Recaida Lla BFM 2002
Recaida Lla BFM 2002
Recaida Lla BFM 2002
Version: 25/06/2003
This protocol was designed by the members of the ALL-REZ BFM study committee (principal
investigator: Prof. Dr. med. Dr. h.c. G. Henze, Charité Berlin). The content of this protocol is
confidential and must not be disseminated in either oral or written form without the permission of the
principal investigator.
The protocol contains experimental elements that should be used in participating centers under
controlled conditions only. These are: Protocol II-IDA (arm A), indication of stem cell
transplantation according to MRD results after F2 in S2 patients.
ALL-REZ BFM 2002 III Protocol version: 25.06.2003
Main investigators
ALL-REZ Studienzentrale
Charité – Universitätsmedizin Berlin, CVK
Augustenburger Platz 1, D – 13353 Berlin
Germany
Tel: + 49(0) 30/ 450 566 354; Fax: + 49(0) 30/450 566 901; Email: [email protected]
2
Klinikum im Friedrichshain
Klinik für Strahlentherapie/Radioonkologie
Standort Moabit-Turmstraße
Turmstr. 21, D- 10559 Berlin
Germany
Telefon: + 49(0) 30/ 3976 – 3611; Telefax: + 49(0) 30/ 3976 – 3609
3
Name Email
Reference laboratories
Test Phone/Fax Address
Contact
Cytomorphology, ALL-REZ Studienzentrale,
cytochemistry, treatment Charité CVK
response Klinik für Pädiatrie m.S.
Tel: +49 (0) 30 450 566 354 Onkologie/Hämatologie
Dr. A.v. Stackelberg Fax: +49 (0) 30 450 566 901 Augustenburger Platz 2,
D – 13353 Berlin
Laboratory Tel: +49 (0) 30 450 566 050 Germany
Fax: +49 (0) 30 450 566 903
Molecular Genetics, ALL-REZ Studienzentrale,
Cytogenetics Charité CVK
Dr. Dr. K. Seeger Tel: +49 (0) 30 450 566 088 Klinik für Pädiatrie m.S.
Fax:+49 (0) 30 450 566 946 Onkologie/Hämatologie
Augustenburger Platz 2,
D – 13353 Berlin, Germany
MRD - Testing ALL-REZ Studienzentrale,
C. Eckert Tel: +49 (0) 30 450 566 088 Charité CVK
Fax: +49 (0) 30 450 566 946 Klinik für Pädiatrie m.S.
Onkologie/Hämatologie
Augustenburger Platz 2,
D – 13353 Berlin, Germany
Immunology Immunol. Zellmarkerlabor,
Prof. Dr. WD. Ludwig Tel: +49 (0) 30 9417 1362 Charité, Campus Berlin-Buch
Fax: +49 (0) 30 9417 1308 Robert Rössle Klinik – MDC
Lindenberger Weg 80,
D – 13122 Berlin – Buch,
Germany
MRD Testing after bone
marrow transplantation MRD-/Chimärismuslabor,
Klinik für Kinderheilkunde und
PD Dr. P. Bader Tel: +49(0) 7071 29-83809 Jugendmedizin
Fax: +49(0) 7071 29-5365 Hoppe-Seyler-Straße 1
D – 72076 Tübingen, Germany
ALL-REZ BFM 2002 VI Protocol version: 25.06.2003
Signatures
Abstract
The protocol ALL-REZ BFM 2002 aims at the optimization of treatment for children with
relapsed acute lymphoblastic leukemia. It is designed as a prospective controlled randomized
multi-center study. The participating centers include all hospitals treating children with relapsed
acute lymphoblastic leukemia in Germany and Austria as well as some centers in Switzerland.
The study is based on the results of five consecutive trials performed by the ALL-REZ BFM study
group since 1983. Thus the study meets the criteria of evidence-based therapy, which has been
developed over nearly 20 years. Multi-agent chemotherapy in short intensive courses, which are
separated by treatment-free intervals, has proved to be a successful form of induction and
consolidation therapy. It is followed by preventative (or therapeutic) cranial irradiation and
continuation therapy. A number of risk factors, particularly the time of relapse, site of relapse, and
the ALL immunophenotype, allow the stratification of patients into a group that has an acceptable
prognosis after treatment with chemotherapy alone and a second group that has a high risk of
subsequent recurrence following the achievement of a second remission. The latter group requires
further intensification of consolidation therapy by allogenic stem cell transplantation (SCT). To
date, the indication for SCT has remained unclear for a large and heterogeneous group of patients
with an intermediate prognosis. During the precursor study ALL-REZ BFM 96, however, the
amount of minimal residual disease (MRD) determined quantitatively with clonal molecular
markers after the second induction therapy element was shown to be a highly significant predictor
of relapse-free survival.
The primary objective of study ALL-REZ BFM 2002 is the randomized comparison of a lower
dosed and less intensive, but continuous consolidation therapy with conventional therapy
administered in treatment blocks. Outcome measures are the reduction of MRD, event-free and
overall survival, and the toxicity associated with each treatment strategy.
The secondary objectives include an improvement of the prognosis in the intermediate risk group
using the stratification in treatment arms with and without allogenic SCT based on the MRD result
after the second treatment element of induction therapy. An additional aim is to improve the
remission induction rate in all groups by increasing the treatment intensity during induction. This
is achieved by shortening the intervals between treatment blocks in keeping with the principles of
guiding therapy as defined in the protocol. A series of biological companion studies aims to
advance our understanding of the disorder and to establish novel prognostic factors that will allow
a risk-adapted therapy.
The accrual of the study is planned for 5 years during which approximately 450 patients will be
enrolled.
Time line
Begin of the ALL-REZ BFM Pilot 02 01.01.2002
End of the pilot study 31.07.2003
Begin of the main study ALL-REZ BFM 2002 01.08.2003
End of patient accrual 31.07.2007
End of the treatment phase 31.07.2008
End of the study 31.07.2012
ALL-REZ BFM 2002 1 Protocol version: 25.06.2003
Table of Contents
1 GENERAL REMARKS ...................................................................................................... 6
1.1 Abbreviations ................................................................................................................ 7
2 INTRODUCTION ............................................................................................................... 8
2.1 Design and results of previous studies .......................................................................... 9
2.1.1 Results of studies ALL-REZ BFM 83-90................................................................................... 9
2.1.2 Results of randomizations ........................................................................................................ 11
2.1.3 Prognostic Factors .................................................................................................................... 11
2.2 Study ALL-REZ BFM 95/96 ...................................................................................... 14
2.2.1 Definition of risk groups (S groups)......................................................................................... 14
2.2.2 Study design ............................................................................................................................. 14
2.2.3 Results for the overall study population and for subgroups ..................................................... 15
2.2.4 The randomized use of filgrastim (G-CSF) .............................................................................. 16
2.2.5 Pilot Studies P99 and P01......................................................................................................... 17
2.3 Extramedullary relapse................................................................................................ 18
2.3.1 Isolated CNS relapse ................................................................................................................ 18
2.3.2 Isolated Testicular relapse ........................................................................................................ 19
2.4 Stem Cell Transplantation........................................................................................... 19
2.5 Results of other studies ............................................................................................... 22
3 AIMS AND RATIONALE OF THE STUDY .................................................................. 23
3.1 Conclusions from previous studies.............................................................................. 23
3.1.1 Chemotherapy .......................................................................................................................... 23
3.1.2 Principal treatment guidelines .................................................................................................. 23
3.1.3 Strategic groups and indications for transplantation................................................................. 23
3.1.4 Minimal Residual Disease........................................................................................................ 23
3.2 Aims of study ALL-REZ BFM 2002 .......................................................................... 23
3.3 Comparison of treatment blocks with continuous chemotherapy ............................... 24
3.3.1 Protocol II – IDA...................................................................................................................... 25
3.3.2 R Blocks ................................................................................................................................... 25
3.3.3 Comparison of cumulative drug doses ..................................................................................... 26
3.3.4 Toxicity .................................................................................................................................... 26
3.3.5 Randomization.......................................................................................................................... 26
3.3.6 Monitoring................................................................................................................................ 26
3.4 Stratification according to MRD after the second treatment element ......................... 27
3.5 Increased treatment intensity during initial therapy as a result of shorter intervals
between the initial treatment blocks ............................................................................ 28
3.6 Improvement of the remission induction rate in strategic group S4 ........................... 28
3.7 Standardization and monitoring of treatment of L-asparaginase ............................... 29
3.8 Additional aims and modifications.............................................................................. 29
3.8.1 Simplification of continuation therapy – the use of 6-mercaptopurine and oral methotrexate . 29
3.8.2 Autologous SCT for an isolated CNS relapse with unfavorable prognosis .............................. 29
3.8.3 Experimental treatment approaches for high risk groups ......................................................... 30
3.8.3.1 STI571 for BCR-ABL-positive patients................................................................................... 30
3.8.3.2 Re-intensification for S3/4 patients with a positive MRD result prior to SCT ......................... 31
3.9 Scientific companion studies....................................................................................... 31
3.9.1 Prognostic relevance of MRD at additional time points........................................................... 31
3.9.2 Prognostic relevance of MRD prior to SCT ............................................................................. 32
3.9.3 Monitoring of L-asparaginase activity...................................................................................... 32
3.10 Summary of rationale – risk-benefit analysis.............................................................. 32
4 STUDY DESIGN .............................................................................................................. 36
4.1 Features of the study ................................................................................................... 36
4.2 Study Organization...................................................................................................... 36
4.3 Inclusion and exclusion criteria................................................................................... 36
ALL-REZ BFM 2002 2 Protocol version: 25.06.2003
1 GENERAL REMARKS
The concept of this treatment protocol was approved by the members of the study committee in
February 2001 and presented to the plenary session of the BFM study group in September 2001. The
pilot phase with the objective to prove the feasibility of the protocol therapy ran from January 2002 to
July 2003. The main study began on 1st August 2003. It is anticipated to conclude on 30th July 2008.
The specific type and combination of therapeutic instructions described in this protocol do not
represent the recommendation of a universally accepted form of treatment. Rather, these instructions
represent guidelines that are part of a study aimed at the optimization of therapy. For ethical and legal
reasons, therefore, it is not permissible to treat patients according to this protocol in centers that do not
participate in the study and thus do not meet the requirements of documentation and ongoing feedback
with the study center. Patients and/or their legal guardians have to be informed accordingly.
The highest degree of diligence was used during the preparation of this protocol. Nevertheless, errors
cannot be completely ruled out. Therefore, it is important to point out that the treating physician
ultimately is responsible for the treatment. The principal investigator does not assume any legal
responsibility for consequences that may result from the implementation of recommendations made in
this protocol. Registered trademarks were identified. However, it cannot be concluded from the
absence of such identification that a registered trademark does not apply.
The aids for data documentation, which are provided in this protocol such as the documentation of
treatment blocks and order sets for infusions, are merely recommendations. It is, for example,
impossible to summarize the entire information that is pertinent to a particular treatment block in a
legible form on a single page. Each participating center, therefore, may design its own aids for the
documentation as it considers them appropriate. The study center accepts any form of documentation
that shows the same information in unequivocal fashion and allows a review of the diagnosis,
treatment and course.
The study center offers an extensive spectrum of additional services. They include the central review
of bone marrow and peripheral blood smears, CSF cytospin and tumor touch preparations. All patient
data collected during the study will be carefully documented and reviewed at regular intervals. All
centers participating in this study have access to consultation for any diagnostic and therapeutic issue.
Typically, a response will be provided within 24 hours. In the case of severe adverse effects of therapy
the study center will contact the treating center immediately.
The services provided by the study center also include a consultation regarding the indication for stem
cell transplantation (SCT) in individual patients. Due to the ongoing feedback with other transplant
centers the recommendations reflect the most up-to-date practice of stem cell transplantation. Upon
request the study center is also willing to assist with the referral of patients to transplant centers. Bone
marrow transplant beds are also available at the study center.
ALL-REZ BFM 2002 7 Protocol version: 25.06.2003
1.1 Abbreviations
6-MP 6-mercaptopurine
6-TG 6-thioguanine
ALL acute lymphoblastic leukemia
ARA cytosine arabinoside
Asp asparaginase
BFM Berlin-Frankfurt-Münster
CCG Children's Cancer Group
CML chronic myeloid leukemia
CPM cyclophosphamide
C(C)R complete (continuous) remission
DNR daunorubicin
Dexa dexamethasone
EFS event-free survival
HD high-dose
G-CSF granulocyte-colony stimulating factor
HLA human leukocyte antigen
IDA idarubicin
IFO ifosfamide
i.t. intrathecal
i.v. intravenous
MFD matched family donor
MRC Medical Research Council
MRD minimal residual disease
MSD matched sibling donor
MTX methotrexate
MUD matched unrelated donor
p probability
PBC peripheral blood cell count
PEG polyethylene glycol
POG Pediatric Oncology Group
PPG poor prognosis group
Pred prednisone
REZ relapse
SCT, SZT stem cell transplantation
SRV survival
U unit(s)
VCR vincristine
VDS vindesine
CNS central nervous system
ALL-REZ BFM 2002 8 Protocol version: 25.06.2003
2 INTRODUCTION
The Berlin-Frankfurt-Münster (BFM) study group has evaluated approaches to the treatment of
children with a relapse (REZ) of acute lymphoblastic leukemia (ALL) in multi-center studies since
1983. The study group comprises more than 100 centers in Germany, Austria and Switzerland. It can
be assumed that almost all children with relapsed ALL in Germany and Austria are enrolled.
Compared to the primary disease, the probability of cure is significantly lower for children with a
relapse. The overall probability of survival after 5 years is approximately 35%. The primary objective
of the ALL-REZ BFM studies, therefore, is to improve the chance of cure for these children. Proven
therapeutic interventions include chemotherapy, radiation therapy and stem cell transplantation (SCT).
In addition, companion research studies aim to advance our insight into this disorder. The results of
the ALL-REZ BFM relapse studies have to be viewed together with the results of the treatment studies
for primary ALL. Since the primary ALL-BFM studies have achieved a continual improvement of
results and a decreased rate of recurrence, we anticipate that a decreasing number of patients will be
available for the relapse studies and that the cases of leukemia will be more resistant as a result of the
more intensive risk-adapted primary therapy (Schrappe et al., 2000). The number of patients treated on
the pilot and main studies over the years as well as the number of patients with protocol violations is
shown in fig. 1.
140
120
number of violation
100
80
60
treatment
40
treatment violation
20
pilot study
0 main study
83 85 87 89 91 93 95 97 99
year of relapse
ALL-REZ BFM 2002 9 Protocol version: 25.06.2003
REZ A C E R1 R2 R1 R2 R1 R2 R1 R2 D 24
BFM
B C R1 R2 R1 R2 R1 R2 R1 R2 D 24
83
C C R1 R2 R1 R2 D12
group week 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29
A R C F R1 R2 R1 R2 R1 R2 R1 R2 D24
85/
87 B (R) C R1 R2 R1 R2 R1 R2 R1 R2 D 24
C (R) C R1 R2 R1 R2 R1 R2 D12
group week 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29
90 A/B R C R1 R2 R3 R1 R2 R3 R1 R2 R3 D24
C R C R1 R2 R3 R1 R2 R3 D12
Fig. 3: Event-free survival of protocol patients, studies ALL-REZ BFM83-95, PPG excluded; status 09/01
1,0
,8
pEFS
,6
,4
,2
0,0
0 5 10 15
Years
________
83: n= 65; cens.= 20; pEFS= .30 .06
__ __ __
85: n= 101; cens.= 36; pEFS= .35 .05
_____
87: n= 151; cens.= 57; pEFS= .38 .04
__ _ __ _
90: n= 374; cens.= 136; pEFS= .36 .03
__ _ _ __
95/96 n= 408; cens.= 253; pEFS= .47 .03
p = 0.017
ALL-REZ BFM 2002 11 Protocol version: 25.06.2003
1,0 1,0
,8 pEFS ,8
,6 ,6
pEFS
,4 ,4
,2 ,2
0,0 0,0
0 2 4 6 8 10 0 2 4 6 8 10
years years
____
with R3: n= 374; cens.= 187; pEFS= .38 .03 5 g/m²: n= 128; cens.= 65; pEFS= .40 .05
______
without R3: n= 317; cens.= 133; pEFS= .33 .03 1 g/m²: n= 141; cens.= 71; pEFS= .38 .05
p= 0.47 p= 0.96
Fig. 6: Event-free survival dependent on time of Fig. 7: EFS dependent on site of relapse, studies 83-96,
relapse, studies 83-96, SCT censored; status 09/01 SCT censored (isol. EM, isolated extramedullary; comb.,
combined bone marrow; isol. BM, isolated bone marrow)
1,0 1,0
,8 ,8
,6 ,6
pEFS
pEFS
,4 ,4
,2 ,2
0,0 0,0
0 2 4 6 8 10 0 2 4 6 8 10
years years
__ __
late: n = 669; cens. = 394; pEFS = .45 .02 isol.EM: n = 233; cens.= 139; pEFS= .52 .04
____
early: n = 421; cens. = 206; pEFS = .26 .03 comb.: n = 300; cens.= 164; pEFS= .44 .03
______
very early: n = 217; cens. = 67; pEFS = .15 .03 isol.BM: n = 774; cens.= 364; pEFS= .23 .02
p < 0.001 p < 0.001
Fig. 8: Event-free survival dependent on immunopheno- Fig. 9: Event-free survival dependent on the interval
type, studies 83-96, SCT censored; status 09/01 between the first two treatment elements; ALL-REZ BFM
90, all documented patients; status 09/01
1,0 1,0
,8 ,8
,6 ,6
pEFS
pEFS
,4 ,4
,2 ,2
0,0 0,0
0 2 4 6 8 10 0 2 4 6 8 10
years years
______
non-T: n = 669; cens.= 394; pEFS= .45 .02 <21 days: n= 94; cens.= 45; pEFS=.48 .05
__ __ _
T: n = 217; cens.= 67; pEFS= .34 .02 21 -25 days: n=129; cens.= 48; pEFS=.37 .04
____
>25 days: n= 78; cens.= 22; pEFS=.27 .05
p < 0.001 p < 0.01
The prognostic impact of treatment intensity during the phase of therapy in blocks was first evaluated
in study ALL-REZ BFM 90. Patients with shorter intervals between the initial treatment blocks had a
more favorable event-free survival (fig.9) (Hartmann et al., 1995).
ALL-REZ BFM 2002 13 Protocol version: 25.06.2003
Fig. 10: Event-free survival dependent on the expression Fig. 11: Event-free survival dependent on expression of
of BCR-ABL, studies 83-96, SCT censored; status 09/01 TEL-AML1, studies 83-96, SCT censored; Stand 09/01
1,0 1,0
,8 ,8
pEFS
pEFS
,6 ,6
,4 ,4
,2 ,2
0,0 0,0
0 2 4 6 8 10 0 2 4 6 8 10
Years Years
______
BCR-ABL-: n = 546; zens = 318; pEFS= .37 .03 TEL-AML1+: n = 70; zens.= 57; pEFS=.68 .08
____
no data: n = 715; zens.= 331; pEFS= .34 .02 no data: n=903; zens.= 415; pEFS=.33 .02
__ __
BCR-ABL+: n = 46; zens.= 18; pEFS= .16 .09 TEL-AML1-: n=306; zens.=136; pEFS=.29 .04
p < 0.001 p < 0.001
ALL-REZ BFM 2002 14 Protocol version: 25.06.2003
Fig. 12: Event-free survival dependent on the strategic Fig. 13: Event-free survival dependent on the strategic
group,studies95/96, SCT censored; status 09/01 group, study 95/96, and SCT; status 09/01
1,0 1,0
,8 ,8
,6 ,6
pEFS
pEFS
,4 ,4
,2 ,2
0,0 0,0
0 2 4 6 8 10 0 1 2 3 4 5 6
years years
______
S1: n = 51; cens. = 40; pEFS = .75 .06 n = 29; cens. = 24; pEFS = .79 .09
__ __
S2: n = 577; cens. = 277; pEFS = .38 .02 n = 325; cens. = 206; pEFS = .48 .04
____
S3: n = 153; cens. = 46; pEFS = .02 .02 n = 69; cens. = 26; pEFS = .25 .06
__ _ __
S4: n = 252; cens. = 60; pEFS = .04 .02 n = 106; cens. = 20; pEFS = .19 .04
p < 0.001 p < 0.001
depending on the availability of a suitable donor and on the subgroup within S2 (for definitions see
tab. 3, p.21).
Similarly, patients in group S3 received induction therapy with F blocks followed by a series of
alternating R blocks. After a complete remission was achieved mandatory SCT followed.
Additionally, the use of G-CSF during the intervals between the first four treatment blocks was
randomized for patients in group S2 and S3. The aim was to evaluate if G-CSF resulted in the
intensification of induction therapy and, as a consequence, the improvement of remission induction
rate and survival.
Patients in group S4 were treated with a novel induction block I followed by S blocks. The design of
these blocks focused on a somewhat reduced treatment intensity and improved therapeutic control.
The aim was to reduce the comparatively high treatment-related mortality and induction death rate
observed in the previous studies. These blocks used idarubicin, etoposide and thiotepa, cytotoxic
agents that had shown activity in vitro against highly drug-resistant leukemic cells. After the
achievement of a complete remission prompt SCT was recommended since an early subsequent
relapse had to be anticipated.
S1 C F1 F2 R1 R2 R1 R2 R1 R2 D 12
S2 R C F1 G? F2 G? R1 G? R2 R1 R2 R1 R2 R1 R2 D 24V
C, cytoreductive pre-phase; D12, continuation therapy 12 months; D24V, continuation therapy 24 months
with VP16 reinduction pulses; R, randomization; , radiation therapy; R1 / R2 / F1 / F2, chemotherapy
blocks
2.2.3 Results for the overall study population and for subgroups
With a study duration of 6 years and 2 months and a median follow up of 2.7 years (5.5 years for study
95, 2.0 years for study 96) the .47±.03 pEFS of study 95/96 is a significant improvement over the
preceding studies (fig.3, p.10). When the criteria underlying the strategic groups S1 to S4 are
retrospectively applied to the previous studies, it is evident that the satisfactory result of group S1
could be reproduced. In group S3 and S4 the event-free survival plateaued at 25% and 19%,
respectively. This result, however, does not represent a significant improvement compared to previous
studies when transplantation is taken into account. Moreover, the trend of the remission induction rate
in group S4 is significantly worse compared with studies 85 and 87 (induction therapy with protocol
F1 and F2). The rate of induction deaths could not be reduced (table 1, p.16). Patients in group S3 and
S4 who were transplanted in CR had an event-free survival of 40%. All patients who were not
transplanted early despite achieving a CR had a subsequent event (fig.15, p.16).
The event-free survival of group S2 was significantly better than in previous studies (fig.16, p.16).
ALL-REZ BFM 2002 16 Protocol version: 25.06.2003
Tab. 1: Results of induction therapy in group S4 dependent on treatment protocol; PPG included
Fig. 15: Event-free survival of patients in group S3 and S4 Fig 16: EFS of patients in group S2; studies 95/96 vs. 83-
(after achieving a CR) with vs. Without SCT; studies 90; PPG excluded; status 09/01
95/96; status 09/01
1,0 1,0
,8 ,8
,6 ,6
pEFS
pEFS
,4 ,4
,2 ,2
0,0 0,0
0 1 2 3 4 5 6 0 2 4 6 8 10
years years
__ __
SZT: n = 71; cens. = 31; pEFS = .41 .06 95/96: n = 313; cens. = 204; pEFS = .49 .04
______
no SZT: n = 31; cens. = 4; pEFS = .00 .00 83-90: n = 514; cens. = 202; pEFS = .39 .02
p = 0.013
Fig. 17: G-CSF randomization: analysis by treatment Fig. 18: G-CSF randomization: intention-to-treat analy-
received ; status 09/01 sis; status 09/01
1,0 1,0
,8 ,8
,6 ,6
pEFS
pEFS
,4 ,4
,2 ,2
0,0 0,0
0 1 2 3 4 5 0 1 2 3 4 5
years
years
__ __
G-CSF-: n = 100; cens. = 60; pEFS = .44 .08 n = 131; cens. = 79; pEFS = .45 .07
______
G-CSF+: n = 108; cens. = 58; pEFS = 40 .08 n = 118; cens. = 63; pEFS = .41 .07
p = 0.30
Fig. 19: Event-free survival dependent on the interval Fig. 20: Intervals between the first 4 blocks of therapy
between the first two treatment elements F1/F2; ALL-REZ treatmentarms with and without G-CSF; ALL-REZ BFM
BFM 95/96, strategie group S2 95/96, strategie group S2/S3 with documentation of
intervals
,8
25
,6
pEFS
D
,4 a 20
y
s
,2
15 G-CSF
0,0
without
0 1 2 3 4 5
10 with
years N = 38 50 38 48 37 42
Study P01 piloted arm A of the current study, ALL-REZ BFM 2002, in group S4. Induction therapy
with blocks F1 and F2 was followed by consolidation therapy using protocol II-IDA and bone marrow
transplantation if a CR was achieved. 73% of patients achieved a CR using this strategy (table 2, p.18).
Fig 21: EFS of children with an isolated CNS relapse Fig 22: EFS of children with an isolated CNS relapse
(early or very early, S2) dependent on sex; ALL-REZ BFM (early or very early, S2) dependent on age at initial
83 - 96; status 09/01 diagnosis of ALL-REZ BFM 83 - 96; status 09/01
1,0 1,0
,8 ,8
pEFS
,6
pEFS
,6
,4 ,4
,2 ,2
0,0 0,0
0 2 4 6 8 10 0 2 4 6 8 10
Years Years
__ __
Girls: n = 40; cens. = 29; pEFS = .66 .08 < 6 Years: n = 73; cens. = 44; pEFS = .54 .06
______
Boys: n = 74; cens. = 26; pEFS = 29 .06 6 Years: n = 41; cens. = 11; pEFS = .17 .07
p < 0.001 p < 0.001
0 haploidentical SCT
83 85 87 90 95 96
ALL-REZ BFM
Since the early 1990s SCT from an HLA-identical unrelated donor (matched unrelated donor, MUD)
has become increasingly available. Presently a suitable donor can be identified for approximately 75%
of all patients within three months. Stem cell transplantation from an unrelated donor has a
significantly higher treatment-related mortality and morbidity due to T-cell depletion and graft versus
host disease than SCT from a related donor. Therefore, it was usually performed only in patients with
a particularly unfavorable prognosis (fig.25, p.21).
In some regards ALL-REZ BFM 95/96 clearly defined the indication for allogeneic SCT based on the
risk group. Examples are the mandatory stem cell transplantation for group S3 and S4 and the lack of
an indication for group S1. In the large and heterogeneous group S2, however, the indication for SCT
from an unrelated donor was not clearly defined (fig.26, p.21). Time and site of relapse, initial blast
count, expression of BCR-ABL and cytologic response to treatment proved to be relevant prognostic
factors within strategic group S2 (Beyermann et al., 1997; Buehrer et al., 1996). The cryptic
translocation t(12;21) (TEL-AML1) was shown to be a parameter that correlates with favorable
prognostic factors but at this point - with a short follow-up - does not have independent prognostic
relevance (Seeger et al., 1998; Seeger et al., 2001).
Based on these risk factors the further stratification of group S2 into subgroups S2A to S2D was
introduced by the study committee and the Pediatric Working Group for Bone Marrow and Peripheral
Blood Stem Cell Transplantation (Paed-AG-KBT) as a guideline regarding the indication for SCT
(fig.24, p.21 and table 3, p.21). The following recommendations were made.
S2A chemotherapy, optional MFD-SCT
S2B MFD-SCT, chemotherapy vs. high-resolution MUD-SCT
S2C MFD-SCT, MUD-SCT, haplo-identical or autologous SCT
S2D chemotherapy, possibly autologous SCT
ALL-REZ BFM 2002 21 Protocol version: 25.06.2003
The results of SCT in patients of group S2 are shown in table 4 and fig 26.
Fig. 24: Event-free survival of S2 subgroups A-D, studies 83- Tab. 3: Definition of subgroups S2 A-D
96, SCT censored; status 09/01
1,0
Site isol. comb. isol.
BM KM extramed.
,8
Time late late early early/
very early
pEFS
,2
0,0
0 2 4 6 8 10
years
______
S2A: n = 221; cens. = 140; pEFS = .53 .04
__ __
S2B: n = 401; cens. = 218; pEFS = .36 .03
____
S2C: n = 153; cens. = 92; pEFS = .24 .06
__ _ __
S2D: n = 184; cens. = 93; pEFS = .39 .04
p < 0.001
Tab. 4: Results in group S3/S4 and S2 after SCT, studies 90-96; status 09/01
strategic group S3 / S4 S2
SCT MFD MUD autologous MFD MUD autologous
N % N % N % N % N % N %
Total 49 100 57 100 16 100 54 100 24 100 13 100
treatment-related mortality 7 14 12 21 - - 1 2 11 46 - -
second malignancy 1 2 2 4 - - - - 1 4 - -
Relapse 21 43 18 32 13 81 17 32 3 13 10 77
CCR 20 41 25 44 3 19 36 67 9 38 3 23
MFD, HLA-matched family donor; MUD, HLA- matched unrelated donor
Fig. 25: Event-free survival of groups S3/S4 after stem Fig. 26: Event-free survival of group S2 after stem cell
cell transplantation, studies 90-96; status 09/01 transplantation, studies 90-96; status 09/01
1,0 1,0
,8 ,8
,6 ,6
pEFS
pEFS
,4 ,4
,2
,2
0,0
0,0
0 2 4 6 8
0 2 4 6 8
years years
______
MFD-SZT: n = 49; cens. = 20; pEFS = .37 .08 n = 54; cens. = 36; pEFS = .63 .07
__ __
MUD-SZT: n = 57; cens. = 25; pEFS = .41 .07 n = 24; cens. = 9; pEFS = .35 .11
____
autologous: n = 16; cens. = 3; pEFS = .19 .10 n = 13; cens. = 3; pEFS = .18 .12
p = 0.19 p < 0.001
ALL-REZ BFM 2002 22 Protocol version: 25.06.2003
3.1.1 Chemotherapy
The results of the ALL-REZ BFM studies hold up favorably in an international comparison. The
principle of treatment consists of intensive multi-agent chemotherapy, which is organized in treatment
blocks during induction and consolidation therapy. This treatment phase is followed either by
conventional continuation therapy with or without VP16 reinduction pulses (for patients with a good
or intermediate prognosis after chemotherapy alone) or, alternatively, by stem cell transplantation (for
patients with an intermediate or unfavorable prognosis). This principle remains unchanged. Induction
and consolidation therapy using the blocks I and S in strategic group S4 resulted in a lower remission
induction rate compared to the historical controls of the precursor studies. The highest remission
induction rates were achieved with blocks F1 and F2 in studies ALL-REZ BFM 85 and 87. As a
consequence the current study will use a uniform induction therapy with blocks F1 and F2 for group
S4 as well.
Treatment with L-asparaginase was heterogeneous in the preceding studies. Depending on preexisting
sensitization to conventional E. coli L-asparaginase, PEG-L-asparaginase was used as a first line or
second line treatment. Studies of L- asparaginase activity in the serum were performed at doses
between 500 and 1000 units/m2. In case of allergy Erwinia L-asparaginase was used as an alternative.
Since study ALL-REZ BFM 96 did not detect a difference of event-free survival between the arms
with and without G-CSF, the current study will no longer employ G-CSF with the goal of intensifying
therapy. Instead, G-CSF will only be used with a classical supportive care indication in patients who
experience particularly frequent complications and - as a result - long delays of therapy.
preceding BFM relapse studies, as well as the targeted use of stem cell transplantation. An additional
aim is to advance our insight into this disease. The specific aims of study ALL-REZ BFM 2002 are as
follows.
To determine in a prospective randomized comparison whether treatment in blocks (R
blocks) vs. continuous chemotherapy (protocol II-IDA) during consolidation therapy is
more efficacious in maintaining a complete remission and in reducing minimal residual
disease; to compare the toxicities of both strategies. Outcomes measures are the
probability of event-free and overall survival, treatment related mortality, toxicity
(assessed according to modified WHO criteria), and the level of minimal residual disease
at specified time points.
To determine - in a comparison with historical controls - if the stratification of patients in
group S2 based on the MRD result after the second treatment element (F2) into a
subgroup with and another without allogeneic SCT from an HLA-identical unrelated
donor results in an increase of event-free survival for the entire group or for the
subgroups. Outcomes measures are the probability of event-free and overall survival and
treatment-related mortality.
To determine - in a comparison with historical controls - if the standardization of
induction therapy through a shortening of intervals between the treatment blocks (in
accordance with the guidelines for administering therapy) improves the remission
induction rate. Outcomes are event-free and overall survival as well as the length of the
intervals between blocks.
To improve the remission induction rate in strategic group S4 using a modified
induction/consolidation therapy.
To standardize the treatment with L-asparaginase using the routine monitoring of L-
asparaginase activity.
3.3.2 R Blocks
The treatment arm using R blocks is largely equivalent to the previously used standard therapy.
Compared with the design of study ALL-REZ BFM 96 the following modifications of the R blocks are
introduced.
Following induction blocks F1/2, block R2 is used first followed by R1. With the administration of
eight R blocks in group S2 the cumulative anthracycline dose remains unchanged and merely the
sequence of blocks is altered with the following rationale: the interval between diagnosis and the first
dose of antracycline is shortened; a better comparability to the antracycline-containing protocol II-IDA
is achieved.
According to the new sequence protocol II-IDA is followed by block R1 without daunomycin. Thus
the cumulative dose of anthracyclines is reduced by 35mg/m2 in this arm and the further sequence of R
blocks is synchronized in both arms.
ALL-REZ BFM 2002 26 Protocol version: 25.06.2003
3.3.4 Toxicity
In addition to anti-leukemic efficacy both strategies will be compared with regard to toxicity. In arm A
toxicity will be documented from the start of protocol II-IDA until the start of the second part (day 1-
28) and from the start of the second part (day 29) until the start of the subsequent R block. In arm B
the toxicity of the first three R blocks will be documented at the start of each subsequent R block. The
form enclosed in the appendix (p125) and the criteria recommended by the WHO will be used to
document toxicity.
3.3.5 Randomization
Enrollment in the arm containing protocol II-IDA (arm A) vs. R blocks (arm B) is determined by
randomization. The randomization is performed at the beginning of relapse therapy once the patient is
registered and written consent has been received.
3.3.6 Monitoring
In addition to the mandatory bone marrow aspirates after the first (F1) and the second treatment
element (F2) further aspirates are scheduled at the beginning of the subsequent R blocks (first R2, first
R1, second R2) and on day 15 and 28 of protocol II-IDA as well as at the start of the first R1 block
following protocol II-IDA, respectively. The anti-leukemic efficacy of both strategies will be
compared using MRD. This will provide an answer in the short term as to which strategy is more
superior to eliminate minimal residual disease.
ALL-REZ BFM 2002 27 Protocol version: 25.06.2003
Tab. 6 Indication for transplantation based on subgroups 2A-C vs. result of MRD (status 08/01)
Fig. 27: Event-free and overall survival dependent on MRD status after the second treatment
element, group S2, ALL-REZ BFM 95/96 (status 8/01)
1,0 1,0
,8 ,8
pOS
,6 ,6
pEFS
,4 ,4
,2 ,2
0,0 0,0
0 1 2 3 4 5 6 0 1 2 3 4 5 6
years years
______
MRD neg.: n= 43; cens.= 39; pEFS= .89 .06 n= 43; cens.= 41; pOS= .98 .02
____
MRD pos.: n= 29; cens.= 18; pEFS= .46 .13 n= 29; cens.= 19; pOS= .27 .20
p = 0.001 p < 0.001
induction rate in this retrospectively defined group of patients with the induction blocks F1 and F2,
this type of induction therapy will be used again (table 1, p.16). As a consequence, induction therapy
with F blocks is uniform in all strategic groups.
3.8.2 Autologous SCT for an isolated CNS relapse with unfavorable prognosis
Additional risk factors allow the prognostic stratification of the group of children with an early or very
early isolated CNS relapse. In a multi-variate analysis sex, age at first diagnosis of ALL, blast
immunophenotype and time of relapse had independent prognostic significance. Based on subgroup
analysis a stratification of the entire cohort into a prognostically favorable and a prognostically
unfavorable group can be derived (table 7, p.30). Children in the prognostically favorable group
(CNS-S, 60%) are treated with chemotherapy and radiation therapy as designed for group S2. Children
ALL-REZ BFM 2002 30 Protocol version: 25.06.2003
in the prognostically unfavorable risk group (CNS-H), however, require an intensification of therapy
(fig 28 and fig 29). Since the risk of allogeneic SCT from an unrelated donor does not appear justified
in view of the questionable graft-versus-leukemia effect in the CNS (Borgmann et al., 1995a) and
since the Italian study group AIEOP published good results for autologous SCT in children with an
isolated CNS relapse (Messina et al., 1998), a modified autologous SCT with immune modulation,
reinduction and continuation therapy is used in this group. SCT should be planned following block R2
in week 16. The study center will send a corresponding treatment recommendation to the registering
institution if the above mentioned risk factors are present.
Tab. 7: Stratification of children with an early or very early isolated CNS relapse into a high (H) and standard (S) risk
group
Fig. 28: EFS of children with an isolated CNS relapse Fig. 29: OS of children with an isolated CNS relapse (early
(early or very early, S2) after stratification into a high (H) or very early, S2) after stratification into a high (H) and
and standard (S) risk group; ALL-REZ BFM 83-96; status standard (S) risk group; ALL-REZ BFM 83-96; status 09/01
09/01
1,0 1,0
,8 ,8
pOS
,6 ,6
pEFS
,4 ,4
,2 ,2
0,0 0,0
0 2 4 6 8 10 0 2 4 6 8 10
Years Years
____
CNS-S: n = 61; cens. = 43; pEFS = .64 .07 n = 61; cens. = 45; pOS = .65 .07
__
CNS-H: n = 50; cens. = 11; pEFS = 17 .06 n = 50; cens. = 19; pOS = 31 .06
p < 0.001 p = 0.003
lymphoblastic and myeloid cell lines including the amplification of the oncogene (Schindler et
al.,2000, Weisberg & Griffin., 2000). The development of drug resistance during the treatment of
BCR-ABL-positive ALL can potentially be decreased by a combination of STI571 with other
cytotoxic agents. In vitro MTT cytotoxicity assays show a synergistic effect with most commonly used
agents including vincristine, doxorubicin, cyclophosphamide and etoposide. Only for methotrexate an
antagonistic effect could be demonstrated in most of the cell lines tested (Kano et. al, 2001). However,
the MTT assay proved not to be suitable for the evaluation of the cytotoxic efficacy of methotrexate
and this finding, therefore, requires confirmation with other methods.
Approximately four patients with a first relapse of a BCR-ABL positive ALL are registered in ALL-
REZ BFM studies each year. Since the molecular definition is already known at the time of relapse
there is an opportunity to evaluate the efficacy of STI571 in an initial therapeutic window. The
evaluation of STI571 is the objective of a separate study in accordance with the “Arzneimittelgesetz”
[drug act]. A two-week cytoreductive pre-phase with STI571 will be used instead of the
dexamethasone pre-phase. Efficacy will be evaluated after 2 weeks based on the complete blood count
and a bone marrow aspirate. Treatment with STI571 continues in parallel to the treatment elements
specified in the protocol as long as they include the use of dexamethasone. During protocol II-IDA
(arm A) continuous treatment for three weeks is planned whereas treatment in arm B is given weekly
in parallel to the R blocks.
The dose follows the recommendations established by the International meeting for the use of STI571
in children with Philadelphia chromosome-positive ALL (June 25th 2001, Hannover Germany). The
exact administration and documentation of therapy will be coordinated with the study center. The use
of STI571 will be evaluated in a separate cooperative study of several European study groups in order
to reach conclusive results given the small case numbers. The efficacy of the treatment will be
compared to historical controls. In addition, BCR-ABL mRNA will be measured as a quantitative
parameter during the course of treatment.
3.8.3.2 Re-intensification for S3/4 patients with a positive MRD result prior to SCT
If the poor prognosis of patients in group S3/S4, who have a positive MRD result prior to SCT, is
confirmed during the course of the current study, the evaluation of a re-intensification block is planned
that aims at the elimination of MRD prior to SCT. To this end, a regimen including fludarabine, high-
dose cytarabine and daunoxome (FLAD) will be considered, which was designed based on the
experience with protocol IDA-FLAG (fludarabine, high-dose cytarabine, idarubicin and G-CSF) in the
treatment of relapsed AML (Bellott et.al, 2001; Fleischhack et.al, 1998; McCarthy et,al. 1999). A
corresponding concept is currently being evaluated by the MRC/UKALL study group. This group
reported a particularly high relapse rate in children with a positive MRD result prior to SCT possibly
as a result of a comparatively intensive T cell depletion during the conditioning phase prior to SCT
(Knechtli et al., 1998).
A corresponding treatment plan will be added as an amendment to the current protocol if the need
arises. Treatment will be planned in collaboration with the British study group.
leukemic efficacy of the randomized treatment elements and to determine the prognostic relevance of
MRD at these time points. This applies to all strategic groups. In children with an isolated
extramedullary relapse the clonal probes will be selected in close collaboration with the MRD
laboratories in Heidelberg (Dr. Flohr ALL-BFM), Hannover (Dr. Schrauder, ALL-BFM-HR) and
Hamburg (Dr. zur Stadt, COALL) taking into account the clone specific DNA sequences derived from
samples at primary diagnosis. Instructions regarding the shipment of samples and lab addresses are
listed in chapters 9.6, 9.7, 9.8 (p.73-74). Requisitions are listed in the appendix (p.130).
which may occur up to 6 years after the diagnosis of the initial relapse. Treatment-induced deaths are
possible given the overall intensity and toxicity of therapy, particularly of allogeneic SCT, which is
required in a subset of patients. Late organ toxicity and rare cases of second malignancies are possible
following the renewed and intensified chemotherapy. Particularly after SCT a proportion of patients
may develop significant late effects that are frequently associated with chronic graft-versus-host
disease.
With this background the intensification of therapy appears justified for groups with an unfavorable
prognosis. A time of 4 years is planned for the accrual of patients. Follow-up observations for at least
another 5 years are required to capture frequent late events. Late effects beyond this time frame will be
assessed by additional specifically designed studies.
The results of this study have a direct impact on the future treatment of children with relapsed ALL.
Successful studies of the ALL-REZ BFM group are used as a standard therapy in many countries that
are not participating in this study. The consulting activity of the study center, therefore, by far exceeds
the geographic region of the main study.
Protocol II-IDA vs. R blocks
Arm A (protocol II-IDA) is an approach to further optimization of therapy in comparison to standard
treatment in arm B (R blocks) in order to consolidate a remission. The aim is to optimize the anti-
leukemic efficacy and to reduce organ toxicity (see chapter 3.2, p.23). In addition, a significant
reduction of cost is anticipated due to the decreased need for in-patient treatment during protocol II-
IDA. The protocol allows a better degree of treatment control compared to the treatment blocks used
to date. The risk of increased toxicity associated with the new treatment arm containing protocol II-
IDA, therefore, is rated low. Extensive experience using a comparable protocol is available from
primary ALL therapy and by now also from the treatment of relapse.
Stratification according to the MRD result after the second treatment element
Monitoring of MRD in patients of group S2 allows the distinction of patients with a high vs. a low risk
of recurrence. On this basis the indication for stem cell transplantation can be determined
unequivocally. The indication is no longer dependent on a subjective assessment by the treating
physician and the families or on the local interests of the treating centers. This has to be regarded as a
definite advantage for the patients. The risk that a good prognosis patient is treated too intensively
with bone marrow transplantation or, conversely, that a patient with a high probability of relapse does
not gain access to the necessary intensification of therapy by allogeneic SCT is significantly reduced
by a stratification that is based on the MRD result after the second treatment element. Table 6 (p.27)
shows that the indication for transplantation based on MRD is present in a larger number of patients
than in the previous studies. This fact, however, corresponds with the still unsatisfactory results in the
entire group S2 with an event-free survival of 35-40% and the inability to predict a subsequent relapse
for individual patients in this group.
The use of MRD for the stratification of this patient group, therefore, is necessary on ethical grounds
given the current status of our knowledge. In this instance neither the use of a standard arm nor the
randomized introduction of this criterium make sense since historically there has been no standard
approach. Potential statistical interactions with the main study question, therefore, have to be tolerated.
A favorable effect on the EFS in this group can be determined using historical controls.
Shortening of treatment-free intervals
The design of the current study realizes the aim of intensifying induction therapy by shortening the
intervals between treatment blocks. This aim was already formulated in study ALL-REZ BFM 96 but
in clinical practice was not always realized according to protocol. In this regard this protocol
modification, therefore, represents a standardization of treatment. A delay of therapy for non-medical
reasons will be avoided. The increased treatment intensity during the initial phase of therapy can in
principle be associated with increased toxicity. The guidelines for administering chemotherapy that are
specified in the protocol are designed to avoid excessive risks. In view of the success achieved and the
data published so far, we anticipate that the patients will benefit from this intensification in form of an
ALL-REZ BFM 2002 34 Protocol version: 25.06.2003
increased remission induction rate and event-free survival. The risk of higher toxicity, therefore,
appears justified.
The efficacy of the increased treatment intensity can be compared with historical control using the first
end point of the study, the remission induction rate.
Standardization of induction therapy for group S4
Induction therapy with blocks F1 and F2 in group S4 was already used in studies ALL-REZ BFM 85
and 87. The toxicity of this therapy, therefore, is known and can be estimated. In a comparison with
historical controls this induction therapy showed an advantage over the induction blocks I and S of
study ALL-REZ BFM 96. Particularly, the hope for decrease of deaths during induction did not
materialize. Induction therapy using F blocks, therefore, represents a benefit for the patients with a
decreased risk. An additional advantage is the standardized design of the induction therapy in all
strategic groups.
Standardization of L-asparaginase therapy
The precise definition of treatment with L-asparaginase is also intended to standardize the clinical
practice when compared to the precursor studies. The risk of ineffective treatment due to silent
inactivation will be minimized by the mandatory monitoring of L-asparaginase activity. Endpoints for
this evaluation of L-asparaginase therapy are the duration of L-asparaginase activity in the serum and
the occurrence of hypersensitivity reactions.
Modification of continuation therapy
The use of daily oral 6-mercaptopurine and weekly oral methotrexate during continuation therapy
results in an improvement of the quality of life for patients during this long treatment phase. This
therapy is well documented during primary therapy of ALL and usually is better tolerated. In addition,
problems of compliance with bi-weekly intravenous injections are avoided. A decreased efficacy is not
anticipated since the treatment will be adjusted based on white cell counts and liver function tests.
Autologous SCT for children with prognostically unfavorable isolated CNS relapse.
The introduction of autologous SCT for children with a prognostically unfavorable isolated CNS
relapse is based on new results that allow the delineation of a group of patients with a very
unfavorable prognosis. Since the event-free survival in this patient group is less than 20% after
chemotherapy alone, intensification of consolidation therapy is absolutely mandatory. The anti-
leukemic efficacy of autologous SCT for this group is solely based on data in the literature. The
experience of the ALL-REZ BFM group does not support the efficacy of allogeneic SCT for isolated
extramedullary relapse. The stratification proposed in this protocol allows a standardized approach for
children with high-risk CNS relapse and defines a specific group of standard-risk patients for whom
there is certainly no indication to further intensification of therapy.
Modification of therapy for high risk patients during the course of the study
Possible modifications of therapy for patients with BCR-ABL-positive leukemia or for patients in
group S4 are mentioned in this study in order to ensure a close feedback with the study center.
Pertinent protocol modifications will be presented in a timely fashion to the responsible committees.
Such treatment modifications constitute separate protocols with independent reviews by the
institutional research ethics board and are not part of the current study protocol.
Diagnostic monitoring during the course of the study
The confirmation of remission by bone marrow aspiration prior to the start of the treatment blocks
during induction and consolidation therapy was not standardized in the previous studies. Mandatory
bone marrow aspirates were specified in the protocol only until a cytologic remission was achieved.
The current protocol uniformly specifies bone marrow aspirates up until the start of the first block R1
following protocol II-IDA (arm A) and up until the start of the second block R1 (arm B), respectively.
These aspirates allow the confirmation of remission for all patients by cytology. Patients with an
incipient relapse prior to a scheduled SCT will be detected systematically and excluded. MRD results
at these time points allow a direct evaluation of the anti-leukemic efficacy of the used treatment
ALL-REZ BFM 2002 35 Protocol version: 25.06.2003
elements. MRD results can be used as a first end point for the randomized main question of the study.
At the same time the prognostic relevance of additional time points can be evaluated and taken in to
account by subsequent studies. The direct benefit for the patient is limited to the monitoring of
remission by cytology. The MRD data derived from additional bone marrow aspirates will not be
communicated but evaluated prospectively. The bone marrow aspirates are performed during the
sedation or general anesthetic for the lumbar punctures required by the protocol at the start of each
block. Dependent on local practice the bone marrow aspirate may require additional or increased
sedation/anesthesia. A moderate degree of pain, which in most cases may not require treatment, has to
be anticipated after bone marrow aspiration.
ALL-REZ BFM 2002 36 Protocol version: 25.06.2003
4 STUDY DESIGN
Observation patients are defined as study patients who are not protocol patients. These patients meet at
least one of the following exclusion criteria at diagnosis or during the duration of treatment. They are
evaluated as a separately.
Patients will be excluded from the study if:
they have completed the 18th year of life at the time the relapse is diagnosed.
curative therapy is declined either by patient himself/herself of the respective legal guardian
the patient is pregnant
the patient is breast feeding
essential parts of the relapse therapy are declined either by the patient or his/her legal guardian or
cannot be administered because of medical reasons.
no consent is given for transmission of data
the patient has a severe concomitant disease that does not allow treatment according to the
protocol (e.g. malformation syndromes, cardiac malformations, metabolic disorders).
Patient with systemic diseases such as Down syndrome, cystic fibrosis or diabetes mellitus are eligible
for enrolment in this study. Due to the anticipated increase of toxicity dose reductions are suggested
after discussion with the study center.
Information about a subsequent relapse or relapse after SCT will also be collected as part of this
study. The study center also provides treatment recommendations for this group of patients.
Registration of a patient has to occur within fourteen days from the diagnosis of relapse. The result of
the randomization will be communicated within one business day.
a remission and a condition that allows transplantation. The high treatment intensity during induction
is intended to improve the remission induction rate and the quality of remission prior to SCT.
Randomization will determine the best strategy to achieve a high remission induction rate, quality of
remission prior to SCT and event-free survival. SCT is planned at an early time point, usually after the
third R block in Arm B and at the end of protocol II-IDA in arm A, respectively.
arm
A Prot II - IDA
S1 F1 F2 R
R2 R1 R2
R1 R2 R1 D 12
B
A Prot II - IDA R1 R2 R1 D 24 / V
S2 F1 F2 R R1 R2 S
B R2 R1 R2 SCT
A Prot II - IDA
S3/4 F1 F2 R SCT
B R2 R1 R2
BMA/
MRD (S1) (S2)
D12/D24, 12/24 months continuation therapy; R, Randomization; S, Stratification; V, VP16-reinduction pulse; , local radiation therapy;
, time point of bone marrow aspirate for stratification of post-remission therapy in S2; SCT, stem cell transplantation;
BMA, bone marrow aspirate; MRD, minimal residual disease; F1, F2, R2, R1, Protokoll II-IDA: blocks of chemotherapy.
ALL-REZ BFM 2002 41 Protocol version: 25.06.2003
generally following protocol II-IDA or the third R block, respectively. If a matching related or
unrelated donor cannot be identified, experimental SCT should be performed.
The prospective monitoring of MRD is used to assess the quality of remission prior to SCT. At this
point no decision is made based on the MRD result prior to SCT. If the unfavorable prognostic value
of a highly positive MRD result (>10-3 ) is confirmed, an amendment of the protocol for these patients
during the course of the study is possible.
4.10.2 Orchiectomy
Orchiectomy is the local therapy of choice in case for a clinically involved testis. The procedure is
performed at the beginning therapy if the clinical finding is unequivocal or during the course of
therapy if histopathological confirmation is required. In this case the decrease in size of the testis can
be used as an indicator for the response to therapy. During orchiectomy a testicular prosthesis should
be implanted. The cosmetic result is typically better than that of testicular atrophy following local
irradiation with 24Gy. Subclinical involvement of the clinically not involved contralateral testis has to
be investigated by biopsy. Depending on the result local irradiation is given according to the
guidelines described in chapter 4.9.3.
4.11.2 Indications
A schematic overview of the indications for different forms of transplantation in the various risk
groups as determined by the study committee and the Pediatric Working Group for Bone Marrow and
Stem Cell Transplantation is shown in table 11 (p.46).
ALL-REZ BFM 2002 45 Protocol version: 25.06.2003
Mandatory stem cell transplantation is recommended for all patients of group S3 and S4. Group 1
donors (MSD) are considered as a first choice followed by group 2 donors (MD). If a suitable donor
cannot be identified within two to three months there is an option to perform a transplant using a group
3 donor (MMD). Whether a haploidentical parent or a HLA-mismatched unrelated donor is preferred
in this group depends on the individual HLA constellation and should be clarified in discussion with
the study centers of ALL SZT-BFM 2003 and ALL-REZ BFM.
The transplant indication for patients with bone marrow involvement in group S2 depends on the
MRD result after the second treatment element (F2). An expeditious search for an unrelated donor is
essential for patients with a MRD result of > 10-3 since the donor search will only start after the
relevant MRD results following the second treatment element has been received. This result can be
expected at the earliest one to three weeks after the beginning of the first R block (arm B) or of
protocol II-IDA (arm A). Transplant from an unrelated donor in group S2, therefore, can generally be
performed no sooner than after 5 R blocks or after two R blocks following the completion of protocol
II- IDA, respectively. For patients in group S2 with a MRD result > 10-3 only donors of group 1
(MSD) and group 2 (MD) are considered as unrelated donors. If no matching MSD or MD can be
identified the patient receives chemotherapy and radiotherapy as specified in the protocol followed by
continuation therapy. If stratification based on MRD testing is not available, the indication for
transplantation is determined according to conventional criteria.
Patients in group S2 with an isolated CNS involvement and high-risk criteria (see chapter 3.8.2, p.29)
proceed to autologous SCT.
S2
MRD
S1 CNS S3/S4
<10-3 n.d. 10-3
subgroup* A B/C A B/C A/B/C SR HR
MSD-SCT - - + + + + - - +
MD-SCT - - - - + + - - +
MMD-SCT - - - - - - - - +
autologous SCT - - - - - - - + -
Legend: MSD, matched sibling donor (group 1); MD, matched ( 9/10 AG) family/unrelated donor (group
2); MMD, mismatched (< 9/10 AG) family/unrelated donor (group 3); MRD, minimal residual disease; CNS-
HR, isolated CNS relapse, high risk group; CNS-SR, isolated CNS relapse, standard risk group.
* for the definition of groups S2A-D, see table 3 (p.21); for the definition of groups CNS-SR/HR, see table 7
(p.30)
It is essential to initiate the search for an unrelated donor early in order to be able to perform
transplantation at the best point in time. The transplant center should be contacted in time to discuss
donor selection, preparation, appointments and possible alternative strategies.
The Institute for Transplantation Diagnostics and Cell Therapeutics at the University Hospital
Düsseldorf offers high resolution HLA-C typing as part of a study free of charge (contact PD. Dr. D.
Dilloo, tel 0211/8116224, or Dr. J. Enczmann, tel 0211/8118684).
Autologous SCT
Henze, Gunter, Prof. Dr. Dr. h.c.
Kühl, Jorn, Dr. med.
Charité-Universitatsmedizin Berlin, CVK
Klinik for Pediatrie mit Schwerpunkt Hämatologie/Onkologie, OHC
Augustenburger Platz 1
Tel: 49-30-450-566032
Fax: 49-30-450-566906
E-mail: [email protected]
ALL-REZ BFM 2002 48 Protocol version: 25.06.2003
4.11.5 Documentation
Following the registration of a patient the primary treating center receives the notification of the
randomization result, a summary of all clinical data available to the study center and a statement
regarding the indication for transplantation. Feedback regarding the already known types of donors
and the intention to perform a SCT will be provided using a form attached to the first statement. Once
the MRD result after the second treatment element (F2) is available for patients in group S2 with bone
marrow involvement the treating center receives a second updated statement. When the patient is
admitted to a transplant center for SCT the ALL-REZ BFM study center is notified using the data
form prepared by the Pediatric SCT Registry. The study center forwards the data to the Pediatric SCT
Registry and corresponding SCT coordinators and sends a summary of the available patient data to the
transplant center.
On day 100 after SCT the course of therapy will be documented by the transplant center using the
documentation form of the SCT Registry (Form Med A), which is sent to the ALL-REZ BFM study
center. The study center ensures the transfer of data to the Pediatric SCT Registry and the SCT
coordinators.
5 TREATMENT ELEMENTS
5.3 Block F1
Niveles Mxt hora 48
Folinato hora 48 y 54
5.4 Block F2
drug route dose day
dexamethasone DEXA p.o. 20 mg/m2/d 1 2 3 4 5
vincristine VCR i.v. 1.5 mg/m2/d 1
cytarabine ARA-C 3h infusion 2 x 3 g/m2/d 1 2
E.Coli L-asparaginase Coli-ASP* 6h infusion 10.000 U/m2 4
methotrexate MTX intrathecal based on age 5
cytarabine ARA-C intrathecal based on age 5
prednisone PRED intrathecal based on age 5
* in case of allergic reaction or silent inactivation chose an alternative preparation according to the guidelines specified in the
protocol
5.5 R2-Block
drug route dose day
dexamethasone DEXA p.o. 20 mg/m2/d 1 2 3 4 5
thioguanine 6-TG p.o. 100 mg/m2/d 1 2 3 4 5
vindesine VDS i.v. 3 mg/m2/d 1
methotrexate MTX 36h Infusion 1 g/m2 1
ifosfamide IFO 1h Infusion 400 mg/m2/d 1 2 3 4 5
daunorubicin DNR 24h Infusion 35 mg/m2 5
E.Coli L-asparaginase Coli-ASP* 6h Infusion 10.000 U/m2 6
methotrexate MTX intrathecal based on age 1
cytarabine ARA-C intrathecal based on age 1
prednisone PRED intrathecal based on age 1
* in case of allergic reaction or silent inactivation chose an alternative preparation according to the guidelines
specified in the protocol
in case of CNS envolvement repeat intrathecal chemotherapy on day 5
ALL-REZ BFM 2002 51 Protocol version: 25.06.2003
5.6 R1-Block
* in case of allergic reaction or silent inactivation chose an alternative preparation according to the guidelines specified in the protocol. In case of CNS involvement an additional dose
of intrathecal chemotherapy is given on day 8.
ALL-REZ BFM 2002 53 Protocol version: 25.06.2003
6 DRUGS
6.1.1 L-asparaginase
L-asparaginase is administered starting on day 4 of protocol of F1 and F2, day 6 of protocol R1 and
R2 and on day 1 of protocol II-IDA.
Native E.coli L-asparaginase is used for all patients unless there was an allergic reaction or silent
inactivation during primary therapy. The drug is administered on day 4 of block F1/F2, on day 6 of the
R blocks, and on day 1, 6 11 and 16 of protocol II-IDA at a dose of 10,000units/m2 as a 6-hour
infusion. The infusion of L-asparaginase should be started at a reduced rate and increased stepwise.
The measurement of L-asparaginase activity in the serum is mandatory five days after each
administration of E.coli L-asparaginase and will be performed by the pharmacology lab at the
University Children’s Hospital Münster (Prof. Dr. Boos). If these results demonstrate a silent
inactivation or if an overt allergic reaction is observed, PEG-L-asparaginase (oncospar, medac) will be
used instead at a dose of 1,000units/m2 BSA infused intravenously over two hours as long as this
preparation was tolerated during primary therapy. During protocol II-IDA PEG-L-asparaginase is
administered on day 1 and 11. Mandatory measurement of PEG-L-asparaginase activity is performed
2, 7 and 14 days after administration. If an allergy to this preparation is already known or occurs
during relapse therapy, Erwina L-asparginase (Erwinase, Ipsen, Ltd.), will be used as a third
preparation at a dose of 10,000 units/m2 BSA on days 4, 6 and 8 of the F blocks, on day 6, 8 and 10 of
the R blocks and on day 1, 3, 5, 7, 9, 11, 13, 15, 17 and 19 of protocol II-IDA, respectively. L-
asparginase activity will be measured 48 hours after each application.
After an allergic reaction the alternative L-asparaginase preparation is introduced during blocks F and
R blocks with the next block or protocol and during protocol II-IDA at the next scheduled
administration of the alternate preparation during the ongoing protocol.
During L-asparaginase therapy vital signs should be closely monitored. All necessary measures to treat
allergic reactions including an anaphylactic shock must be available.
6.1.2 Cyclophosphamide
Cyclophosphamide is administered during protocol II-IDA on day 29 at a dose of 1g/m2 a an
intravenous infusion over 1 hour. Mesna is administered intravenousy at a dose of 400 mg/m2 BSA
prior to as well as 4 and 8 hours after the cyclophosphomide infusion. Sufficient hydration with 3000
ml/m2 was to be ensured for 24 hours from the start of cyclophosphamide (see infusion orders).
6.1.3 Cytarabine
Cytarabine is administered during block F2 on day 1 and day 2 (two doses of 3g/m2 BSA) and during
block R1 on day 5 (two doses of 2g/m2 BSA). The interval between the two daily doses is 12 hours,
the duration of infusion is 3 hours. Attention should be paid to the administration of a sufficient
amount of fluids and to conjunctivitis prophylaxis. Prior to each infusion of cytarabine vitamin B6
should be given intravenously at a dose of 100mg/m2 BSA. Antiemetic prophylaxis using 5-HT3
antagonists is started one hour prior to cytarabine infusion (3 hours in case of oral administration) and
continued at 12 hourly intervals, e.g. ondansetron 5mg/m2 BSA p.o. or i.v. (see infusion orders sets in
the appendix, p.116,117)
During protocol II-IDA cytarabine is administered on days 31 to 34 and on days 38 to 41 at a dose of
75 mg/m2 BSA i.v.
In addition, cytarabine is part of the intrathecal therapy at an age-dependent dose (chapter 5.2, table
12, p.49).
ALL-REZ BFM 2002 54 Protocol version: 25.06.2003
6.1.4 Daunorubicin
Daunorubicin is infused on day 5 of block R2 at a dose of 35 mg/m2 BSA in normal saline over 24
hours (following the ifosfamide infusion). If a peripheral venous access is used a concentration of
0.05mg/ml should not be exceeded. The amount of normal saline infused in parallel to the ifosfamide
infusion has to be decreased accordingly. If a central venous access is used any concentration can be
selected.
6.1.5 Dexamethasone
Dexamethasone is administered at a dose of 20 mg/m2 BSA/day on day 1 through 5 of protocol F1, F2,
R1 and R2 and at a dose of 6mg/m2 BSA/day on day 1 through 14 and at a tapering dose (decrease the
dose by half every three days) on day 15 through 23 of protocol II-IDA. The daily dexamethasone
dose should be divided in two or three doses.
6.1.6 Etoposide
Etoposide is administered orally as a reinduction pulse during continuation therapy for group S2, at a
dose of 50mg/m2 BSA/day for 10 days. A total of four reinduction pulses are scheduled.
6.1.7 Idarubicin
Idarubicin is administered on day 1, 8, 15 and 22 of protocol II-IDA at a dose of 6 mg/m2 BSA as an
infusion over 6 hours. The drug is dissolved in 20-40 ml normal saline/mg idarubicin. If a peripheral
venous access is used a dilution to at least 0.01 mg/ml should be selected.
Children under the age 2 years should receive a reduced dose of idarubicin after discussion with the
study center. Renal or hepatic dysfunction may also require a dose reduction dependent on the degree
of impairment.
6.1.8 Ifosfamide
Ifosfamide is administered during block R2 on day 1 through 5 at a dose of 400 mg/m2 BSA as an
intravenous infusion over 1 hour. On day 1, ifosphamide is administered prior to the methotrexate
infusion, on day 2 after completion of the methotrexate infusion, and on day 5 prior to the
daunorubicin infusion. Mesna (200 mg/m2BSA) is administered intravenously prior to as well as 4 and
8 hours after each ifosfamide infusion. Simultaneously, the administration of a sufficient amount of
fluids has to be ensured.
6.1.9 Methotrexate
Methotrexate is administered on day 1 of block F1, R1 and R2 at a dose of 1000 mg/m2 BSA over 36
hours. One tenth of the solution is infused during the first half hour, the remaining nine tenth over 35.5
hours. In parallel forced alkaline diuresis with 3000 ml/m2BSA/24 hours is used on day 1 and 2 (see
infusion orders, appendix, p.115). Serum methotrexate levels are measured at the start, at the end and
48h after the start of the MTX infusion. The 48 hour level has to be measured immediately since it is
the basis of the folinic acid rescue. The result has to be communicated promptly to the responsible
physician.
In addition, methotrexate is part of the intrathecal chemotherapy at an age-dependent dose (see chapter
5.2, table 12, p. 49).
increased toxicity has to be anticipated. If the methotrexate level at 48h is greater than 1.0 mol/L, an
increased dose and potentially an increased number of doses of folinic acid are required in accordance
with the rescue schema (see appendix, p.120). If the methotrexate level at 48 hours is greater than 2
mol/L, it is in addition recommended to prolong the duration of forced alkaline diuresis. The
measurement of methotrexate levels and the corresponding administration of folinic acid are continued
at 6 hourly intervals until the methotrexate level falls below 0.25 mol/L.
6.1.11 6-Mercaptopurine
6-Mercaptourine is administered orally at a dose of 100 mg/m2 BSA from day 1 to day 5 of block R1.
It is also administered daily during continuation therapy. The recommended dose is 50 mg/m2 BSA
with dose adjustments according to the white cell count (see guidelines for administering therapy,
p.60). Oral 6-mercaptopurine should be administered in the evening.
6.1.12 Prednisone
Prednisone is part of the intrathecal chemotherapy at an age-dependent dose (chapter 5.2., table 12, p.
49).
6.1.13 6-Thioguanine
6-Thioguanine is administered orally at a dose of 100 mg/m2 on day 1 to 5 of block R2. In addition, it
is administered orally at a dose of 60 mg/m2 from day 29 to 42 of protocol II-IDA. Thioguanine should
be given as a single dose in the evening.
6.1.14 Vincristine
Vincristine is administered strictly intravenously at a dose of 1.5 mg/m2 BSA on day 1 and 6 of block
F1 and R1, on day 1 of block F2 as well as on day 1, 8, 15 and 22 of protocol II-IDA. The maximal
single dose is 2 mg.
6.1.15 Vindesine
Vindesine is administered strictly intravenously at a dose of 3 mg/m2 BSA on day 1 of block R2.
6.2.1 L-asparaginase
L-asparaginase is a bacterial enzyme that catalyzes the conversion of asparagine to aspartate and
ammonium as well as the conversion of glutamine to glutamate and ammonium. The depletion of L-
asparagine in the serum deprives leukemic lymphoblasts of this for them essential amino acid.
Although the cells of the human body are capable of synthesizing asparagine, organs with a high rate
of protein synthesis (liver, pancreas) will experience a relative deficiency of asparagine.
Contraindications include an episode or history of pancreatitis as well as pregnancy.
Documented side effects affect the skin (urticaria, hypersensitivity reaction), nervous system (cerebral
dysfunction with EEG changes, decreased level of consciousness), gastrointestinal system (anorexia,
nausea, vomiting, weight loss, acute hemorrhagic pancreatitis), liver (abnormal liver function,
increased bilirubine, alkaline phosphatase, decreased albumin and cholesterol, decreased
ALL-REZ BFM 2002 56 Protocol version: 25.06.2003
concentrations of clotting factors with abnormal coagulation and fibrinolysis), metabolism (impaired
glucose tolerance, decreased insulin level, hyperglycemia, ketoacidosis), circulation (hypotension,
shock), blood (abnormal blood counts, leukopenia, thrombocytopenia, hemolytic anemia), genito-
urinary system (renal toxicity, microhemoturia, albuminuria, casts, azotemia), and the immune system
(hypersensitivity reactions, urticaria, fever, hypotension, shock).
In case of a hereditary prothrombotic risk and an asparaginase-induced decrease of anti-thrombotic
factors the risk of thrombosis is increased. Treatment with low molecular weight heparin may be
considered in these cases.
6.2.2 Cyclophosphamide
Cyclophosphamide belongs to the group of oxazaphosphorines. It is an alkylating agent and interferes
with the replication and transcription of DNA. Its cytotoxic effect requires activation by microsomal
liver enzymes and intracellular cleavage of acrolein.
Contraindication include acute infections, severe myelosuppression as well as pregnancy and lactation.
Acrolein is toxic to mucosal surfaces and can cause hemorrhagic cystitis after renal elimination.
Mesna prevents this adverse effect by binding acrolein.
Additional documented side effects involve the skin (alopecia, dermatitis), nervous system (neural
toxicity), gastrointestinal system (gastrointestinal symptoms such as nausea, vomiting, diarrhea and
stomatitis), liver (liver damage), metabolism and endocrine system (hyperuricemia, impaired
spermatogenesis and ovulation), vascular system (irritation of vascular intima), blood (impaired
hematopoiesis) and genito-urinary system (renal injury and injury to the urinary tract).
Immunosuppressive effects as well as inflammation of skin and mucosal tissues (e.g. dermatitis,
stomatitis) are described. Cyclophosphamide interacts with other agents and measures that impair the
function of the bone marrow (increased cytotoxicity) and with anti-diabetic medications (resulting in
lower blood sugar).
6.2.3 Cytarabine
Cytarabine inhibits the synthesis of pyrimidines and is a member of the family of anti-metabolite
agents. Contraindications include acute infections, severe myelosupression as well as pregnancy and
lactation.
Documented side effects involve the skin (alopecia, skin reactions, dermatitis) and mucosal tissues
(ulcerations of the oral mucosa and the GI tract, stomatitis, conjunctivitis), musculo-skeletal system
(myalgia, arthralgia), nervous system (dysfunction of the central nervous system, neuritis, rarely
leukoencephalopathy, rarely paraplegia), gastrointestinal system (gastrointestinal symptoms such as
nausea, vomiting, diarrhea), liver (liver damage), metabolism (hyperuricemia), endocrine system
(impaired spermatogenesis and ovulation), heart (cardiac arrhythmia), respiratory system
(bronchospasm, pulmonary edema), blood (impaired hematopoiesis), genito-urinary system (abnormal
renal function) and the immune system (hypersensitivity reaction, immunosuppression). Interaction
with other agents and measures that suppress the function of the bone marrow results in increased
toxicity.
6.2.4 Daunorubicin
Daunorubicin is an anthracycline and belongs to the group of cytotoxic antibiotic drugs. The cytotoxic
effect is predominantly due to direct DNA damage.
Contraindications include acute infections, severe myelosuppression, a dose exceeding the maximal
cumulative anthracycline dose (risk of life-threatening cardiac damage), myocardial damage as well as
pregnancy and lactation. A limitation of the use of this agent should be considered in case of
pancytopenia, isolated leukopenia or thrombocytopenia, clinical heart failure, abnormal renal and liver
function, uncontrolled infection and a poor general health status of the patient.
ALL-REZ BFM 2002 57 Protocol version: 25.06.2003
Documented side effects affect the skin (reversible alopecia, dermatitis, local irritation),
gastrointestinal tract (ulcerative stomatis, nausea, vomiting, diarrhea), metabolism (hyperuricemia),
endocrine system (impaired spermatogenesis and ovulation, azoospermia, amenorrhea, irreversible
infertility), heart and circulation (cardiomyopathy - dependent on the dose, results in global cardiac
insufficiency, which may end in fatal cardiac failure-, bradycardia, cardiac arrhythmias), vascular
system (single cases of irritation of the intima after i.v. injection), blood (myelosuppression with
leukopenia, thrombocytopenia, anemia), genito-urinary system (urate nephropathy) and the immune
system (immunosuppression, allergic reactions). Interactions have to be considered with medications
such as other cytotoxic agents (increased cytotoxicity), cardiotoxic drugs (increased cardiotoxicity of
daunorubicin), irradiation (increased cardiotoxicity), hepatotoxic drugs (e.g. methotrexate, increased
hepatoxicity) and drugs that decrease the excretion of uric acid (e.g. sulfonamide and certain
diuretics).
Anthracyclines have to be infused strictly intravenously. Extravasation results in tissue ulceration and
irreversible local damage. Instructions for the management of an anthracycline extravasation are given
in chapter 8.1.3, p. 64.
6.2.5 Dexamethasone
Dexamethasone belongs to the halogenated glucocorticoids. Its mechanisms of action are multifold. It
blocks the release of arachidonic acid, the starting compound of prostaglandin and leukotriene
synthesis by inhibiting phospholipase 2. This results in anti-inflammatory, immunosuppressive and
ulcerogenic effects. Lymphoblastic leukemic cells express glucocorticoid receptors that have greater
affinity to dexamethasone than other glucocortocoids. Binding of dexamethasone to these receptors
results in programmed cell death of leukemic lymphoblasts.
Contraindications are gastrointestinal ulcers, severe osteoporosis, a psychiatric disorder, herpes
simplex, herpes zoster (viremic phase), varicella, a time period of approx. 8 weeks before to 2 weeks
after vaccinations, amoebal infection, systemic fungal disease, poliomyelitis with the exception of the
bulbar/encephalitic form, lymphadenitis after BCG vaccination, narrow and wide-angle glaucoma.
Parenteral administration of depot preparations and crystalline suspensions is not indicated in children
under the age of 6 years and children between 6 and 12 years, respectively. Restriction of its use has to
be considered in patients with a history of tuberculosis (reactivation!) and in case of severe infections.
Side effects are described involving the skin (striae, petechiae, ecchymoses, acne, delayed wound
healing), muskulo-skeletal system (muscle weakness, osteoporosis, avascular necrosis of femoral and
humeral head), eyes (glaucoma, cararact), psychiatric symptoms (depression, irritability, euphoria),
gastrointestinal tract (epigastric pain, peptic ulcer, pancreatitis), electrolytes, metabolism and
endocrine system (cushingoid face, truncal obesity, impaired glucose tolerance, diabetes mellitus,
sodium retention and edema, increased renal loss of potassium, adrenal insufficiency, decreased
growth in children, abnormal secretion of sex hormones – e.g. amenorrhoea, hirsutism, impotence),
circulation (hypertension), vascular system (increased risk of thrombosis, vasculitis – withdrawal
syndrome after long term use) and the immunsystem (allergic reaction including – rarely- shock,
immunosuppression, increased infectious risk).
Interactions have been observed with other medications such as cardiac glycosides (glycoside effect is
enhanced by hypokalemia), diuretics, loop diuretics (additional potassium secretion depending on the
mineralocorticoid effect), antidiabetics (increased blood sugar), oral anticoagulants (decreased
anticoagulant activity), induction of cytochrome p450 (e.g. rifampicin, phenytoin, barbiturates,
primidone: decreased effect of corticoids), non-steroidal anti-inflammatory drugs (risk of
gastrointestinal bleeding and ulceration), ACE inhibitors (increased risk of abnormal blood counts),
chloroquin, hydroxychloroquin, mefloquin (increased risk of myopathies and cardiomyopathies),
somatotropin (decreased somatotropin activity), protirelin (decreased rise of TSH), laxatives
(increased potassium loss) and salicylates (increased risk of gastrointestinal bleeding).
ALL-REZ BFM 2002 58 Protocol version: 25.06.2003
6.2.6 Etoposide
Etoposide is a derivative of epipodophyllotoxin. It inhibits cell division in the pre-mitotic phase and is
predominantly cytotoxic during the late S- or early G-phase. Etoposide interferes with DNA repair by
inhibiting topoisomerase II. Contraindications and side effects - see cyclophosphomide.
6.2.7 Idarubicin
Idarubicin is anthracycline that belongs to the group cytotoxic antibiotics. Contraindications and side
effects see daunorubicin.
6.2.8 Ifosfamide
Ifosfamide belongs to the group of oxaphosphorines. It is an alkylating agent that interferes with DNA
replicaton and transcription. The cytotoxic effect requires activation by microsomal liver enzymes and
intracellular cleavage of acrolein. For contraindications and side effects see cyclophosphamide.
6.2.9 Methotrexate
Methotrexate is a folate antagonist and belongs to the group of anti-metabolites. It interferes with
synthesis of purines and pyrimidines by inhibiting dihydrofolate reductase.
Contra indications include acute infection, severe myelosuppression, abnormal liver function,
gastrointestinal ulceration, renal insufficency (nephrotoxic even at low dose; at high doses causes
additional renal impairment by precipitation of methorexate), as well as pregnancy and lactation.
Documented side effects involve the skin (exanthems, toxic skin reactions - e.g. exanthems, pruritus,
photosensitivity, very rarely Lyell syndrome -, alopecia, dermatitis), the musculo-skeletal system
(osteoprosis), the gastrointestinal tract (gastrointestinal abnormalities, e.g. nausea, vomiting, diarrhea,
intestinal hemorrhage, ulceration of the oral mucosa and gastrointestinal tract, stomatitis), liver (liver
damage), metabolism and the endocrine system (hyperuricemia) impaired spermatogenesis and
ovulation), the vascular system (vasculitis), the respiratory system (pulmonary infiltrates, fibrosis) the
blood (impaired hematopoiesis), the genito-urinary system (renal dysfunction), the immune system
(allergic reactions immunosuppression). Inflammation of skin and mucosal surfaces (e.g. dermatitis,
stomatitis) and teratogenic effects are described.
Interactions with other medications and measures that are myelosuppressive and enhance the toxicity
of methotrexate are known. Non-steroidal anti-inflammatory drugs, phentoin, barbiturates,
tetracyclines, chloramphenicol, sulfonamides, p-amino benzoic acid, p-amino hippuric acid and
metamizole can increase the toxicity of methotrexate.
An impairment of the ability to eliminate high-dose methotrexate can result in life-threatening
complications. Guidelines for the management of decreased elimination of methotrexate are described
in chapter 8.1.2, p. 64.
6.2.10 Mercaptopurine
6-Mercaptopurine is a purine analog that belongs to the anti-metabolite drugs. It results in chromatin
damage through the incorporation of false nucleotides into DNA.
The use of mercaptopurine is contraindicated during pregnancy and lactation.
Documented side effects involve the gastrointestinal tract (gastrointestinal symptoms, nausea,
vomiting, anorexia, ulceration of the oral mucosa and of the gastrointestinal tract), the liver (abnormal
liver function, liver damage), the genitourinary tract (secondary hyperuricemia) metabolism and the
endocrine system (impaired spermatogenesis and ovulation), blood (abnormal hematopoiesis,
leukopenia, thrombocytopenia). Drug-induced fever, pancreatitis and secondary leukemia have also
been described. Interaction of 6-mercaptupurine with allopurinol and anticoagulants has to be kept in
mind.
ALL-REZ BFM 2002 59 Protocol version: 25.06.2003
6.2.11 Prednisone
Prednisone belongs to the non-halogenated glucocorticoids. For contraindications and side effects see
dexamethasone.
6.2.12 6-Thioguanine
6-Thioguanine belongs to the anti-metabolites. The use of 6-thioguanine is contraindicated in Lesh-
Nyhan Syndrome (decreased efficacy) as well as during pregnancy and lactation.
Documented side effects affect the gastrointestinal tract (gastrointestinal abnormalities, nausea,
vomiting, anorexia, ulceration of the oral mucosa and of the gastrointestinal tract, stomatitis, necrosis
of the intestinal mucosa, intestinal perforation), liver (abnormal liver function, jaundice, veno-
occlusive disease, centrilobular liver necrosis), metabolism and endocrine system (impaired
spermatogenesis and ovulation) and blood (impaired hematopoieis, leukopenia, thrombocytopenia).
Interaction with other medications and measures that are myelosuppressive and increase toxicity are
known. Interactions with busulfan include nodular hyperplasia of the liver, portal hypertension and
esophageal varices.
6.2.13 Vincristine
Binding of vincristine to tubulin results in a blockade of mitosis. Vincristine belongs to the group of
vinca alkaloids. Contraindications include acute infections, severe mylosuppression as well as
pregnancy and lactation.
The predominant adverse effect is neurotoxicity. Loss of deep tendon reflexes, parathesias, cranial
nerve palsies, marked weakness particularly of the extremities, marked myalgia as well as a syndrome
of inadequate ADH secretion (SIADH) have been observed. Abnormal function of the autonomous
nervous system is possible resulting in constipation, paralytic ileus, urinary retention, hypotonia and
impotence.
Other potential side effects include nausea, vomiting, alopecia and myelosuppression.
Vincristine has to be administered strictly intravenously. Extravasation results in tissue ulceration and
irreversibe local damage. Guidelines for the treatment of a vinca alkaloid extravasation are described
in chapter 8.1.3, p. 64.
6.2.14 Vindesine
Vindesine is a derivative of vinblastine and belongs to the group of vincalacaloids. For mechanism of
action, contraindications and side effects see vincristine.
ALL-REZ BFM 2002 60 Protocol version: 25.06.2003
7.2 F blocks
Both F blocks should be administered on time and regardless of the peripheral blood counts. Similar to
the practice of stem cell transplantation, platelet counts are maintained above 15 to 20,000 x 109/L
with HLA-matched platelet transfusions until a remission is achieved. Thus critical clinical situations
are safely manageable. Even fever and the in this scenario almost always necessary antibiotic therapy
alone are not a sufficient reason to delay therapy. The early achievement of a remission has priority
and frequently is an essential prerequisite for the long term control of infections. If the patient's
clinical condition is critical, for example in the case of blood pressure problems, sepsis, coagulopathy
and severe mucositis with massive protein loss, the ultimate decision rests with the treating physician.
Creatinine <100 >= 100 > 250 > 450 > 800
[µmol/L]
Stomatitis none soreness, erythema, ulcers, ulcers, liquid diet oral nutrition not
erythema hardly any solid possible
food
Diarrhea none Transient tolerable not tolerable, Bloody diarrhea,
2 days > 2 days treatment dehydration
required
Constipation none mild moderate subileus Ileus
8 SUPPORTIVE CARE
The main problem of intensive multi-agent chemotherapy is the combination of marked
immunosuppression, direct organ and mucosal toxicity and the resulting immunodeficieny toward
potentially pathogenic microorganisms. A number of protective and supportive measures are urgently
required to prevent potentially serious harm associated with therapy.
8.1 Emergencies
The local area should be kept warm (in contrast to the cooling recommended for anthracycline
extravasations).
If a necrosis develops despite these local measures early surgical revision should be considered.
The amphotericin suspension is carefully spread over the entire oral mucosa and then swallowed. If
prophylaxis with amphotericin solution is not feasible or if thrush becomes apparent despite
prophylaxis, fluconazole (approx. 2 mg/kg/d) can be used as an alternative. Hepatic toxicity and
possible drug resistance have to be considered.
The inhalation of amphothericin B BID is urgently recommended. 2 ml Amphotericin B stock
solution (1 vial = 50mg, dissolved in 10 ml distilled water) is used for one inhalation. The inhalations
proved useful in the prevention of infections with Aspergillus fumigatus in the bronchial system.
Severe large ulcerations generally are not limited to the mouth. They require close monitoring and a
consistent and early replacement of protein and electrolyte losses. In addition, sufficient analgesia
should be ensured including opiates as needed.
The mucosal area under the tongue generally is representative of the status of the entire
gastrointestinal tract. It remains almost always accessible to inspection and assessment even in cases
with marked swelling and pain.
This approach is simply an example that has to be supplemented by clinical findings and
microbiological results and requires modification according to the experience of the local treating
physician. Delays in the change of antibiotic medications may provide an irretrievable advantage to
problem organisms such as pseudomonas, coagulase negative staphylococci or aspergillus. If there is a
clinical suggestion of an infection with pseudomonas medication with certain efficacy such as
amikacin should be added. If an atypical pneumonia is suspected the combination of antibiotics should
include a macrolide antibiotic such as erythromycin.
Vigilance and clinical expertise are more important than pedantic adherence to a schema !
8.4.3 GCSF
GSCF (Filgrastim) was used in a randomized fashion during the precursor study ALL-REZ BFM 96. It
resulted in a shortening of treatment intervals. A prognostic impact, however, could not be
demonstrated.
Therefore, GCSF is used in protocol ALL-REZ BFM 2002 only with a supportive care indication. It is
used in patients who had a poor tolerance of therapy during preceding treatment blocks and who are
considered at risk of significant complications due to prolonged during periods of aplasia. The
decision to use of G-CSF lies with the treating clinician. G-CSF is used at a dose of 5 mg/kg body
weight/day s.c. or as a 4-hour infusion. It is administered 24 hours after the end of the preceding
chemotherapy block. G-CSF is discontinued if the neutrophil count exceeds 3.0 x109/L on two
subsequent occasions following the nadir of cell counts.
9 DIAGNOSTIC TESTS
The relapse of ALL requires a comprehensive diagnostic evaluation in order to classify the disease
according to established parameters and to adapt treatment to the risk profile of the patient. An
additional aim of relapse study ALL-REZ BFM 2002 is to define parameters that may provide insight
into the origin, course and prognosis of the disease and be helpful in the development of novel,
specific, efficacious and risk-adapted therapeutic strategies.
9.1 Definitions
9.2.2 CNS
Every time a relapse is diagnosed a diagnostic lumbar puncture is performed. This lumbar puncture
can be used to administer the first dose of intrathecal chemotherapy. If a CNS relapse is suspected a
CSF volume of at least 10 mL has to be collected since this sample is possibly the only material for the
design of a clonal probe to monitor MRD. The CSF has to be promptly assessed by cytology or
prepared for such an assessment.
If a CNS relapse is suspected and the CSF is unremarkable a cranial MRI should be performed to
detect a localized involvement. Such an involvement may have to be confirmed by biopsy.
9.2.3 Testis
In the interest of a precise diagnostic evaluation, the immunophenotyping of lymphoblasts and the
detection of molecular markers should also be performed in cases of isolated testicular relapse.
The biopsy sample should be sent in sterile saline to the local pathologist and to the study center for
review.
ALL-REZ Studienzentrale
First BM-syringe from both Berlin
sites! Prof. Dr. Dr. h.c. G. Henze
Diagnosis 2-3 x 5 ml heparin. BM
- molecular genetics
5 - 10 ml heparin. PB
- cytology
isol. extramed. manifestation
smears, unstained (5 PB, 5
BM, 2 CSF)
Robert-Rössle-Klinikum Buch
2 ml hepar. BM Prof. Dr. W.-D. Ludwig
2 unstained smears - immunphenotyping
after 1. R2 day 15
after 1. R1 day 29
after 2. R2 after
completion
prior to and after continu-
ation therapy
immediately prior to SCT
ALL-REZ Studienzentrale
smears, unstained (2 PB, Berlin
2 BM, 2 CSF) Prof. Dr. Dr. h.c. G. Henze
in case of a chromosomal - molecular genetics
translocation: - cytology
1 x 5 ml hepar. BM
1 x 5 ml hepar. PB
* Chimerism studies after SCT are performed according to protocol ALL BFM SZT 2003 in parallel in Tübingen
(PD Dr. P. Bader) and in Vienna (Prof. Dr. Dr. T. Lion).
ALL-REZ BFM 2002 70 Protocol version: 25.06.2003
9.8.3 Immunology
Samples for immunophenotyping include at least 2 ml of heparinized bone marrow and 5 ml of
heparinized peripheral blood and should be sent to:
Prof Dr. W.-D. Ludwig
Immunologisches Zellmarkerlabor
Charité, Campus Berlin-Buch
Robert Rössle Klinik – MDC
Lindenberger Weg 80
13122 Berlin-Buch
Germany
Tel: 030 - 9417-1362
Fax: 030 - 9417 - 1308
ALL-REZ BFM 2002 75 Protocol version: 25.06.2003
10 PATIENT SAFETY
The study center regularly assesses the stoppage criteria to ensure that any significant increase of
treatment-related mortality or relapse either in comparison to historical controls within the randomized
prospective comparison is recognized and acted upon according to the guidelines described in chapter
11.5, p.79. Additionally, adverse events are documented by the treating centers and severe adverse
events are reported immediately to the study center.
11.1 Definitions
Complete remission (CR)
-regenerating bone marrow with less than 5% blasts (M1) and
-peripheral blood without blasts and with evidence of regeneration and
-absence of extramedullary leukemic involvement.
Partial remission (PR)
Blast percentage in the bone marrow > 5% and < 25% (M2).
Early response
Patients have an early response if CR is achieved after the first treatment element (bone marrow
aspirate on day 15 following F1) or if the bone marrow is aplastic with blasts < 5% and the criteria of
CR are fulfilled within the subsequent four weeks.
Late response
Patients have a late response if a CR is achieved between day 15 and day 29 of protocol II-IDA in arm
A or after the fourth treatment element (first block R1) and prior to the fifth treatment element (second
block R2) in arm B.
Non-response (NR)
Non-response is diagnosed in patients who have not achieved a complete remission by day 29 of
protocol II-IDA in arm A or by the start of the fifth block (second block R2) in arm B.
Induction death (ID)
An induction death is a treatment- and/or disease-related death that occurs during induction prior to
the confirmation of a CR.
Treatment-related death (TRD)
A treatment-related death is a death with a temporal and/or causal relationship to treatment that occurs
during continuous CR.
specific evaluation of the efficacy of chemotherapy, patients with SCT are censored at the time of
SCT. To evaluate SCT separate Kaplan-Meier analyses and survival calculations starting with the time
point of SCT are planned.
The comparison of the therapeutic efficacy of protocol II-IDA (arm A) vs. the R blocks (arm B) is
possible based on the MRD results that were obtained prior to and during corresponding treatment
periods in both arms at corresponding time points. Since the vast majority of patients have already
achieved a complete remission following block F2, only highly sensitive semi-quantitative molecular
tests will allow the detection of a further reduction of leukemic cells in the bone marrow following
morphologic remission.
The feasibility of the protocol will be assessed using the ratio of actually administered therapy vs.
therapy specified by the protocol according to the treatment schedule. Treatment-related morbidity
will be recorded using modified WHO criteria and the toxicity data collection form enclosed in the
appendix. Treatment-related deaths will be evaluated by individual analysis of autopsy reports and
medical summaries.
if the pEFS is smaller than in the precursor study ALL-REZ BFM 96 with p<0.01; if p<0.05, the
data safety monitoring committee is assembled
if during the course of the study a treatment concept becomes known that is clearly superior to
relapse study ALL-REZ BFM 2002
Randomization will be stopped in favor of the prognostically favorable arm if the pEFS differs
between both randomization arms A and B in all strategic groups at a p<0.05 or within one strategic
group at a p<0.01. The data safety monitoring board is notified at a p<0.10.
ALL-REZ BFM 2002 80 Protocol version: 25.06.2003
data registry personnel that are responsible for the correct and complete documentation of the study
data. These research assistants will receive regular training sessions during which study-specific issues
are discussed. This provides assistance to the local study physicians regarding the time-consuming
documentation and ensures the complete and prompt documentation of data.
ALL-REZ BFM 2002 82 Protocol version: 25.06.2003
12 ETHICS
apply. By definition, this study is a treatment optimization study. All drugs used are either approved
for the respective indication in children or can be designated as standard medication because they have
been used in numerous studies for the respective indication in children. Beyond the liability insurance
of the treating center a proband insurance was considered necessary by the ethics review board and,
therefore, has been arranged for all patients treated in Germany.
The study is funded by the ‘Deutsche Kinderkrebsstiftung’[German Childhood Cancer Foundation].
The study protocol must be followed exactly. Any deviation from the specified diagnostic/therapeutic
measures and time points, for which the local study physician is responsible, has to be documented
and justified (e.g. emergency measures).
13 REFERENCES
1. Bader, P., Beck, J., Frey, A., Schlegel, P.G., Hebarth, H., Handgretinger, R., Einsele, H.,
Niemeyer, C., Benda, N., Faul, C., Kanz, L., Niethammer, D. & Klingebiel, T. (1998). Serial
and quantitative analysis of mixed hematopoietic chimerism by PCR in patients with acute
leukemias allows the prediction of relapse after allogeneic BMT. Bone Marrow Transplant, 21,
487-95.
2. Bader, P., Klingebiel, T., Schaudt, A., Theurer-Mainka, U., Handgretinger, R., Lang, P.,
Niethammer, D. & Beck, J.F. (1999). Prevention of relapse in pediatric patients with acute
leukemias and MDS after allogeneic SCT by early immunotherapy initiated on the basis of
increasing mixed chimerism: a single center experience of 12 children. Leukemia, 13, 2079-86.
3. Bader, P., Stoll, K., Huber, S., Geiselhart, A., Handgretinger, R., Niemeyer, C., Einsele, H.,
Schlegel, P.G., Niethammer, D., Beck, J. & Klingebiel, T. (2000). Characterization of lineage-
specific chimaerism in patients with acute leukaemia and myelodysplastic syndrome after
allogeneic stem cell transplantation before and after relapse. Br J Haematol, 108, 761-8.
4. Balis, F.M., Holcenberg, J.S., Poplack, D.G., Ge, J., Sather, H.N., Murphy, R.F., Ames, M.M.,
Waskerwitz, M.J., Tubergen, D.G., Zimm, S., Gilchrist, G.S. & Bleyer, W.A. (1998).
Pharmacokinetics and pharmacodynamics of oral methotrexate and mercaptopurine in children
with lower risk acute lymphoblastic leukemia: a joint children's cancer group and pediatric
oncology branch study. Blood, 92, 3569-77.
5. Bellott, R., Auvrignon, A., Leblanc, T., Perel, Y., Gandemer, V., Bertrand, Y., Mechinaud, F.,
Bellenger, P., Vernois, J., Leverger, G., Baruchel, A. & Robert, J. (2001). Pharmacokinetics of
liposomal daunorubicin (DaunoXome) during a phase I- II study in children with relapsed acute
lymphoblastic leukaemia. Cancer Chemother Pharmacol, 47, 15-21.
6. Berman, E. (1993). A review of idarubicin in acute leukemia. Oncology (Huntingt), 7, 91-8, 104;
discussion 104-7.
7. Bertelli, G. (1995). Prevention and management of extravasation of cytotoxic drugs. Drug Saf,
12, 245-55.
8. Bertelli, G., Gozza, A., Forno, G.B., Vidili, M.G., Silvestro, S., Venturini, M., Del Mastro, L.,
Garrone, O., Rosso, R. & Dini, D. (1995). Topical dimethylsulfoxide for the prevention of soft
tissue injury after extravasation of vesicant cytotoxic drugs: a prospective clinical study. J Clin
Oncol, 13, 2851-5.
9. Beyermann, B., Adams, H.P. & Henze, G. (1997). Philadelphia chromosome in relapsed
childhood acute lymphoblastic leukemia: A matched-pair analysis. Berlin-Frankfurt-Munster
Study Group. J Clin Oncol, 15, 2231-7.
10. Biondi, A., Valsecchi, M.G., Seriu, T., D'Aniello, E., Willemse, M.J., Fasching, K., Pannunzio,
A., Gadner, H., Schrappe, M., Kamps, W.A., Bartram, C.R., van Dongen, J.J. & Panzer-
Grumayer, E.R. (2000). Molecular detection of minimal residual disease is a strong predictive
factor of relapse in childhood B-lineage acute lymphoblastic leukemia with medium risk
features. A case control study of the International BFM study group. Leukemia, 14, 1939-43.
11. Borgmann, A., Baumgarten, E., Schmid, H., Dopfer, R., Ebell, W., Gobel, U., Niethammer, D.,
Gadner, H. & Henze, G. (1997). Allogeneic bone marrow transplantation for a subset of children
with acute lymphoblastic leukemia in third remission: A conceivable alternative? Bone Marrow
Transplant, 20, 939-44.
12. Borgmann, A., Hartmann, R., Schmid, H., Klingebiel, T., Ebell, W., Gobel, U., Peters, C.,
Gadner, H. & Henze, G. (1995a). Isolated extramedullary relapse in children with acute
lymphoblastic
13. leukemia: A comparison between treatment results of chemotherapy and bone marrow
transplantation. Bone Marrow Transplant, 15, 515-21.
ALL-REZ BFM 2002 85 Protocol version: 25.06.2003
14. Borgmann, A., Schmid, H., Hartmann, R., Baumgarten, E., Hermann, K., Klingebiel, T., Ebell,
W., Zintl, F., Gadner, H. & Henze, G. (1995b). Autologous bone-marrow transplants compared
with chemotherapy for children with acute lymphoblastic leukaemia in a second remission: A
matched-pair analysis. The Berlin-Frankfurt-Munster Study Group. Lancet, 346, 873-6.
15. Buchanan, G.R. (1990). Diagnosis and management of relapse in acute lymphoblastic leukemia.
Hematol Oncol Clin North Am, 4, 971-95.
16. Buchanan, G.R., Boyett, J.M., Pollock, B.H., Smith, S.D., Yanofsky, R.A., Ghim, T., Wharam,
M.D., Crist, W.M., Vietti, T.J., Johnson, W. & et al. (1991). Improved treatment results in boys
with overt testicular relapse during or shortly after initial therapy for acute lymphoblastic
leukemia. A Pediatric Oncology Group study. Cancer, 68, 48-55.
17. Buchanan, G.R., Rivera, G.K., Pollock, B.H., Boyett, J.M., Chauvenet, A.R., Wagner, H.,
Maybee, D.A., Crist, W.M. & Pinkel, D. (2000). Alternating drug pairs with or without periodic
reinduction in children with acute lymphoblastic leukemia in second bone marrow remission: A
Pediatric Oncology Group study. Cancer, 88, 1166-74.
18. Bührer, C., Hartmann, R., Fengler, R., Dopfer, R., Gadner, H., Gerein, V., Gobel, U., Reiter, A.,
Ritter, J. & Henze, G. (1993). Superior prognosis in combined compared to isolated bone
marrow relapses in salvage therapy of childhood acute lymphoblastic leukemia. Med Pediatr
Oncol, 21, 470-6.
19. Bührer, C., Hartmann, R., Fengler, R., Rath, B., Schrappe, M., Janka-Schaub, G. & Henze, G.
(1996). Peripheral blast counts at diagnosis of late isolated bone marrow relapse of childhood
acute lymphoblastic leukemia predict response to salvage chemotherapy and outcome. J Clin
Oncol, 14, 2812-7.
20. Bührer, C., Hartmann, R., Fengler, R., Schober, S., Arlt, I., Loewke, M. & Henze, G. (1994).
Importance of effective central nervous system therapy in isolated bone marrow relapse of
childhood acute lymphoblastic leukemia. Blood, 83, 3468-72.
21. Creutzig, U. & Schellong, G. (1980). Treatment of relapse in acute lymphoblastic leukaemia of
childhood. Dtsch Med Wochenschr, 105, 1109-12.
22. Dopfer, R., Henze, G., Bender-Götze, C., Ebell, W., Ehninger, G., Friedrich, W., Gadner, H.,
Klingebiel, T., Peters, C., Riehm, H. & et al. (1991). Allogeneic bone marrow transplantation for
childhood acute lymphoblastic leukemia in second remission after intensive primary and relapse
therapy according to the BFM- and CoALL-protocols: Results of the German Cooperative
Study. Blood, 78, 2780-4.
23. Dörffel, W., Hartmann, R., Schober, S., Veerman, A.J., Pieters, R., Klumper, E. & Henze, G.
(1993). Drug resistance testing as a basis for tailored therapy in children with refractory or
relapsed acute lymphoblastic leukemia. In Drug resistance in leukemia and lymphoma, Kaspers,
G.J., Pieters, R., Twentyman, P.R., Weisenthal, L.M. & Veerman, A.J. (eds) pp. 353-7.
Harwood Academic Publishers: Chur.
24. Eckert, C., Biondi, A., Seeger, K., Cazzaniga, G., Hartmann, R., Beyermann, B., Pogodda, M.,
Proba, J. & Henze, G. (2001). Prognostic value of minimal residual disease in relapsed
childhood acute lymphoblastic leukaemia. Lancet, 358, 1239-41.
25. Erb, N., Harms, D.O. & Janka-Schaub, G. (1998). Pharmacokinetics and metabolism of
thiopurines in children with acute lymphoblastic leukemia receiving 6-thioguanine versus 6-
mercaptopurine. Cancer Chemother Pharmacol, 42, 266-72.
26. Feig, S.A., Ames, M.M., Sather, H.N., Steinherz, L., Reid, J.M., Trigg, M., Pendergrass, T.W.,
Warkentin, P., Gerber, M., Leonard, M., Bleyer, W.A. & Harris, R.E. (1996). Comparison of
idarubicin to daunomycin in a randomized multidrug treatment of childhood acute lymphoblastic
leukemia at first bone marrow relapse: A report from the Children's Cancer Group. Med Pediatr
Oncol, 27, 505-14.
27. Fleischhack, G., Hasan, C., Graf, N., Mann, G. & Bode, U. (1998). IDA-FLAG (idarubicin,
fludarabine, cytarabine, G-CSF), an effective remission-induction therapy for poor-prognosis
ALL-REZ BFM 2002 86 Protocol version: 25.06.2003
43. McCarthy, A.J., Pitcher, L.A., Hann, I.M. & Oakhill, A. (1999). FLAG (fludarabine, high-dose
cytarabine, and G-CSF) for refractory and high-risk relapsed acute leukemia in children. Med
Pediatr Oncol, 32, 411-5.
44. Messina, C., Valsecchi, M.G., Arico, M., Locatelli, F., Rossetti, F., Rondelli, R., Cesaro, S.,
Uderzo, C., Conter, V., Pession, A., Sotti, G., Loiacono, G., Santoro, N., Miniero, R., Dini, G.,
Favre, C., Meloni, G., Testi, A.M., Werner, B., Silvestri, D., Arrighini, A., Varotto, S., Pillon,
M., Basso, G., Zanesco, L. & et al. (1998). Autologous bone marrow transplantation for
treatment of isolated central nervous system relapse of childhood acute lymphoblastic leukemia.
AIEOP/FONOP-TMO Group. Associzione Italiana Emato-Oncologia Pediatrica. Bone Marrow
Transplant, 21, 9-14.
45. Muller, H.J., Beier, R., Loning, L., Blutters-Sawatzki, R., Dorffel, W., Maass, E., Muller-
Weihrich, S., Scheel-Walter, H.G., Scherer, F., Stahnke, K., Schrappe, M., Horn, A.,
Lumkemann, K. & Boos, J. (2001). Pharmacokinetics of native Escherichia coli asparaginase
(Asparaginase medac) and hypersensitivity reactions in ALL-BFM 95 reinduction treatment. Br
J Haematol, 114, 794-799.
46. Muller, H.J., Loning, L., Horn, A., Schwabe, D., Gunkel, M., Schrappe, M., von Schutz, V.,
Henze, G., Casimiro da Palma, J., Ritter, J., Pinheiro, J.P., Winkelhorst, M. & Boos, J. (2000).
Pegylated asparaginase (Oncaspar) in children with ALL: drug monitoring in reinduction
according to the ALL/NHL-BFM 95 protocols. Br J Haematol, 110, 379-84.
47. Nachman, J., Sather, H.N., Gaynon, P.S., Lukens, J.N., Wolff, L. & Trigg, M.E. (1997).
Augmented Berlin-Frankfurt-Munster therapy abrogates the adverse prognostic significance of
slow early response to induction chemotherapy for children and adolescents with acute
lymphoblastic leukemia and unfavorable presenting features: a report from the Children's Cancer
Group. J Clin Oncol, 15, 2222-30.
48. Neuendank, A., Hartmann, R., Bührer, C., Winterhalter, B., Klumper, E., Veerman, A.J. &
Henze, G. (1997). Acute toxicity and effectiveness of idarubicin in childhood acute
lymphoblastic leukemia. Eur J Haematol, 58, 326-32.
49. Oakhill, A., Pamphilon, D.H., Potter, M.N., Steward, C.G., Goodman, S., Green, A., Goulden,
P., Goulden, N.J., Hale, G., Waldmann, H. & Cornish, J.M. (1996). Unrelated donor bone
marrow transplantation for children with relapsed acute lymphoblastic leukaemia in second
complete remission. Br J Haematol, 94, 574-8.
50. Pui, C.H., Bowman, W.P., Ochs, J., Dodge, R.K. & Rivera, G.K. (1988). Cyclic combination
chemotherapy for acute lymphoblastic leukemia recurring after elective cessation of therapy.
Med Pediatr Oncol, 16, 21-6.
51. Reid, J.M., Pendergrass, T.W., Krailo, M.D., Hammond, G.D. & Ames, M.M. (1990). Plasma
pharmacokinetics and cerebrospinal fluid concentrations of idarubicin and idarubicinol in
pediatric leukemia patients: a Childrens Cancer Study Group report. Cancer Res, 50, 6525-8.
52. Ribeiro, R.C., Rivera, G.K., Hudson, M., Mulhern, R.K., Hancock, M.L., Kun, L., Mahmoud,
H., Sandlund, J.T., Crist, W.M. & Pui, C.H. (1995). An intensive re-treatment protocol for
children with an isolated CNS relapse of acute lymphoblastic leukemia. J Clin Oncol, 13, 333-8.
53. Riehm, H., Feickert, H.J., Schrappe, M., Henze, G. & Schellong, G. (1987). Therapy results in
five ALL-BFM studies since 1970: implications of risk factors for prognosis. Hamatol
Bluttransfus, 30, 139-46.
54. Ritchey, A.K., Pollock, B.H., Lauer, S.J., Andejeski, Y. & Buchanan, G.R. (1999). Improved
survival of children with isolated CNS relapse of acute lymphoblastic leukemia: A pediatric
oncology group study. J Clin Oncol, 17, 3745-52.
55. Rivera, G.K., Hudson, M.M., Liu, Q., Benaim, E., Ribeiro, R.C., Crist, W.M. & Pui, C.H.
(1996). Effectiveness of intensified rotational combination chemotherapy for late hematologic
relapse of childhood acute lymphoblastic leukemia. Blood, 88, 831-7.
ALL-REZ BFM 2002 88 Protocol version: 25.06.2003
56. Sadowitz, P.D., Smith, S.D., Shuster, J., Wharam, M.D., Buchanan, G.R. & Rivera, G.K. (1993).
Treatment of late bone marrow relapse in children with acute lymphoblastic leukemia: A
Pediatric Oncology Group study. Blood, 81, 602-9.
57. Schindler, T., Bornmann, W., Pellicena, P., Miller, W.T., Clarkson, B. & Kuriyan, J. (2000).
Structural mechanism for STI-571 inhibition of abelson tyrosine kinase. Science, 289, 1938-42.
58. Schmid, H., Baumgarten, E., Ebell, W., Fengler, R. & Henze, G. (1996). Hochdosistherapie mit
autologer Stammzell-Reinfusion, Immunmodulation und Re-Induktion für Kinder mit Rezidiven
einer Hochrisiko-ALL, ALL-REZ BFM. Abteilung Onkologie/Hämatologie, Kinderklinik des
OHC, Charité - Virchow Klinikum der Humboldt-Universität zu Berlin.
59. Schrappe, M., Reiter, A., Zimmermann, M., Harbott, J., Ludwig, W.D., Henze, G., Gadner, H.,
Odenwald, E. & Riehm, H. (2000). Long-term results of four consecutive trials in childhood
ALL performed by the ALL-BFM study group from 1981 to 1995. Berlin-Frankfurt-Munster.
Leukemia, 14, 2205-22.
60. Schroeder, H., Garwicz, S., Kristinsson, J., Siimes, M.A., Wesenberg, F. & Gustafsson, G.
(1995). Outcome after first relapse in children with acute lymphoblastic leukemia: A population-
based study of 315 patients from the Nordic Society of Pediatric Hematology and Oncology
(NOPHO). Med Pediatr Oncol, 25, 372-8.
61. Schroeder, H., Gustafsson, G., Saarinen-Pihkala, U.M., Glomstein, A., Jonmundsson, G.,
Nysom, K., Ringden, O. & Mellander, L. (1999). Allogeneic bone marrow transplantation in
second remission of childhood acute lymphoblastic leukemia: A population-based case control
study from the Nordic countries. Bone Marrow Transplant, 23, 555-60.
62. Seeger, K., Adams, H.P., Buchwald, D., Beyermann, B., Kremens, B., Niemeyer, C., Ritter, J.,
Schwabe, D., Harms, D., Schrappe, M. & Henze, G. (1998). TEL-AML1 fusion transcript in
63. relapsed childhood acute lymphoblastic leukemia. The Berlin-Frankfurt-Munster Study Group.
Blood, 91, 1716-22.
64. Seeger, K., Stackelberg, A.V., Taube, T., Buchwald, D., Korner, G., Suttorp, M., Dorffel, W.,
Tausch, W. & Henze, G. (2001). Relapse of TEL-AML1--positive acute lymphoblastic leukemia
in childhood: a matched-pair analysis. J Clin Oncol, 19, 3188-93.
65. Stackelberg, A., Hartmann, R., Ritter, J., Nuernberger, W., ., Klingebiel, T., Kretschmann, A. &
Henze, G. (1999). Male gender as an independent adverse risk factor for children with isolated
CNS relapse of ALL (abstract). Israeli-German Bi-National Conference: Current Concepts in
Pediatric Hematology-Oncology. January 26-29, Eilat, Israel, Abstr Vol, 21.
66. van Dongen, J.J., Seriu, T., Panzer-Grumayer, E.R., Biondi, A., Pongers-Willemse, M.J., Corral,
L., Stolz, F., Schrappe, M., Masera, G., Kamps, W.A., Gadner, H., van Wering, E.R., Ludwig,
W.D., Basso, G., de Bruijn, M.A., Cazzaniga, G., Hettinger, K., van der Does-van den Berg, A.,
Hop, W.C., Riehm, H. & Bartram, C.R. (1998). Prognostic value of minimal residual disease in
acute lymphoblastic leukaemia in childhood. Lancet, 352, 1731-8.
67. Vieira Pinheiro, J.P., Muller, H.J., Schwabe, D., Gunkel, M., Casimiro da Palma, J., Henze, G.,
von Schutz, V., Winkelhorst, M., Wurthwein, G. & Boos, J. (2001). Drug monitoring of low-
dose PEG-asparaginase (Oncaspar) in children with relapsed acute lymphoblastic leukaemia. Br
J Haematol, 113, 115-9.
68. Villani, F., Galimberti, M., Comazzi, R. & Crippa, F. (1989). Evaluation of cardiac toxicity of
idarubicin (4-demethoxydaunorubicin). Eur J Cancer Clin Oncol, 25, 13-8.
69. Weisberg, E. & Griffin, J.D. (2000). Mechanism of resistance to the ABL tyrosine kinase
inhibitor STI571 in BCR/ABL-transformed hematopoietic cell lines. Blood, 95, 3498-505.
70. Wheeler, K., Richards, S., Bailey, C. & Chessells, J. (1998). Comparison of bone marrow
transplant and chemotherapy for relapsed childhood acute lymphoblastic leukaemia: The MRC
UKALL X experience. Medical Research Council Working Party on Childhood Leukaemia. Br J
Haematol, 101, 94-103.
ALL-REZ BFM 2002 89 Protocol version: 25.06.2003
71. Winick, N.J., Smith, S.D., Shuster, J., Lauer, S., Wharam, M.D., Land, V., Buchanan, G. &
Rivera, G. (1993). Treatment of CNS relapse in children with acute lymphoblastic leukemia: A
Pediatric Oncology Group study. J Clin Oncol, 11, 271-8.
72. Wofford, M.M., Smith, S.D., Shuster, J.J., Johnson, W., Buchanan, G.R., Wharam, M.D.,
Ritchey, A.K., Rosen, D., Haggard, M.E., Golembe, B.L. & et al. (1992). Treatment of occult or
late overt testicular relapse in children with acute lymphoblastic leukemia: A Pediatric Oncology
Group study. J Clin Oncol, 10, 624-30.
73. Wolfrom, C., Hartmann, R., Brühmüller, S., Fengler, R., Reiter, A., Ritter, J. & Henze, G.
(1997). Similar outcome on boys with isolated and combined testicular acute lymphoblastic
leukemia relapse after stratified BFM salvage therapy. Haematol Blood Transfus, 38, 647-651.
ALL-REZ BFM 2002 90 Protocol version 25.06.2003
14 Appendices
ALL-REZ BFM 2002 91 Protocol version 25.06.2003
Appendix1
Disclosure and Consent
The legal guardians have to be informed that they have the choice to decline the protocol therapy
and to decide in favour of treatment alternatives, an alternative treatment arm or to decline any
treatment. One of the previous ALL-REZ BFM protocols may be considered as a treatment
alternative, which achieved a high rate of continuous remissions without an increase of severe
complications when compared to the international literature. Additionally, it has to be disclosed
that treatment alternatives are available during any phase of the disease and that no disadvantage
will arise from such a decision for the affected patient.
The randomization (assignment of treatment by chance) of treatment element protocol II-IDA vs.
the sequence of blocks R2, R1, R2 aims to determine if the modified protocol II can achieve a
higher remission induction rate and a longer event-free survival. The high efficacy and
tolerability of protocol II have already been demonstrated in primary BFM-treatment studies for
acute lymphoblastic leukemia. It is not known, however, if the use of this protocol during relapse
therapy is superior to the administration of R blocks. The legal guardians have to be informed
about this as well as the merit of the scientific question for future therapies before treatment is
started and before the central randomization is perfomed. They have to be informed that they may
decline the randomization and choose together with the treating physician a treatment arm
without further impact on the overall treatment and without disadvantages for patient. Further,
they have to be informed about the diagnostic tests and scientific studies, which are planned
during the treatment, their purpose and any associated risks as well as the option to decline these
investigations at any point in time.
It is recommended to have the legal guardian and – depending on the circumstances - the patient
sign a consent form for treatment of relapse according to protocol ALL-REZ BFM 2002, which
documents the contents of the disclosure. In cases where the signature of the legal gaurdian
and/or patient is not obtained the disclosing physician and a witness must sign a summary of
items that were covered during the disclosure and carefully document the decisoin of the legal
guardian/patient.
Written consent of the legal guardian/patient to the transmission and processing of personal as
well as diagnostic and therapeutic data to the study center, the Childhood Cancer Registry in
Mainz and central diagnostic laboratories is indispensible. If a patient completes the 18th year of
life during the course of the treatment a separate consent for the processing of data has to be
obtained. For the purpose of documentation, analysis and transmission of data to third parties the
study center anonymizes all personal data using a code for electronic patient identification (PID).
A copy of the patient information and consent to treatment and a copy of the consent to the
transmission and processing of personal data have to handed to the patient or his/her legal
guardian.
ALL-REZ BFM 2002 94 Protocol version 25.06.2003
Prinicipal Investigator: Prof. Dr. med. Dr. h.c. G. Henze, Charité, Klinik f. Pädiatrie m.S. Onkologie/Hämatologie, Augustenburger Platz 1, 13353
Berlin
The persons listed below discussed in detail the disease and treatment of patients with a relapse
of acute lymphoblastic leukemia on _____________.
During the disclosure the following topics were addressed (please check).
Diagnosis
Prognosis without appropriate therapy.
Expected prognosis with treatment protocol ALL-REZ BFM 2002.
Expected prognosis with tretament alternatives (e.g. ALL-REZ BFM 87/90/96)
Effects of chemotherapy (elimination of the leukemic cells, restoration of normal bone marrow function;
need for a combination of different successive treatment phases)
Side effects of chemotherapy (nausea, vomiting; temporary hair loss; effects on the bone marrow and the
peripheral blood counts; immunosuppression; occurrence of severe infections, which in rare cases cannot
be controlled; potential damage to organs; potential effect on fertility; risk of development a malignancy
later on; rarely, not controllable life-threatening toxicity.
Mechanism of action of radiation therapy (elimination of leukemic cells e.g. in the central nervous
system and its membranes)
Side effects of radiation therapy (lethargy syndrome after CNS radiation, potential late effects)
ALL-REZ BFM 2002 95 Protocol version 25.06.2003
Aims of the protocol (optimization and standardization of therapy to improve the prognois of children
with relapsed ALL using a risk-adapted treatment concept with chemotherapy, radiation therapy and
possibly stem cell transplantation; randomized comparison of a continuous chemotherapy with therapy
in blocks during consolidation therapy; MRD testing as a basis for the decision regarding stem cell
transplantation in group S2; increased treatment intensity through shorter intervals between blocks in
accordance with protocol guidelines; standardization of induction therapy and mandatory stem cell
transplantation in the high risk groups S3 and S4; standardized use of L-asparaginase with individual
monitoring of pharmacological efficacy during tretament blocks; answer to scientific questions which
may be important for the treatment of future patients)
Randomization (use of either protocol II-IDA or treatment blocks R2-R1-R2 as part of the consolidation
therapy; potential advantages and disadvantages; nature and purpose of randomization; merit of the
scientific question.
Potential primary or secondary indication for transplantation (information about the associated
transplantation protocol with separate disclosure and consent)
Scientific companion studies (research into the molecular, genetic, immunologic and other directly
related features of acute lymphoblastic leukemia; use of leukemic cells collected during protocol ALL-
REZ BFM 2002; storage of left over cells and use only after approval by the ethics board after
consideration of the potential individual benefit for the patient and only after anonymization)
Current scientific knowledge
Transmission of patient-related data
Insurance coverage of the patient by liabililty insurance and proband insurance
Freedom of choice for the patient
Topics that are not marked were not covered.
The patient and/or his/her legal guardians have decided
O in favour of participation and treatment according to protocol ALL-REZ BFM 2002
against participation and treatment according to protocol ALL-REZ BFM 2002
in favour of randomization
O against randomization
O in favour of participation in scientific companion studies after approval by the
ethics board and consideration of the benefit for the child
O In favour of participation in scientific companion studies only after additional
information and consent
O against participation in scientific companion studies
I hereby declare that I have informed the legal guardian(s)/patient identifed above in detail about
the nature, meaning, impact and risks of treatment protocol ALL-REZ BFM 2002 and that I have
handed to them/him/her a copy of the patient information form and consent to treatment form.
Stamp of the
Hospital
ALL-REZ BFM 2002 96 Protocol version 25.06.2003
Principal Investigator: Prof. Dr. med. Dr. h.c. G. Henze, Charité, Klinik f. Pädiatrie m.S. Onkologie/Hämatologie, Augustenburger Platz 1, 13353
Berlin
The persons listed below discussed in detail the disease and treatment of patients with a relapse
of acute lymphoblastic leukemia on ______________.
You hearby confirm that today you have been informed in detail by the physician named above
about your/your child’s disease and about your/your child’s planned treatment. The disease is
acute lympoblastic leukemia which has recurred (relapse). You have been informed that this
disease cannot be controlled without appropriate treatment. You have been informed about the
probability of success that is expected after treatment with the proposed protocol as well as with
other previously tested treatments.
In particular, the following aspects of treatment have been explained to you:
As part of the treatment protocol ALL-REZ BFM 2002 treatment is given according to a plan that
is used in approximately 100 hospitals in Germany, Austria and Switzerland. More than 100
patients up to 18 years of age will be treated each year according to this plan so that a total of
approximately 450 patients will participate in this study. The experience gained in the preceding
studies was used in the planning of the overall treatment. The main goal of this protocol is to
improve the chance of cure by adjusting the treatment to the variable risk of suffering another
relapse. In addition, novel insights into the disease and its treatment will be gathered (treatment
optimization study). The treatment plan, the anticipated duration of treatment according to
ALL-REZ BFM 2002 97 Protocol version 25.06.2003
protocol ALL-REZ BFM 2002 and the difference to other treatment concepts and previous
treatment protocols were explained to you.
The current concept uses a combination of medications (chemotherapy) and radiation therapy
and/or stem cell transplantation. The chemotherapy consists of successive treatment blocks and
phases during which various medications (cytotoxic drugs) are combined to eliminate the
leukemic cells. The combination of several medications, the additional use of radiation therapy
and - if needed - of stem cell transplantation is aimed at preventing any leukemic cell from
escaping treatment.
In this treatment plan the effectiveness of a prolonged chemotherapy block (protocol II-IDA) will
be evaluated, which has been used in a similar form for many years in the treatment of new cases
of ALL. Since it is not known if it is better to use a prolonged treatment block or three short
blocks during the treatment of relapse it will be determined randomly if protocol II-IDA or the
blocks R2-R1-R2 will be used. This process of random selection is called randomization. Using
randomization one can later determine if the prolonged block or the three short blocks achieve a
better prognosis. You have been informed about the possibilty to participate in the
randomization. If you decline the randomization there will be no disadvantages for you/your
child.
The side effects and risks of chemotherapy have been explained to you in detail. The following
issues have been mentioned: the occurrence of nausea, vomiting, temporary hair loss, effects on
the mucosal membranes, blood cell formation in the bone marrow and blood counts, the high risk
due to potentially life-threatening infections, possible late effects such damage to organs,
potential impairment of fertility, the need for contraception and the risk of developing other
malignancies later on. It was mentioned that side effects may not always be controllable and may
even be fatal.
The purpose of radiation therapy - the elimination of leukemic cells in the central nervous system
and its membranes or in the testes - as well as potential side effects and late effects have been
explained to you. A detailed explanation through the radiation therapist will occur prior to this
treatment. You have been informed about the possibilty to participate in studies aimed at the
recognition of late effects as part of a radiation therapy study.
You have been informed that the treatment plan includes a stem cell transplantion if the chance
for a cure is unfavorable. This can often already be determined at the time the relapse is
diagnosed. It is, however, also possible that it becomes apparent only during the treatment that
chemotherapy is not effective enough and that leukemic cells remain the bone marrow (minimal
residual disease, MRD). In this case stem cell transplantion should be planned. Special highly
sensitive laboratory tests are used to detect the remaining leukemic cells. Bone marrow will be
collected for these tests at various time points under local or a brief general anesthesia.
The possibility of a stem cell transplantion and its role as a part of the treatment plan have been
discussed with you in detail taking into account your/your child’s special situation. If this
treatment becomes necessary you will receive detailed information about the specific risks and
the procedure from the treating physcian ahead of time and on a separate occasion.
The tests for the detection of remaining leukemic cells in the bone marrow (MRD) at the time the
relapse is diagnosed and during the treatment are used to determine the response to treatment.
The test results will be used to determine the appropriate treatment and the appropriate treatment
strategy within the overall protocol. The treatment should be intensive enough to eliminate all
leukemic cells. At the same time the side effects of the treatment should be kept to a minimum.
ALL-REZ BFM 2002 98 Protocol version 25.06.2003
The MRD test uses specific markers of the leukemic cells that are determined with molecular and
immunologic laboratory methods. Compared to conventional methods leukemic cells can be
detected with a up to 10,000-fold higher sensitivity. In children with a first diagnosis of acute
lymphoblastic leukemia these test results have been used for the planning of treatment since
1999.
During the relapse protocol ALL REZ BFM 2002 the MRD tests will be performed at the start of
treatment and at different time points during the course of treatment. The result after the second
chemotherapy block is used to decide if further chemotherapy or stem cell transplantation is
necessary. MRD results from all other time points will not be used to change treatment. They
will be analyzed scientifically, however, to investigate and confirm the success of the treatment.
You have been informed that bone marrow aspirates and drawing of blood for the MRD test is
scheduled at the time of the diagnosis of the relapse and up to six times during the course of
chemotherapy for you/your child. The first aspirates until the achievement of a remission are
necessary to establish the diagnosis and to assess the course of the disease with conventional
methods (microscopy). Samples for the MRD test are collected at the same time. After stem cell
transplantation it is recommended to perform additional bone marrow aspirates and MRD
measurements in your/your child’s case.
The potential risks and complications of bone marrow aspirates and the drawing of blood have
been explained to you. You have been informed that to date there have been no reports of severe
or regularly occurring complications after bone marrow aspirates and the drawing of blood for
MRD tests. In rare cases, bleeding and local infections at the aspiration site may occur. The bone
marrow aspirate is performed under local or general anesthesia. Information about general
anesthesia which typically lasts ten minutes will be provided by the anesthetist.
You have been informed that your child's leukemic cells may used for research studies into the
molecular, genetic, immunologic and other immediately disease-related features of acute
lymphoblastic leukemia and may be potentially used for the development of novel treatment
approaches. No additional bone marrow aspirate or phlebotomy is necessary for this purpose.
Only cells that were collected during the diagnostic tests for protocol ALL-REZ BFM 2002 will
be used. Leftover cells will be frozen if they are available. The cells will be used anonymously
after approval by the local research ethics board or after additional information has been provided
to you and your consent has been obtained. They will only be used if a direct benefit for your
child is possible from these investigations.
During the participation in protocol ALL-REZ BFM 2002 the liability insurance of the
participating clinic/center, in which the treatment and the investigations take place, assumes
liability for adverse health effects that are due to negligence. Adverse health effects that are
directly related to the study question are additionally insured by a proband insurance (Gothaer
Versicherung, Probandenversicherungsnummer 37.907.546060, Gothaer Allee 1 50969 Köln). If
you or your child observe an adverse health effect during or after treatment, which may be related
to the participation in the treatment protocol mentioned above, you are obligated to contact the
treating physician promptly.
Declaration and consent:
I consent to my/my child’s treatment according to protocol ALL-REZ BFM 2002.
I have been informed that my consent to this treatment is voluntary, that I may decline
consent to this treatment or to specific treatment elements, that I may revoke my consent at
any time and that I have the right to chose a different treatment or decline any therapy.
ALL-REZ BFM 2002 99 Protocol version 25.06.2003
I agree that one of two treatment elements specified in the protocol (a longer block or three
short blocks) will be selected randomly at the study center (central randomization) and that
this information will be forwarded to the treating physician. I have been informed that the
participation in the randomization is voluntary and that I have the right to decline the
treatment according to a performed randomization and to select one of the two treatment
elements on my own.
I agree to partipate in the MRD and scientific companion studies that are part of the
treatment protocol. The participation in these studies is voluntary. Additional scientific
studies with remaining samples will only be performed after approval by the research
ethics board after consideration of the benefit for my child or only after I receive additional
information and provide consent. I know that I can decline all or individual tests at any
time without giving reasons.
You have the right to delete single words, sentences or paragraphs of this consent form and to
change them if they do not apply to you/your child or if you do not agree.
You consider yourself sufficiently informed and have had sufficient opportunity to ask questions
of the physician mentioned above. You have understood the information provided herein and
received a copy of the consent form.
Treating center:
(letterhead/stamp)
personal data (name, date of birth, address, diagnosis and results of diagnostic tests, treatment
and disease course) are transmitted to the study center of treatment protocol ALL-REZ BFM
2002 and processed as part of and according to the purpose of this protocol. Regarding the
transmission of the data as descibed above I hereby release the treating physician from his/her
obligation to patient confidentiality.
The processing of data (storage, transfer, modification, deletion) by the study center serves the
purpose of medical documentation to improve the diagnosis, confirm laboratory and clinical test
results and monitor treatment in the individual treating centers. This documentation is an
important aid of contemporary treatment and indispensable for the optimal implementation and
coordination of treatment as well as the assesment of treatment success of this protocol. For the
purpose of central documentation, analysis or transmission of results to third parties the study
center anonymizes all personal data using a code for the electronic patient identification (PID).
These data will be transmitted if necessary to the following recipients:
ALL-REZ BFM-Study Center (Director: Prof. Dr. G. Henze), Klinik für Pediatric Hematology/Oncology,
Charité, Augustenburger Platz 1, 13353 Berlin
ALL-BFM-Study Center (Director: Prof. Dr. M. Schrappe), Pediatric Hematology/Oncology,
Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, 30625 Hannover
CoALL-Study Center (Director: Prof. Dr. G. Janka-Schaub), Pediatric Hematology/Oncology, Universi-
tätsklinikum Hamburg, Martinistr. 52, 20246 Hamburg
Kinderkrebsregister (Dr. P. Kaatsch), Insitute for Medical Science and Documentation, Project Group
Pediatric Oncology, Johannes-Gutenberg Universität, Langenbeckstr. 1, 55101 Mainz
Immunology Laboratry (Director: Prof. Dr. W.-D. Ludwig), Medizinische Onkologie / Molekularbiologie,
Max-Delbrück-Centrum, Lindenberger Weg 80, 13122 Berlin-Buch
Molecular Genetic Laboratory (Director: Dr. Dr. K. Seeger), Klinik für Pädiatrie m. S.
Onkologie/Hämatologie, Charité, Augustenburger Platz 1, 13353 Berlin
Cytostatic Laboratory (Director: Prof. Dr. B. Dörken), Med. Klinik m. S. Hämatologie, Onkologie und
Tumorimmunologie, Charité, Robert-Rössle-Str. 10, 13092 Berlin-Buch
Pharmacologial Laboratory Pediatric Oncology (Director Prof. Dr. J. Boos), Klinik und Poliklinik für
Kinderheilkunde, Westfälische Wilhelms-Universität, Albert-Schweitzer-Str. 33, 48149 Münster
ALL-REZ BFM 2002 101 Protocol version 25.06.2003
Radiological Late-Effect Study (Director: Prof. Dr. N. Willich), Klinik und Poliklinik für
Strahlentherapie, Westfälische Wilhelms-Universität, Albert-Schweitzer-Str. 33, 48149 Münster
In case of stem cell transplantation the data will be transmitted to
Pediatric Stem Cell Transplant Registry (Director: Prof. Dr. T. Klingebiel), Pädiatrische Hämato-
logie/Onkologie, Johann-Wolfgang-Goethe Universität, Theodor-Stern-Kai 7, 60590 Frankfurt
The coordinating and transplanting center, at which the transplantation will be performed (center and
address will be forwarded to the patient/legal guardian by the treating physician)
MRD-Lab (Director: PD Dr. P. Bader), Allg. Pädiatrie Hämatologie/Onkologie, Universitätsklinikum
Tübingen, Hoppe-Seyler.Str. 1, 72076 Tübingen
Any person that has access to the data is obliged to protect the privacy of these data and to
conform with pertinent legislation according to EU guidelines for data protection, federal law for
data protection and applicable state laws.
I agree to the transmission and processing of personal data as well as data describing the
disease, treatment and diagnostic tests to the extent described above and for the exclusive
purposes described above.
In addition, I agree to the collection, isolation, transfer and analysis as well as the anonymized
storage of samples of blood, tissue and genetic material potentially derived from this material that
was collected as part of the treatment protocol by the treating physician and the laboratories
mentioned above. All samples will be anonymized and stored indefinitely after the analysis or
will be destroyed upon your request.
Patient-related data, test results and other medical data will be stored for a time period of at least
10 years and then destroyed. Data used as part of the medical record will be stored for 30 years.
You may at any time decline the processing of the your/your child's data, request information
about your/your child's data and ask for the correction of data. The results of the treatment
protocol and investigations will be published in scientific journals without information that will
allow your/your child’s identificaton.
Consent to the processing of data is voluntary and can be revoked at anytime without
disadvantages for the patient. In this case all stored personal data and the corresponding code will
be deleted as far as this is not prevented by legal or professional rules for data storage.
APPENDIX 2
Block F1 .................................................................................................................................................103
Block F2 .................................................................................................................................................104
Block R2.................................................................................................................................................105
Block R1.................................................................................................................................................106
Protocol II-IDA........................................................................................................................................107
Order sets
Block F1 .................................................................................................................................................109
Block F2 .................................................................................................................................................110
Block R2.................................................................................................................................................111
Block R1.................................................................................................................................................112
Block F1
Please fill in this form and the end of the block and send it to the ALL-REZ BFM study center in Berlin
(Prof. Dr. med. Dr. h.c. G. Henze, Charité, Klinik f. Paediatrie m.S. Haematologie/Onkologie,
Augustenburger Platz 1, 13353 Berlin).
2
dexamethasone 20 mg/m /d p.o. mg DEXA
2
vincristine * 1.5 mg/m /d i.v. mg VCR
2
methotrexate 1 g/m 36 h infusion g MTX
2
E.coli L- Asp. **10,000 U/m 6 h infusion U Coli-ASP
day 1 2 3 4 5 6
Please consult the specific instructions regarding administration of medications as well as the protocol
guidelines.
Block F2
Please fill in this form and the end of the block and send it to the ALL-REZ BFM study center in Berlin
(Prof. Dr. med. Dr. h.c. G. Henze, Charité, Klinik f. Paediatrie m.S. Haematologie/Onkologie,
Augustenburger Platz 1, 13353 Berlin).
2
dexamethasone 20 mg/m /d p.o. mg DEXA
2
vincristine* 1.5 mg/m i.v. mg VCR
2
cytarabine 2x3 g/m /d 3 h infusion g ARA-C
2
E. coli L- asp.** 10,000 U/m 6 h infusion U Coli-ASP
day 1 2 3 4 5
Please consult the specific instructions regarding administration of medications as well as the protocol
guidelines.
Block R2
Please fill in this form and the end of the block and send it to the ALL-REZ BFM study center in Berlin
(Prof. Dr. med. Dr. h.c. G. Henze, Charité, Klinik f. Paediatrie m.S. Haematologie/Onkologie,
Augustenburger Platz 1, 13353 Berlin).
2
dexamethasone 20 mg/m /d p.o. mg
2
thioguanine 100 mg/m /d p.o. mg 6-TG
2
vindesine 3 mg/m i.v. mg VDS
2
methotrexate 1 g/m 36 h infusion g MTX
2
ifosfamide 400 mg/m /d 1 h infusion mg IFO
2
daunorubicin 35 mg/m 24 h infusion mg DNR
2
E.coli L- Asp. ** 10,000 U/m 6 h infusion U Coli-ASP
day 1 2 3 4 5 6
Please consult the instructions regarding the administration of medications as well as the protocol
guidelines.
In case of CNS involvement intrathecal chemotherapy is repeated on day 5.
Block R1
Please fill in this form and the end of the block and send it to the ALL-REZ BFM study center in Berlin
(Prof. Dr. med. Dr. h.c. G. Henze, Charité, Klinik f. Paediatrie m.S. Haematologie/Onkologie,
Augustenburger Platz 1, 13353 Berlin).
day 1 2 3 4 5 6
Please consult the instructions regarding the administration of medications and the protocol guidelines.
Protocol II-IDA
Please fill in this form at the end of the block and send it to the ALL-REZ BFM study center in Berlin (Prof. Dr. med. Dr. h.c. G. Henze, Charité, Klinik f. Pädiatrie
m.S. Onkologie/Hämatologie, Augustenburger Platz 1, 13353 Berlin).
body surface area [m2] ____,_________ start of the block: _____________________ end of the block: _______________
2
dexamethasone 6 mg/m /d p.o. ________ mg DEXA
2
vincristine* 1.5 mg/m /d i.v. ________ mg VCR
2
idarubicin 6 mg/m /d 6 h infusion ________ mg IDA
2
E.coli L–Asp. ** 10,000 U/m 6 h infusion ________ U Coli-ASP
2
cyclophosphamide 1 g/m /d 1 h infusion ________ g CPM
2
cytarabine 75 mg/m /d i.v. ________ mg ARA-C
2
thioguanine 60 mg/m /d p.o. ________ mg 6-TG
day 1 8 15 22 29 36 43
Please consult the instructions regarding the administration of medications as well as the protocol guidelines.
In case of CNS involvement additional intrathecal chemotherapy is given on day 8.
* The maximal dose of vincristine is 2 mg.
** In case of an allergic reaction or silent inactivation chose an alternative preparation according to the protocol guidelines.
ALL-REZ BFM 2002 108 Protocol version 25.06.2003
Order Sets
The order sets and worksheets printed below are meant to aid the practical administration of therapy. They, of
course, cannot take into account the particular additions of each hospital and are merely meant as general
framework that requires individual modification. The heparinization of all infused solutions (400 U/L), which is
customary at our center, for example, is therefore deliberately not included.
ALL-REZ BFM 2002 109 Protocol version 25.06.2003
Block F1
patient: date of birth: _________
Triple intrathecal chemotherapy day 1: approx. 1 hour after the start of MTX
Block F2
patient: date of birth: _________
Block R2
patient: date of birth: _________
Triple intrathecal chemotherapy day 1 (in case of CNS involvement also on day 5)
approx.1 hour after the start of MTX
Age MTX ARA-C PRED MTX ________ mg
< 1 year 6 16 4 mg simultan.
1 year 8 20 6 mg intra- ARA-C________ mg
2 years 10 26 8 mg thecal
>=3 years 12 30 10 mg PRED ________ mg
IFO 400 mg/m2 (see infusion orders ifosfamide during block R2)
Block R1
patient: date of birth: _________
Triple intrathecal chemotherapy day 1: approx.1 hour after the start of MTX
* for a peripheral venous access the specified concentration should not be exceeded; the parallel
infusion of normal saline has to be reduced accordingly; in case of a central venous access any
concentration may be selected.
Triple intrathecal chemotherapy day 1, 15 (in case of CNS involvement also on day 8)
Leucovorin rescue
Leucovorin 15 mg/m2 i.v. at 48 hours Leucovorin ________ mg
Leucovorin 15 mg/m2 i.v. at 54 hours Leucovorin ________ mg
Parallel infusion start with MTX (hour 0), infuse twice the volume over 48 hours
0.9% NaCl 1500 ml/m2 0.9% NaCl ________ ml
2
+ 5% glucose 1500 ml/m 5% glucose ________ ml
+ KCl 30 mmol/l (glucose + NaCl) KCl ________ mmol
+ Na-Bicarbonate 40 mmol/l (glucose + NaCl) NaHCO3 ________ mmol
Labs: Na, K, Ca, Cl, Mg, total protein, AST, ALT, alk. Phosph., bili, crea
Prior to as well as 24 and 48 hours after the start of the MTX infusion
MTX level prior to as well as 36 hours and 48 hours after the start of the MTX infusion
The MTX level at hour 48 has to be measured immediately and the result has to be
communicated to the physician!
(may change the leucovorin rescue, see appendix)
ARA-C infusion
Vit B6 100 mg/m2 i.v. prior to each ARA-C infusion 4 x Vit B6 ________ mg
Conjunctivitis prophylaxis q6h (eye drops)
Parallel infusion
0.9%NaCl 1000 ml/m2 0.9%NaCl ________ ml
+ 5% glucose 1000 ml/m2 5% glucose ________ ml
+ KCl 30 mmol/l (glucose + NaCl) KCl ________ mmol
infuse twice each time over 24 hours
Antiemetic prophylaxis:
Labs: Na, K, Ca, Cl, Mg, total protein, AST, ALT, alk. Phosph., bili, crea
at the start as well as 24 hours and 48 hours after the start of each ARA-C infusion
ARA-C infusion
Vit B6 100 mg/m2 i.v. prior to each ARA-C infusion 2 x Vit B6 ________ mg
Conjunctivitis prophylaxis q6h (eye drops)
Parallel infusion
0.9%NaCl 1000 ml/m2 0.9%NaCl ________ ml
+ 5% glucose 1000 ml/m2 5% glucose ________ ml
+ KCl 30 mmol/l (glucose + NaCl) KCl ________ mmol
infuse over 24 hours
Antiemetic prophylaxis:
Labs: Na, K, Ca, Cl, Mg, total protein, AST, ALT, alk. Phosph., bili, crea
at the start as well as 24 hours and 48 hours after the start of each ARA-C infusion
Parallel infusion
0.9% NaCl 750 ml/m2 0.9% NaCl ________ ml
+ 5% glucose 750 ml/m2 5% Glucose ________ ml
+ KCl 30 mmol/l (glucose + NaCl) KCl ________ mmol
Infuse on day 3-5 each time over 24 hours; on day 1-2 the parallel infusion to MTX is
sufficient.
Antiemetic prophylaxis:
Labs: Na, K, Ca, Cl, total protein, AST, ALT, alk. Phosph., bili, crea
at the start of each IFO infusion
Labs: AST, ALT, total protein, bili at hour 0; electrolytes, crea at hour 0 and 24
5
75 mg/m²
4
60 mg/m²
3
45 mg/m²
2
30 mg/m²
1
15 mg/m²
0.25 no rescue
42 54 66 78 90
36 48 60 72 84 96
hours after the start
of the MTX infusion
48h 15 mg/m²
54h 15 mg/m²
end of rescue
deviations: MTX36h > 10.0 µmol/L and/or MTX48h > 0.5 µmol/L
determine a MTX level every 6 hours (may include a level at 42 hours) !
Rescue every 6 hours. LVC i.v. until MTX level 0.25 µmol/L
dose: according to the diagram using the MTX level
measured 6 hours earlier (if MTX at 42h > 5.0 µmol/L
use the MTX level at 42h, however).
start: as soon as the MTX level at 48h (or 42h) is available
MTX48h > 2.0 µmol/L: - forced alkaline diuresis at 3 l/m²
MTX48h > 5.0 µmol/l: - carboxypeptidase (see chapter emergencies)
- forced alkaline diuresis 4.5 l/m²
- LCV dose (mg) = weight(kg) x MTX level at 42h (µmol/l)
- additional LCV doses are calculated based on the
methotrexate level measured 6 hour earlier until this level
falls below 5 µmol/L.
ALL-REZ BFM 2002 121 Protocol version 25.06.2003
APPENDIX 3
molecular studies: BCR/ABL yes no; TEL/AML1 yes no; MLL-abnormality yes no
Current relapse
_________________________________________________________________________________
_________________________________________________________________________________
related to relapse
related to treatment complication
related to BMT
related to a second malignancy
_________________________________________________________________________________
reason: ________________________________________
_________________________________________________________________________________
_________________________________________________________________________________
causality:
Is the pre-existing condition of the patient or an unrelated disease responsible for this event ?
Toxicity form
Please document the maximal toxicity that occurred during the entire period from the beginning of the
protocol until 14 days after the end of the protocol and prior to the start of the next treatment block,
respectively.
Grade 0 1 2 3 4
General wellbeing very good good intermediate poor very poor
Hb[g/l] normal for age 100 80 65 < 65
9
WBC [x10 /L] 4 <4 <3 <2 <1
9
Neutrophils [x10 /L] 2 <2 < 1.5 <1 < 0.5
9
Platelets [x10 /L] 100 < 100 < 75 < 50 < 10
Infection none minor moderate severe life-threatening
no organism organism isolated; with hypotension
isolated; on i.v. on i.v. antibiotics
antibiotics
Fever [ C] none < 38 40 > 40 > 40
< 24 hours 24 hours
Nausea none oral intake sufficient decreased intake no oral intake TPN required
arm A
prot. II-IDA
arm B
block R2
block R1
block R2
block R1
block R2
block R1
block R2
block R1
Radiation therapy
name: ____________________ date of birth: __________ strategic group: _____ arm: _____
At diagnosis:
toxicity forms
arm A protocol II-IDA (part 1) _________
arm A protocol II-IDA (part 2) _________
for BMT:
copy of the BMT discharge summary _________
During follow-up
follow-up (yearly)
ALL-REZ BFM 2002 128 Protocol version 25.06.2003
Date
Transaminases + + + + + + + + + + +
Bilirubin + + + + + + + + + + +
Creatinine + + + + + + + + + + +
Blood pressure + + + + + + + + + + +
Height + + + + + + + + + + +
Weight + + + + + + + + + + +
Echocardiogram + + + + + + + + +
EKG + + + + + + + + +
Chest X ray + + + + + + + + +
Karnofsky/Lansky index + + + + + + + +
Learning + + + + + + + +
Skin + + + + + + + + +
Neurologic exam + + + + + + + +
Pulmonary function test + + + + + +
Coagulation + + + + +
Ophthalmologic exam + + + + + + +
Dental exam + + + + + + +
In case of a mild or severe impairment pls. provide diagnostic results and details in
the following (e.g. SF of the echocardiogram, biochemistry etc.):
Comments:
_________________________________________________________________________________
_________________________________________________________________________________
_________________________________________________________________________________
Current medication:_________________________________________________________________
_________________________________________________________________________________
_________________________________________________________________________________
Appendix 4
Requisitions
Immunophenotyping ..............................................................................................................................132
13353 BERLIN
_________________________________________________________________________________
_________________________________________________________________________________
sample:
requested test(s):
diagnosis of relapse
_______________ _________________
date signature
ALL-REZ BFM 2002 132 Protocol version 25.06.2003
Immunology
REQUISITION IMMUNOPHENOTYPING
DNA INDEX (only for ALL BFM-Study)
Sample: ___ml peripheral blood ___ml peripheral blood in EDTA (PNH/ lymphocyte
subsets)
___ml bone marrow ___ml pleural effusion, CSF
lymph node
...................................................... ..............................................................................
place, date of sample collection legible signature and stamp of the requesting
physician
ALL-REZ BFM 2002 133 Protocol version 25.06.2003
1. Samples
Draw bone marrow (at least 2 ml), peripheral blood ( amount depends of WBC or % blasts),
CSF(>300/3 cells/µl), pleural effusion, ascites into a heparinized syringe (50 U heparin/ml
sample).
Send lymph node or tissue biopsies unfixed in culture media (e.g. RPMI or MEM media) or in
a buffered saline solution (e.g. Hanks BSS or PBS), to which 10-15% fetal calf serum have
been added if possible.
PLEASE INCLUDE ONE UNSTAINED SLIDE PER SAMPLE!
For the PNH test (FACS) und the analysis of lymphocyte subsets please send 5-10 ml
peripheral blood in EDTA.
2. Packaging
If possible use plastic containers that cannot break. Do not use natural cork caps for tubes.
Remove needles form syringes and cap syringes well.
3. Shipment
Please send all samples by courier and label as “important diagnostic sample”.
If possible do not ship samples in which cells rapidly lose viability (e.g. lymph node, ascites,
pleural effusion, CSF) during the weekend.
4. Requisition
Please fill in the form completely (name, first name, date of birth of the patient;
clinically/morphologically suspected diagnosis; initial test or relapse; previous immunological
result if known; clinical results; date of sample collection; legible signature of the
physician and address of the sending hospital).
Universitätsklinikum Tübingen
Probennummer:
Probenmenge:
Universitätsklinikum Tübingen
Klinik für Kinderheilkunde und Jugendmedizin
MRD-/ Chimärismuslabor PD Dr. P. Bader
Labor C02 Raum 305
Hoppe-Seyler-Straße 1
72076 Tübingen
Tel.: + 49 (0)7071 29-83809
Fax.: + 49 (0)7071 29-5365 Email: [email protected]
APPENDIX 5
Participating Centers
Prof.Dr.G.Mau Frau OÄ Dr. D. Möbius
OA Dr. Eberl Frau OÄ Dr. E. Holfeld
Stadt.Klinikum-Kinderklinik Carl-Thiem-Klinikum Cottbus
Holwedestr.16 Kinderklinik
38118 BRAUNSCHWEIG Thiemstr. 111
Tel: (0531) 595-1222 (Pforte)-1338 (Stat) O3048 COTTBUS
Fax: (0531) 595-1400 Tel.: (0355) 46-2332 (Stat.)
Fax (0355) 46-2182
OA Dr. H.-J. Spaar
OA Dr. Th. Lieber Prof. Dr. W. Andler
Kliniken d. Freien Hansestadt Bremen Dr. Th. Wiesel
Prof. Hess-Kinderklinik Vestische Kinderklinik
St.-Jürgen-Str. Universität Witten / Herdecke
28205 BREMEN Dr. Friedrich-Steiner-Str. 5
Tel.: (0421) 497-1 (Pforte) -5413 (Stat.) 45711 DATTELN
Fax (0421) 497-3421 Tel.: (02363) 975-506 (Stat.)
Fax (02363) 642-11
Prof. Dr. M. Kirschstein
Allg. Krankenhaus - Kinderabteilung Frau CÄ Dr. C. Niekrens
Siemensplatz 4 Städt. Krankenanstalten
29223 CELLE Kinderklinik
Tel.: (05141) 72-2040 (Stat.) Wildeshauser Str. 92
Fax (05141) 72-2049 (Stat.) 27753 DELMENHORST
Tel.: (04221) 99-4401 (Poli)
OA Dr. K. Hofmann Fax (04221) 99-4405
Frau OÄ Dr. I. Krause
Klinikum Chemnitz gGmbH OA Dr. H. Breu
Klinik für Kinder – u. Jugendmedizin Frau OÄ Dr. H. Olschewski
Flemmingstr. 4 Städt. Kliniken Dortmund - Kinderklinik
O9116 CHEMNITZ Beurhausstr. 40
Tel.: (0371) 3332-4124 (Stat.) 44123 DORTMUND
Fax (0371) 3332-4125 Tel.: (0231) 50-21721 (Stat.)
Fax (0231) 50-20105
OA Dr. R. Frank
Landkrankenhaus / Kinderklinik PD Dr. M. Suttorp
Ketschendorfer Str. 33 Frau Dr. I. Lauterbach
96450 COBURG Universitätsklinikum Carl Gustav Carus
Tel.: (09561) 22-5553(Stat.) Klinik u. Polikl. für Kinderheilkunde
Fax (09561) 22-5552 Fetscherstr. 74
O1307 DRESDEN
Prof. Dr. E. B. Lang Tel.: (0351) 458-2340 (Stat.)
Frau OÄ Dr. R. Siegler Fax (0351) 458-4337
St. Vincenz-Hospital / Kinderabteilung
Südring 41 Frau Dr. V. Scharfe
48653 COESFELD Städt. Krankenhaus Dresden-Neustadt
Tel.: (02541) 89-2022 Kinderklinik
Industriestr. 40
PD Dr. G. Weinmann O1129 DRESDEN
OA Dr. A. Lemmer Tel.: (0351) 856-2550 (Stat.)
Klinikum Erfurt GmbH Fax (0351) 856-2500
Klinik für Kinderheilkunde
Am Schwemmbach 32 A
99012 ERFURT
Tel.: (0361) 781-4603 (Stat.)
Fax (0361) 781-4502
ALL-REZ BFM 2002 139 Protocol version 25.06.2003