m²/d, and vincristine 1.4 mg/m². Thirty patients received whole-brain RT (45 Gy). Twenty-two older patients deferred RT to diminish the risk of delayed neurotoxicity; these patients are compared with 12 older patients who completed the entire treatment regimen. Most patients (n = 35) received high-dose cytarabine after RT. RESULTS: Objective response rate to the induction chemotherapy regimen was 90%; overall median survival is 60 months. Grade 3 or 4 myelosuppression was seen in 30 patients, primarily in association with cytarabine; grade 3 nephrotoxicity due to MTX was seen in two patients. Older patients had similar median survival with or without the addition of RT: 32 versus 33 months, respectively. However, late neurotoxicity was significantly more common in those older patients who received RT (P = .00004). Patients younger than 60 years who received the complete treatment regimen have not reached median disease-free or overall survival. CONCLUSION: Increasing the dose of MTX and adding procarbazine and vincristine improved disease control and overall survival in patients with newly diagnosed PCNSL. Younger patients in particular fared extremely well with this treatment regimen. In older patients, deferring whole-brain RT did not compromise overall survival but did reduce treatment-related toxicity.
Article
Purpose: To evaluate response rate, response duration, overall survival (OS), and toxicity in primary CNS lymphoma (PCNSL) after systemic and intraventricular chemotherapy with deferred radiotherapy. Patients and Methods: From September 1995 to July 2001, 65 consecutive patients with PCNSL (median age, 62 years) were enrolled onto a pilot and phase II study evaluating chemotherapy without radiotherapy. A high-dose methotrexate (MTX; cycles 1, 2, 4, and 5) and cytarabine (ARA-C; cycles 3 and 6)–based systemic therapy (including dexamethasone, vinca-alkaloids, ifosfamide, and cyclophosphamide) was combined with intraventricular MTX, prednisolone, and ARA-C. Results: Sixty-one of 65 patients were assessable for response. Of these, 37 patients (61%) achieved complete response, six (10%) achieved partial response, and 12 (19%) progressed under therapy. Six (9%) of 65 patients died because of treatment-related complications. Follow-up is 0 to 87 months (median, 26 months). The Kaplan-Meier estimates for median time to treatment failure (TTF) and median OS were 21 months and 50 months, respectively. For patients older than 60 years, median survival was 34 months, and the median TTF was 15 months. In patients younger than 61 years, median survival and median TTF have not been reached yet; the 5-year survival fraction is 75%. Systemic toxicity was mainly hematologic. Ommaya reservoir infection occurred in 12 patients (19%), and permanent cognitive dysfunction possibly as a result of treatment occurred in only two patients (3%). Conclusion: Primary chemotherapy based on high-dose MTX and ARA-C is highly efficient in PCNSL. Response rate and response duration in this series are comparable to the response rates and durations reported after combined radiotherapy and chemotherapy. Neurotoxicity was infrequent.
Article
BACKGROUND High-dose thiotepa, busulfan, and cyclophosphamide (TBC) with autologous stem cell transplantation (ASCT) has been used in patients with central nervous system (CNS) involvement by non-Hodgkin lymphoma (NHL). Despite limited penetration into the CNS, rituximab is active in primary CNS NHL. Therefore, high-dose rituximab was combined with TBC for ASCT in patients with CNS NHL.METHODSA single-arm phase 2 trial using high-dose rituximab with cytarabine for stem cell mobilization followed by high-dose rituximab combined with thiotepa, busulfan, and cyclophosphamide (R-TBC) for ASCT was conducted. Doses of rituximab at 1000 mg/m2 were given on days 1 and 8 of mobilization and on days −9 and −2 of TBC. The primary endpoint was efficacy.RESULTSThirty patients were enrolled. Eighteen patients had primary CNS NHL (12 with complete remission (CR)/first partial remission (PR1) and 6 with CR/PR2), and 12 patients had secondary CNS lymphoma (5 with CR/PR1 and 7 with CR/PR2 or beyond). All patients were in partial or complete remission. Twenty-nine patients proceeded to R-TBC ASCT. Two patients developed significant neurotoxicity. The 100-day nonrelapse mortality rate was 0%, and 1 patient died because of nonrelapse causes 5 months after ASCT. For all patients, at a median follow-up of 24 months (range, 12-40 months), the estimated 2-year progression-free survival rate was 81% (95% confidence interval, 59%-92%), and the 2-year overall survival rate was 93% (95% confidence interval, 76%-98%). There were no relapses or deaths among the 18 patients with primary CNS lymphoma.CONCLUSIONS For patients with CNS involvement by B-cell NHL and especially for patients with primary CNS NHL, R-TBC ASCT shows encouraging activity and merits further study. Cancer 2014. © 2014 American Cancer Society.
Article
ABSTRACT The prognosis for patients with central nervous system (CNS) involvement by recurrent or refractory diffuse large B-cell lymphoma is poor, with overall-survival (OS) of 4-10 months. High-dose chemotherapy (HDC) and autologous stem-cell transplant (ASCT) is a potential treatment alternative. We reviewed patients with recurrent primary (PCNSL) or secondary (SCNSL) CNS lymphoma referred for consolidation HDC-ASCT utilizing thiotepa, busulfan and cyclophosphamide (TBC). Among the 17 patients included, all had achieved a complete remission after salvage induction chemotherapy, which incorporated methotrexate in 82% of patients. Two patients failed stem-cell harvesting and 15 (88%) underwent transplant. The estimated 3-year progression-free survival (PFS) and OS were both 93% (95%CI 61-99%); median PFS and OS were not reached. There was no transplant-related mortality. These results confirm the benefit of TBC followed by ASCT in select patients with recurrent PCNSL and suggest a potential role for the regimen in those with SCNSL; further investigation is warranted.
Article
A multicenter phase II study was conducted to assess the efficacy of rituximab, methotrexate, procarbazine, and vincristine (R-MPV) followed by consolidation reduced-dose whole-brain radiotherapy (rdWBRT) and cytarabine in primary CNS lymphoma. Patients received induction chemotherapy with R-MPV (five to seven cycles); those achieving a complete response (CR) received rdWBRT (23.4 Gy), and otherwise, standard WBRT was offered (45 Gy). Consolidation cytarabine was given after the radiotherapy. The primary end point was 2-year progression-free survival (PFS) in patients receiving rdWBRT. Exploratory end points included prospective neuropsychological evaluation, analysis of magnetic resonance imaging (MRI) white matter changes using the Fazekas scale, and evaluation of the apparent diffusion coefficient (ADC) as a prognostic factor. Fifty-two patients were enrolled, with median age of 60 years (range, 30 to 79 years) and median Karnofsky performance score of 70 (range, 50 to 100). Thirty-one patients (60%) achieved a CR after R-MPV and received rdWBRT. The 2-year PFS for this group was 77%; median PFS was 7.7 years. Median overall survival (OS) was not reached (median follow-up for survivors, 5.9 years); 3-year OS was 87%. The overall (N = 52) median PFS was 3.3 years, and median OS was 6.6 years. Cognitive assessment showed improvement in executive function (P < .01) and verbal memory (P < .05) after chemotherapy, and follow-up scores remained relatively stable across the various domains (n = 12). All examined MRIs (n = 28) displayed a Fazekas score of ≤ 3, and no patient developed scores of 4 to 5; differences in ADC values did not predict response (P = .15), PFS (P = .27), or OS (P = .33). R-MPV combined with consolidation rdWBRT and cytarabine is associated with high response rates, long-term disease control, and minimal neurotoxicity.