Vous êtes sur la page 1sur 8

Leukemia & Lymphoma, February 2010; 51(2): 261–268

ORIGINAL ARTICLE: CLINICAL

Central nervous system involvement at presentation in childhood acute


lymphoblastic leukemia: management experience and lessons

RAM K. MARWAHA, KETAN P. KULKARNI, DEEPAK BANSAL, & AMITA TREHAN

Division of Pediatric Hematology–Oncology, Advanced Pediatric Center, PGIMER, Chandigarh, India


Leuk Lymphoma Downloaded from informahealthcare.com by Michigan University on 11/08/14

(Received 28 July 2009; revised 30 October 2009; accepted 3 November 2009)

Abstract
This study was designed to analyze the spectrum of central nervous system (CNS) disease at diagnosis, traumatic lumbar
puncture (TLP), role of cranial irradiation, prognostic parameters, and survival outcome in patients with CNS involvement
amongst 747 patients with acute lymphoblastic leukemia managed at our center. Twenty-five and six patients had CNS
disease and TLP, respectively. Patients with CNS involvement had significantly higher mean presenting leukocyte count
( p ¼ 0.021) and incidence of hyperleukocytosis ( p ¼ 0.01) compared to those without it. The outcome was poor with three
For personal use only.

patients in continuous complete-remission, nine relapsers, eight deaths, and eight therapy defaulters. Three patients did not
opt for therapy. CNS involvement was significantly associated with inferior survival by log-rank ( p ¼ 0.03) analysis but not by
Cox-multivariate ( p ¼ 0.145) analysis. CNS involvement is a high-risk indicator. Poor outcome in our cohort indicates the
need for the revaluation of our treatment protocols with the inclusion of risk-stratified systemic therapy, categorization of
CNS involvement into CNS1/CNS2/CNS3, and appropriate use of intrathecal therapy.

Keywords: Central nervous system, event free survival, prognostic factors, relapse

PGIMER, Chandigarh from January 1990 to De-


Introduction
cember 2005 were analyzed. Information concerning
Acute lymphoblastic leukemia (ALL) with high-risk demographic profile, symptoms, clinical features,
features has an inferior outcome [1,2]. High-risk laboratory investigations, therapy administered, and
indicators, factors influencing treatment and out- outcome was recorded. The study was approved by
come of ALL in developing countries have not been Ethics Review Board of PGIMER, Chandigarh. The
adequately studied. Central nervous system (CNS) follow-up data are as of February 2008 with a mean
disease at presentation and lumbar puncture (LP) follow-up duration of 66 months.
have been associated with increased risk and poorer CNS status at presentation was classified into (a)
survival outcome [1–4]. However, data from devel- CNS1: absence of lymphoblasts in cerebrospinal
oping nations describing CNS involvement at pre- fluid (CSF) (b) CNS2/3: (i) presence of unequivocal
sentation and prognostic importance are scant [5,6]. blasts in cytocentrifuged CSF irrespective of CSF cell
In this communiqué, we describe the clinical count (Group A) or (ii) overt CNS leukemia: the
spectrum and our experience of managing patients presence of new onset focal neurological deficit or
with ALL with CNS involvement at presentation and raised intracranial pressure at presentation (Group
lessons learnt thereby. B), and (c) traumatic lumbar puncture (TLP):
defined as 10 or more erythrocytes/mL with identifi-
able lymphoblasts in CSF (Group C) [1].
Material and methods
Bulk disease was defined as the presence of one
Seven hundred forty-seven children, age1–14 years, or more of the following: enlarged lymph node
with ALL enrolled in the pediatric oncology clinic of (42 cm), hepatomegaly (45 cm), and splenomegaly

Correspondence: Dr. Ram Kumar Marwaha, Professor, Division of Pediatric Hematology–Oncology, Advanced Pediatric Center, PGIMER, Sec 12,
Chandigarh 160012, India. Tel: þ91-172-2755303/09815466662. Fax: þ91-172-2744450. E-mail: rammarwaha1@rediffmail.com or
ketanpkulkarni@gmail.com

ISSN 1042-8194 print/ISSN 1029-2403 online ! 2010 Informa Healthcare USA, Inc.
DOI: 10.3109/10428190903470323
262 R. K. Marwaha et al.

(45 cm). High-risk disease: Age 52 or 410 years, after January 2001, CNS irradiation was administered
presenting leukocyte count (PLC) 4 50.0 6 109/L, only to patients with CNS disease or with ‘high-risk’
bulk disease, T cell disease. Till recently, immuno- disease. The change in protocol after January 2001
phenotyping and cytogenetics were not routinely was a move-forward in our endeavor to cure patients
available for risk assessment. with ALL in keeping with the contemporary proto-
cols. Owing to risk concerns of irradiation, young
children under 3 years of age received intrathecal
Treatment protocol
triple therapy (methotrexate, cytosine arabinoside
The treatment protocol used at our center was a and hydrocortisone) at 12 weekly intervals till they
modified version of UKALL X protocol [7–9]. attained the age of 3 years. The dose of intrathecal
Remission induction chemotherapy consisted of methotrexate was 8, 10, and 12 mg in children under
vincristine, prednisolone, L-asparaginase, and in- 2, 2–3, and 43 years of age, respectively.
trathecal methotrexate. More recently (after 2000), The maintenance therapy included daily oral 6-
patients with high-risk disease and/or those affording mercaptopurine (60–75 mg/m2/day), weekly oral
Leuk Lymphoma Downloaded from informahealthcare.com by Michigan University on 11/08/14

the drug were administered doxorubicin in addition methotrexate (15–20 mg/m2/week) and 4 weekly
to the above drugs during remission induction. All pulses of vincristine with prednisolone (40 mg/m2/
patients received either an early or an early and day) or dexamethasone (6 mg/m2/day). The total
delayed intensification (vincristine, prednisolone or duration of therapy was 27 months after attaining
dexamethasone, daunomycin and thioguanine) in the complete remission (CR). Intermediate- or high-
form of 5-day blocks. dose systemic methotrexate was not used in this
CNS-directed therapy (weeks 9–12) in patients treatment protocol.
with/without CNS disease during the years of accrual
is detailed in Table I. Two hundred fifty-two and 280
Lumbar puncture
patients were treated prior to and after January 2001,
For personal use only.

respectively. After January 2001, all patients received LP was performed by an experienced clinician under
14–15 doses of intrathecal methotrexate in contrast to deep sedation after appropriate informed consent in a
6 doses of intrathecal therapy prior to it. Similarly, dedicated area. The patients were kept nil by mouth

Table I. CNS directed therapy in patients with/without CNS involvement at presentation.

Patients January 1990 to December 2000 January 2001 onwards

Patients with Intrathecal methotrexate: six doses [three doses during Intrathecal methotrexate: six doses
CNS disease at induction (weeks 1, 3 and 4); three doses during CNS
presentation directed therapy (weeks 9–12)]
Cranial irradiation: 18 Gray in 10 fractions during weeks Cranial irradiation: 18 Gray in 10 fractions during
9–12 of therapy weeks 9–12 of therapy. Three monthly intrathecal
methotrexate during maintenance phase till end of
therapy (eight to nine doses)
Patients without Intrathecal methotrexate: six doses [three doses during Intrathecal methotrexate: six doses
CNS disease at induction (weeks 1,3 and 4); three doses during CNS
presentation directed therapy (weeks 9–12)]
Cranial irradiation: 18 Gray in 10 fractions during weeks Cranial irradiation: 18 Gray in 10 fractions during
9–12 of therapy weeks 9–12 of therapy to patients with ‘high-risk’
disease. Three monthly intrathecal methotrexate
during maintenance phase till end of therapy
(eight to nine doses)
Traumatic Intrathecal methotrexate: six doses [three doses during Weekly intra-thecal triple (methotrexate, cytosine
lumbar induction (weeks 1,3 and 4); three doses during CNS arabinoside and hydrocortisone) therapy 6 four
puncture directed therapy (weeks 9–12)] doses or till clear CSF
Cranial irradiation: 18 Gray in 10 fractions during weeks Intrathecal methotrexate: three doses (during weeks
9–12 of therapy 9–12 of therapy). Three monthly intrathecal
methotrexate during maintenance phase till end of
therapy (eight to nine doses)
Cranial irradiation: 18 Gray in 10 fractions during
weeks 9–12 of therapy to patients with ‘high-risk’
disease

CNS, central nervous system; CSF, cerebrospinal fluid.


Central nervous system involvement in childhood leukemia 263

for 3–4 h prior to the procedure. Midazolam with and without CNS disease at presentation.
(0.1 mg/kg, repeated as necessary) and/or ketamine Cumulative risk of different types of relapse after
(1 mg/kg, repeated if necessary) were used. LP was remission induction was estimated using the Kalb-
performed with the appropriate size of LP/spinal fleisch and Prentice test and the functions were
needle. Intrathecal chemotherapeutic agents were compared with Gray’s test [10,11]. Deaths were
diluted to volumes up to 5 ml and administered considered as competing events in the estimation of
slowly over 2–3 min whilst diluting with CSF during cumulative incidence for each type of relapse. Cox
administration. In patients receiving triple intrathecal multivariate regression analysis was applied to the
chemotherapy, the medications were mixed prior to predictor variables to identify the factors significantly
administration. Post-procedure, patients were kept in associated with inferior survival outcome.
supine or semiprone position for 30 min to 1 h or till
they were fully awake and observed for headache.
Results
The above protocol was followed during first and
each of the subsequent LPs. Out of the 747 patients accrued, 31 (4.14%) patients
Leuk Lymphoma Downloaded from informahealthcare.com by Michigan University on 11/08/14

Based on the time from the initiation of therapy, had CNS involvement at presentation. There were
relapses were categorized as [10]: (a) very early: 13, 12, and 6 patients with CNS2/3 (Group A), overt
relapse within 18 months of diagnosis, whilst on CNS leukemia (Group B), and TLP (Group C),
chemotherapy, (b) early: relapse after 18 months of respectively. The rate of TLP was 0.8% (n ¼ 6 out of
diagnosis and within 6 months of treatment comple- 747). The mean age at presentation was 69.8 + 15.6
tion, and (c) late: relapse 46 months after treatment months (range: 12–157 months) while the male:
completion. female ratio was 2.75:1. The salient characteristics of
Groups A, B, and C patients are detailed in
Tables II–IV, respectively. Among patients with
Statistical analysis
overt CNS leukemia, cases 7, 9, and 10 had overt
For personal use only.

The statistical analysis was performed from Decem- testicular disease, renal failure, and lytic lesions in
ber 2008 onwards. The percentage, mean, and range dorso-lumbar vertebrae, respectively at diagnosis.
were used to describe continuous and categorical The mean age, PLC, and platelet count at
variables. Qualitative variables in the two groups presentation in Groups A, B, and C are shown in
were compared using the Chi-square test. Quantita- Table V. The results of the comparative analysis of
tive variables were compared using the Student ‘t’ salient clinical and laboratory characteristics in
test. The event free survival (EFS) was calculated by patients with/without CNS disease at presentation
Kaplan–Meier method. EFS was derived from the are enlisted in Table VI. The mean PLC and
date of commencement of treatment to date of the incidence of hyperleukocytosis in patients with
last follow-up or occurrence of an event (induction CNS disease at presentation were significantly higher
death, induction failure, remission death, relapse, or compared to the other patients.
treatment default). Log-rank test was used to Twenty-eight out of 31 patients opted for therapy.
compute ‘p’ value for survival analysis in patients The outcome was dismal (Tables II–V). At the time of

Table II. CNS2/3 status at presentation.

Age Bulk Mediastinal PLC Platelet count EFS


Case (months) Gender disease adenopathy Hemoglobin (6109/L) (6109/L) Outcome (months)

1 48 Female 7 7 6.0 80.0 74.0 Death 0.2


2 24 Female 7 7 6.1 24.7 58.0 Loss to follow-up (in CCR) 29.0
3 132 Male þ 7 5.0 0.8 31.0 CNS relapse 19.0
4 12 Male þ 7 5.3 64.0 37.0 BM relapse 14.0
5 36 Female 7 7 4.1 120.0 10.0 Treatment default 15.0
6 132 Male 7 þ NA NA NA Death 0.7
7 12 Male þ 7 10.0 90.0 119.0 Death 0.5
8 84 Female þ 7 4.2 160.0 11.0 Death 0.8
9 48 Male þ 7 12.0 10.9 358.0 Treatment default 0.1
10 24 Male 7 þ 14.1 28.7 21.0 Death 22.0
11 108 Male 7 7 6.0 260.0 125.0 BM relapse 12.0
12 24 Female 7 7 12.1 144.0 22.0 BM relapse 9.0
13 84 Male þ 7 13.3 5.8 219.0 BM þ CNS þ ocular relapse 38.0

BM, bone marrow; CNS, central nervous system; CCR, Continuous complete remission; EFS, event free survival; PLC, presenting
leukocyte count; NA, Not available.
264 R. K. Marwaha et al.

Table III. Patients with Overt CNS leukemia at presentation.

CNS feature at Bulk Platelet


Case Age Gender SDI presentation disease Hemoglobin PLC count Outcome EFS

1 62 Male 90 Right facial palsy No 8.0 14.4 150.0 Isolated ocular relapse 47
2 87 Male 45 Left facial palsy No 11.1 266.8 18.0 BM þ extrame-dullary 5
relapse
3 124 Male 6 Left hemiparesis, Yes 7.6 420.3 41.0 Did not opt for treatment –
left facial palsy, right
oculomotor palsy
4 110 Male 10 Left facial palsy No 10.3 15.5 29.0 Treatment default 0.2
5 39 Male 87 Paraplegia No 8.1 4.2 78.0 Did not opt for treatment –
6 132 Male 45 Paraplegia Yes 5.2 19.0 25.0 Treatment default 0.4
7 36 Male 120 Paraplegia Yes 7.3 11.9 22.0 Treatment default 0.7
8 127 Male 150 Paraplegia Yes 7.0 8.2 829.0 CCR 26
9 63 Male 90 Conus-medullaris syndrome No 4.3 0.8 19.0 Death 2
Leuk Lymphoma Downloaded from informahealthcare.com by Michigan University on 11/08/14

10 122 Female 60 Decreased hearing No 11.0 20.0 154.0 Did not opt for treatment –
11 14 Male 7 Altered sensoruim Yes 7.0 223.0 5.0 Death before induction –
12 84 Male 4 Raised intracranial pressure 5.0 356.5 43.0 Treatment default 0.2

BM, bone marrow; CCR, continuous complete remission; CNS, central nervous system; EFS, event free survival; SDI, symptom diagnosis
interval; PLC, presenting leukocyte count.

Table IV. Patients with TLP at diagnosis.

Age Bulk Mediastinal PLC Platelet count EFS


Case (months) Gender disease adenopathy Hemoglobin (6109/L) (6109/L) Outcome (months)
For personal use only.

1 49 Female 7 7 12.0 105.0 43.0 Defaulted therapy 0.6


2 38 Male 7 þ 4.2 20.0 5.0 Isolated testicular relapse 25
3 26 Male þ 7 7.1 17.0 26.0 Defaulted therapy 0.4
4 88 Male þ 7 10.3 2.0 450.0 (In CCR) Lost to follow-up 32.0
5 157 Female þ 7 6.7 115.0 70.0 Death 17.0
6 39 Male þ 7 2.5 210.0 8.0 BM þ CNS relapse 7.0

BM, bone marrow; CNS, central nervous system; CCR, continuous complete remission; EFS, event free survival; PLC, presenting leukocyte
count.

Table V. Comparison of salient characteristics in Groups A, B, and C.

Parameter Group A (n ¼ 13) Group B (n ¼ 12) Group C (n ¼ 6)

Age (mean) 59.1 + 24.3 83.4 + 23.2 66.2 + 40.3


PLC (6109/L) (mean) 82.4 + 45 113.4 + 90.6 78.2 + 65.6
Platelet count (6109/L) (mean) 90.4 + 60.3 117.7 + 132.5 100.3 + 141.3
High risk disease (%) 9 (69.2) 8 (66.7) 5 (83.3)
Death 5 2 1
Relapse 5 2 2
Treatment default 2 4 2

PLC, presenting leukocyte count.

analysis, only three patients with CNS disease were in CNS þ ocular. All patients with BM relapse (isolated
continuous CR. Two of them were lost to follow-up or combined) were very early relapsers. Isolated
while a solitary patient was nearing completion of testicular and isolated CNS relapses were ‘early’
therapy. Death and relapse were documented in eight while ocular relapses (isolated or combined) occurred
(29.6%) and nine (33.3%) patients, respectively ‘late’. Incidence of CNS relapse was 11.1% (n ¼ 3) in
(Table V). There were three isolated bone marrow comparison to 8.3% (42 out of 502 patients who
(BM) and one each of isolated CNS, isolated opted for therapy) in patients without CNS involve-
testicular and isolated ocular relapses. Combined ment at diagnosis (p ¼ 0.49). Eight patients defaulted
relapse was observed in three patients: one in BM þ to therapy (Tables IV and V). There was a significant
CNS, one in BM þ extramedullary, and one in BM þ (p ¼ 0.008) improvement in the CNS relapse rate
Central nervous system involvement in childhood leukemia 265

Table VI. Comparison of salient clinical characteristics in patients with and without CNS involvement.

Patients with CNS Patients with CNS1 status


Parameter disease at presentation (n ¼ 31) at presentation (n ¼ 716) p value

Age (months) (range) 69.8 + 15.6 (12–157) 67.2 + 1.41 0.9


Male:female ratio 2.75:1 3.18:1 0.526
SDI* (days) 48.6 57.2 0.417
Bulk disease (%) 50 (n ¼ 15) 40.1 (n ¼ 287) 0.195
Mediastinal adenopathy (%) 16.6 (n ¼ 5) 13.0 (n ¼ 93) 0.400
Hemoglobin (g/dL) 7.52 7.08 0.418
PLC (6109/L) (range) 93.9 + 41.9 (0.8–420.3) 45.8 + 6.0 0.021
Leukocyte count 4100 6 109/L (%) 11 (35.4) 109 (14.6) 0.01
Platelet count (6109/L) (range) 103.3 + 62.6 (5.0–829.0) 78.2 + 6.6 0.238

SDI, symptom diagnosis interval; CNS, central nervous system; PLC, presenting leukocyte count.
Leuk Lymphoma Downloaded from informahealthcare.com by Michigan University on 11/08/14

after the change of protocol in January 2001 from


11.5% (29 out of 252) to 5.7% (16 out of 280).
In the entire cohort, the cumulative risk of isolated
BM, CNS, and testicular relapse at 5 years follow-
up was 13.6 + 2.1, 6.8 + 1.3, and 4.3 + 0.9%,
respectively while that of combined relapse (BM
and/or CNS and/or testicular) was 7.8 + 1.6%. The
EFS in patients with and without CNS involvement
is depicted in Figure 1. Survival in patients with CNS
For personal use only.

involvement at presentation was significantly ( p ¼


0.03 by log-rank test) inferior as against patients
without CNS disease. Table VII depicts the sig-
nificance and odds ratio of the predictor variables for
survival in 530 patients who opted for therapy
according to Cox model. CNS involvement at
presentation was not significantly associated with
adverse outcome (p ¼ 0.145) while CNS irradiation Figure 1. EFS in patients with and without CNS involvement at
significantly (p ¼ 0.001) improved survival. Further- presentation.
more, high PLC and prolonged symptom diagnosis
interval (SDI) were other significant predictors of Table VII. Results for Cox multivariate regression model to
inferior survival outcome. determine the impact of predictor variables on survival.

95% CI for
Discussion odds ratio

Although cure rates of ALL in developed nations are Parameter p value Odds ratio Lower Upper
approaching 80–90% with contemporary risk-strati-
CNS involvement 0.145 1.551 0.860 2.796
fied protocols, survival outcome in developing at presentation
nations continues to remain modest [12–17]. Gender 0.096 0.760 0.551 1.050
Furthermore, data from resource-strained nations CNS irradiation 0.001 0.114 0.085 0.152
concerning incidence of CNS leukemia at diagnosis, Age 0.236 1.117 0.930 1.340
SDI 0.002 1.664 1.316 2.103
management experience, and impact on outcome are
PLC 0.001 1.360 1.208 1.530
largely unavailable. Moreover, CNS disease at Platelet count 0.083 0.855 0.716 1.021
diagnosis and CNS relapse continue to pose a Bulk 0.238 0.843 0.635 1.119
significant challenge in the endeavor to deliver cure Mediastinal adenopathy 0.09 0.757 0.550 1.102
to children with ALL even in resource-plenty nations
CI, confidence interval; CNS, central nervous system; SDI,
[18,19]. Our study was conducted in an attempt to symptom diagnosis interval; PLC, presenting leukocyte count.
address these concerns.
Authors have reported CNS2 and CNS33 status in
about 15–20 and 3% of children with ALL, 1.3% of the cases [5]. We observed CNS disease in
respectively [13]. On the contrary, in a study from 4.14% of the cases. Facial palsy, other cranial nerve
India, CNS involvement was reported in merely palsies, paraplegia, and deafness have been reported
266 R. K. Marwaha et al.

as an initial manifestation of ALL in isolated case without CNS involvement, were the major challenges
reports or very small case series [20–26]. We encountered. Sepsis and/or bleed were contributory
observed four cases each of unilateral facial palsy to majority of the deaths. In the entire cohort
and paraplegia. In addition, occurrence of conus- financial constraints, poor socioeconomic status,
medullaris syndrome and hearing defect in solitary illiteracy, distant locales from a dedicated cancer
cases was noteworthy. In view of these varied treatment facility, faith in alternate systems of
manifestations of CNS disease, a diagnostic delay medicine, and insufficient facilities for shared and
is likely and a high index of suspicion should be acute care were the major hindrances encountered
maintained for prompt diagnosis and management. locally [17]. We observed that even in patients with
Gajjar et al. reported a TLP rate of 16% and a 10% CNS involvement, very early BM relapse was the
rate of bloody LP. In comparison, our rates for TLP commonest while CNS relapse was more frequent
were far lower (0.8%) [27]. Small numbers preclude than other patients with ALL. These observations
identification of risk factors although high PLC and indicate the need of reappraisal of our treatment
thrombocytopenia and bulk disease seem most likely protocols with categorization of CNS disease into
Leuk Lymphoma Downloaded from informahealthcare.com by Michigan University on 11/08/14

[13,27,28]. Prone positioning after intrathecal che- CNS1, CNS2, and CNS3 [13,18,19]. Significant
motherapy could be helpful [18,19]. improvement in the rate of CNS relapse after January
Incidence of high-risk disease in patients with CNS 2001 suggests that the CNS-directed therapy used
involvement was similar in Groups A, B, and C and prior to 2001 was probably inadequate although
was only marginally higher than the 60–65% in- CNS irradiation was used in all the patients. There is
cidence of high-risk disease in India [17,29]. How- a clear need to improve systemic control and use
ever, these patients with CNS involvement had a intrathecal therapy more effectively.
significantly higher mean PLC compared to other Contemporary protocols have demonstrated the
patients with ALL which implicates an added risk. In efficacy and advantages of non-radiotherapy techni-
a recent study, Lazarus et al., in a study in adults, ques for CNS-directed therapy. However, till re-
For personal use only.

reported that a T-cell phenotype, high PLC, and the cently, in resource poor nations, in view of a
presence of a mediastinal mass were associated with multiplicity of factors enumerated above, CNS
CNS leukemia at diagnosis [30]. irradiation was being advocated as a cheap, easily
The outcome was dismal in patients with CNS available, and effective form of therapy. Our experi-
involvement despite low incidence of CNS disease ence depicts the difficulties faced in managing
and TLP and was significantly inferior in comparison patients on an intensive chemotherapy protocol and
to other patients with ALL. It is likely that more in designing remedial measures. The connotations of
patients with CNS disease at presentation belonged sole use of CNS-directed chemotherapy, although
to CNS3 status which could have contributed to the effective and advantageous; in an Indian setting
unfavorable outcome. Larazius et al. have recorded remain to be elucidated. Improvements in infra-
poor survival in adult patients with CNS disease structural and cancer treatment facilities could
despite adequate therapy [30]. A study from South optimize its use. However, the importance of cranial
India observed that CNS disease was independently irradiation cannot be underestimated in a resource-
associated with inferior survival outcome [31]. In our limited setting, more so, since, prior to 2000, very
cohort, CNS involvement was significantly asso- scanty outcome data from India were available
ciated with inferior outcome in univariate log-rank [13,33,34]. Hence, although CNS irradiation must
analysis but in multivariate Cox regression analysis have definitely contributed to the improvement in
only PLC, SDI, and CNS irradiation were significant cure rates of ALL in India as observed in our cohort,
predictors. This implies that with appropriate ther- its effectiveness is offset by substantial rates of
apy, added risk due to CNS involvement could be secondary neoplasms, endocrine disorders, growth
modified and that in our cohort other parameters impairment, and neurocognitive dysfunction pre-
were more significant prognostic determinants. Our cluding its routine use [35,36]. With the use of
study suggests that, as such, presence of any CNS modified Medical research Council United Kingdom
disease/involvement at diagnosis should itself 2003 protocol, our unit has currently adopted the
be considered as an indicator of ‘high-risk’ in policy of prophylactic cranial irradiation only to
addition to the routinely used parameters atleast in patients with increased risk of CNS relapse. Further-
resource-limited settings. Furthermore, there is need more, a recent study from St. Jude Children
to assess the cytogenetic and molecular data in this Research Hospital proposes to completely omit
subgroup to better define their risk-categorization CNS irradiation in all childhood patients with ALL
[1,2,32]. with use of apt systemic therapy without increase in
Treatment default, loss to follow-up, and a higher incidence of CNS relapse or decrement in survival
incidence of relapse and death, than in patients outcome [37].
Central nervous system involvement in childhood leukemia 267

In conclusion, CNS involvement at presentation 14. Gao YJ, Lu FJ, Wang HS. Treating childhood acute
should be considered as a high-risk indicator. Appro- lymphoblastic leukemia in a developing country 1998–2003:
the experience of a single children’s hospital in China.
priate methods and precautions to minimize the risk J Pediatr Hematol Oncol 2006;28:798–802.
of TLP and optimize intrathecal therapy should be 15. Howard SC, Pedrosa M, Lins M, et al. Establishment of a
advocated. Identifications of risk-factors in Indian pediatric oncology program and outcomes of childhood acute
patients with ALL, appropriate risk-stratification, use lymphoblastic leukemia in a resource-poor area. JAMA
2004;291:2471–2475.
of risk-adapted therapy protocols, use of cranial
16. Dabbous IA, Dbouk H, Sibai AM, Bahlawan L. Childhood
irradiation only in high-risk patients, and assessment acute lymphoblastic leukemia managed in tertiary care
of response to therapy along with adequate suppor- center in a developing country. Med Pediatr Oncol 2003;41:
tive facilities should definitely improve the overall 83–84.
survival in ALL and outcome of patients with CNS 17. Kulkarni KP, Marwaha RK, Trehan A, Bansal D. Survival
disease at initial presentation. outcome in childhood ALL: experience from a tertiary care
centre in North India. Pediatr Blood Cancer 2009;53:
168–173.
Declaration of interest: The authors report no 18. Pui CH, Howard SC. Current management and challenges of
Leuk Lymphoma Downloaded from informahealthcare.com by Michigan University on 11/08/14

conflicts of interest. The authors alone are respon- malignant disease in the CNS in paediatric leukaemia. Lancet
sible for the content and writing of the paper. Oncol 2008;9:257–268.
19. Pui CH. Central nervous system disease in acute lympho-
blastic leukemia: prophylaxis and treatment. Hematol Am Soc
Hematol Educ Program 2006:142–146.
References 20. Lee AC, Wong LG. Facial palsy, corticosteroids, and acute
1. Pui CH, Evans WE. Treatment of acute lymphoblastic leukemia. Pediatr Neurol 2007;36:137.
leukemia. N Engl J Med 2006;354:166–178. 21. Juhn YJ, Inoue S. Facial nerve palsy as an early manifestation
2. Vrooman LM, Silverman LB. Childhood acute lymphoblastic of relapse in T-cell acute lymphoblastic leukemia. Ear Nose
leukemia: update on prognostic factors. Curr Opin Pediatr Throat J 1996;75:157–160.
2009;21:1–8. 22. Antony R, Roebuck D, Hann IM. Unusual presentations of
3. Sancho JM, Morgades M, Arranz R, et al. Practice of central acute lymphoid malignancy in children. J R Soc Med 2004;97:
For personal use only.

nervous system prophylaxis and treatment in acute leukemias 125–127.


in Spain. Prospective registry study. Med Clin (Barc) 2008; 23. Shenoy A, Khajuria R, Malik N, Singhi S. Acute paraplegia as
131:401–405. a presenting symptom of acute leukemia. Indian Pediatr
4. Pui CH. Toward optimal central nervous system-directed 1988;25:569–571.
treatment in childhood acute lymphoblastic leukemia. J Clin 24. Iqbal Y, Al-Sudairy R, Abdullah MF, Rafaie T, Al-Omari A.
Oncol 2003;21:179–181. Spinal cord compression as an initial symptom in childhood
5. Advani S, Pai S, Venzon D, et al. Acute lymphoblastic acute lymphoblastic leukemia: rapid decompression with high
leukaemia in India: An analysis of prognostic factors using a dose dexamethasone and chemotherapy alone. Ann Saudi
single treatment regimen. Ann Oncol 1999;10:167–176. Med 2003;23:179–180.
6. Luo XQ, Ke ZY, Huang LB, et al. High-risk childhood acute 25. Harada T, Namiki S, Kawabata I. Acute profound sensor-
lymphoblastic leukemia in China: factors influencing the treat- ineural hearing loss as the initial manifestation of acute
ment and outcome. Pediatr Blood Cancer 2009;52:191–195. leukemia – report of a case. Auris Nasus Larynx 2000;27:
7. Chessells JM, Bailey CC, Richards S. MRC UKALL X. The 359–362.
UK protocol for childhood ALL: 1985–1990. Leukemia 26. Veling MC, Windmill I, Bumpous JM. Sudden hearing loss as
1992;6:157–161. a presenting manifestation of leukemia. Otolaryngol Head
8. Chessells JM, Bailey C, Richards SM. Intensification of Neck Surg 1999;120:954–956.
treatment and survival in all children with lymphoblastic 27. Gajjar A, Harrison PL, Sandlund JT, et al. Traumatic
leukaemia: results of UK Medical Research Council trial lumbar puncture at diagnosis adversely affects outcome in
UKALL X. Medical Research Council Working Party on childhood acute lymphoblastic leukemia. Blood 2000;96:
Childhood Leukaemia. Lancet 1995;345:653. 3381–3384.
9. Chessels JM, Harrison G, Richards SM, et al. Failure of a new 28. Howard SC, Gajjar AJ, Cheng C, et al. Risk factors
protocol to improve treatment results in paediatric lympho- for traumatic and bloody lumbar puncture in children
blastic leukaemia: lessons from the UK Medical Research with acute lymphoblastic leukemia. JAMA 2002;288:
Council trials UKALL X and UKALL XI. Br J Haematol 2001–2007.
2002;118:445–455. 29. Magrath I, Shanta V, Advani S, et al. Treatment of acute
10. Klabfleisch JD, Prentice RL. The statistical analysis of failure lymphoblastic leukaemia in countries with limited resources;
of time data. New York, NY: Wiley; 2002. lessons from use of a single protocol in India over a twenty
11. Grey RJ. A class of K sample tests for comparing the cumula- year period. Eur J Cancer 2005;41:1570–1583.
tive incidence of competing risk. Ann Stat 1988;16: 30. Lazarus HM, Richards SM, Chopra R, et al. Central
1141–1154. nervous system involvement in adult acute lymphoblastic
12. Gaynon PS, Qu RP, Chappell RJ, et al. Survival after relapse leukemia at diagnosis: results from the international ALL
in childhood acute lymphoblastic leukemia: impact of site trial MRC UKALL-XII/ECOG E2993. Blood 2006;108:
and time to first relapse – the Children’s Cancer Group 465–472.
Experience. Cancer 1998;82:1387–1395. 31. Bejel A, George B, Mathews V, et al. Treatment of children
13. Laningham FH, Kun LE, Reddick WE, Ogg RJ, Morris BE, with acute lymphoblastic leukemia in India using a BFM
Pui CH. Childhood central nervous system leukemia: protocol. Pediatr Blood Cancer 2008;51:621–625.
historical perspectives, current therapy, and acute neurological 32. Cario G, Izraeli S, Teichert A, et al. High interleukin-15
sequelae. Neuroradiology2007;49:873–888. expression characterizes childhood acute lymphoblastic
268 R. K. Marwaha et al.

leukemia with involvement of the CNS. J Clin Oncol 2007;25: 35. Pui CH, Cheng C, Leung W, et al. Extended follow-up of
4813–4820. long-term survivors of childhood acute lymphoblastic leuke-
33. Advani SH, Rao DN, Gopal R, et al. Acute lymphoblastic mia. N Engl J Med 2003;349:640–649.
leukemia: end-result analysis of treatment and prognostic 36. Hijiya N, Hudson MM, Lensing S, et al. Cumulative incidence
factors in Indian patients. Am J Hematol 1983;15:35–43. of secondary neoplasms as a first event after childhood acute
34. Advani SH, Iyer RS, Pai SK, et al. Four-agent induction/ ymphoblastic leukemia. JAMA 2007;297:1207–1215.
consolidation therapy for childhood acute lymphoblastic 37. Pui CH, Campana D, Pei D, et al. Treating Childhood Acute
leukemia: an Indian experience. Am J Hematol 1992:39; Lymphoblastic leukemia without cranial irradiation. N Engl J
242–248. Med 2009;360:2730–2741.
Leuk Lymphoma Downloaded from informahealthcare.com by Michigan University on 11/08/14
For personal use only.

Vous aimerez peut-être aussi