Aall1331doc 2
Aall1331doc 2
AALL1331
Risk-Stratified Randomized Phase III Testing of Blinatumomab (IND# 117467, NSC#765986) in First
Relapse of Childhood B-Lymphoblastic Leukemia (B-ALL)
THIS PROTOCOL IS FOR RESEARCH PURPOSES ONLY, AND SHOULD NOT BE COPIED, REDISTRIBUTED OR USED FOR
ANY OTHER PURPOSE. MEDICAL AND SCIENTIFIC INFORMATION CONTAINED WITHIN THIS PROTOCOL IS NOT
INCLUDED TO AUTHORIZE OR FACILITATE THE PRACTICE OF MEDICINE BY ANY PERSON OR ENTITY. RESEARCH
MEANS A SYSTEMATIC INVESTIGATION, INCLUDING RESEARCH DEVELOPMENT, TESTING AND EVALUATION,
DESIGNED TO DEVELOP OR CONTRIBUTE TO GENERALIZABLE KNOWLEDGE. THIS PROTOCOL IS THE RESEARCH
PLAN DEVELOPED BY THE CHILDREN’S ONCOLOGY GROUP TO INVESTIGATE A PARTICULAR STUDY QUESTION OR
SET OF STUDY QUESTIONS AND SHOULD NOT BE USED TO DIRECT THE PRACTICE OF MEDICINE BY ANY PERSON OR
TO PROVIDE INDIVIDUALIZED MEDICAL CARE, TREATMENT, OR ADVICE TO ANY PATIENT OR STUDY SUBJECT. THE
PROCEDURES IN THIS PROTOCOL ARE INTENDED ONLY FOR USE BY CLINICAL ONCOLOGISTS IN CAREFULLY
STRUCTURED SETTINGS, AND MAY NOT PROVE TO BE MORE EFFECTIVE THAN STANDARD TREATMENT. ANY PERSON
WHO REQUIRES MEDICAL CARE IS URGED TO CONSULT WITH HIS OR HER PERSONAL PHYSICIAN OR TREATING PHYSICIAN
OR VISIT THE NEAREST LOCAL HOSPITAL OR HEALTHCARE INSTITUTION.
STUDY CHAIR
Patrick Brown, MD
1650 Orleans Street, CRB1 RM 2M49
Baltimore, MD. 21231
Phone: (410) 614-4915
Fax: (410) 955-8897
E-mail: [email protected]
ABBREVIATIONS
Abbreviation Term
TABLE OF CONTENTS
SECTION PAGE
STUDY COMMITTEE 7
ABSTRACT 10
EXPERIMENTAL DESIGN SCHEMA 11
1.0 GOALS AND OBJECTIVES (SCIENTIFIC AIMS) 12
1.1 Primary Aims 12
1.2 Secondary Aims 12
1.3 Exploratory Aims 12
2.0 BACKGROUND 13
2.1 Overview 13
2.2 Rationale for Selected Approach and Trial Design 13
3.0 STUDY ENROLLMENT PROCEDURES AND PATIENT ELIGIBILITY 22
3.1 Study Enrollment 22
3.2 Patient Eligibility Criteria 27
3.3 Definitions 30
4.0 TREATMENT PLAN 34
4.1 Overview of Treatment Plan 34
4.2 Block 1 (All patients) - 4 Weeks Duration 37
4.3 Block 2 (HR/IR and LR Patients: Post-randomization) – 4 weeks duration 41
4.4 Block 3 (HR/IR Patients on Arm A; LR Patients on Arm C) - 4 weeks 45
4.5 Blinatumomab Block: Cycle 1 (HR/IR Patients in Arm B) - 5 weeks 48
4.6 Blinatumomab Block: Cycle 2 (HR/IR Patients in Arm B) - 5 weeks 51
4.7 Blinatumomab-S: Cycle 1 (Salvage Therapy) - 5 weeks 53
4.8 Blinatumomab-S: Cycle 2 (Salvage Therapy) - 5 weeks 56
4.9 Hematopoietic Stem Cell Transplant 59
4.10 Bridging Maintenance Therapy (All HR/IR Patients, as required) – up to 6 weeks 69
4.11 Continuation 1/Continuation 2 (All LR Patients) - 8 weeks 70
4.12 Maintenance Cycle 1 (All LR patients) – 12 weeks 74
4.13 Maintenance Chemoradiation - 3 weeks 77
4.14 Maintenance-Post Cycle 1 (All LR patients) 78
4.15 Blinatumomab Block: Cycle 1 (Patients in Arm D) - 5 weeks 80
4.16 Blinatumomab Block: Cycle 2/3 (LR Patients in Arm D) - 5 weeks 82
5.0 DOSE MODIFICATIONS FOR TOXICITIES 84
5.1 Asparaginase [Pegaspargase (PEG-Asparaginase) or Erwinia] 84
5.2 Blinatumomab 86
5.3 Cyclophosphamide 89
5.4 Cytarabine (ARAC) 89
5.5 Anthracycline-Mitoxantrone 90
5.6 Etoposide 90
5.7 Intrathecal Methotrexate/Triple Intrathecal Therapy 91
5.8 Intermediate-Dose Methotrexate (ID MTX) and Leucovorin Rescue 92
STUDY COMMITTEE
Sarah Tasian, MD
Children’s Hospital of Philadelphia
Hematology/Oncology
3501 Civic Center Blvd
CTRB, 3rd Floor Room 3100
Philadelphia, PA 19104
Phone: (215) 590-5476
Fax: (215) 590-3770
Email: [email protected]
AGENT NSC# IND#
Hui Zeng, MS Blinatumomab 765986 117467
Children's Oncology Group - Data Center (Gaines) (IND Sponsor: CTEP)
University of Florida Asparaginase E. chrysanthemi 106977 Exempt
6011 NW 1st Place Cytarabine 63878 Exempt
Gainesville FL 32607 Cyclophosphamide 26271 Exempt
Phone: 352 273-0570 Cyclosporine 290193 Exempt
Fax: 352 392-8162 Dexamethasone 34521 Exempt
Email: [email protected] Etoposide 141540 Exempt
Filgrastim 614629 Exempt
COG RESEARCH COORDINATOR Fludarabine 312887 Exempt
Susan Conway, BA, CCRP Hydrocortisone 10483 Exempt
Children's Oncology Group Statistics and Data Center Leucovorin 003590 Exempt
University of Florida Methotrexate 000740 Exempt
6011 NW 1st Place Mercaptopurine 000755 Exempt
Gainesville, FL 32607 Mitoxantrone 301739 Exempt
Phone: 352-273-0559 Mycophenolate Mofetil 724229 Exempt
Fax: 352-392-8162 Pegaspargase 624239 Exempt
E-mail: [email protected] Tacrolimus 717865 Exempt
Thioguanine 00752 Exempt
COG PROTOCOL COORDINATOR Thiotepa 6396 Exempt
Mary Beth Sullivan, BS Vincristine 67574 Exempt
Children’s Oncology Group Operations Center
Study Development Office
222 E. Huntington Dr., Suite 100
Monrovia, CA 91016
Phone: (626) 241-1539
Fax: (626) 445-4334
E-mail: [email protected]
The Children's Oncology Group has received a Certificate of Confidentiality from the federal government,
which will help us protect the privacy of our research subjects. The Certificate protects against the
involuntary release of information about your subjects collected during the course of our covered studies.
The researchers involved in the studies cannot be forced to disclose the identity or any information collected
in the study in any legal proceedings at the federal, state, or local level, regardless of whether they are
criminal, administrative, or legislative proceedings. However, the subject or the researcher may choose to
voluntarily disclose the protected information under certain circumstances. For example, if the subject or
his/her guardian requests the release of information in writing, the Certificate does not protect against that
voluntary disclosure. Furthermore, federal agencies may review our records under limited circumstances,
such as a DHHS request for information for an audit or program evaluation or an FDA request under the
Food, Drug and Cosmetics Act. The Certificate of Confidentiality will not protect against mandatory
disclosure by the researchers of information on suspected child abuse, reportable communicable diseases,
and/or possible threat of harm to self or others.
ABSTRACT
AALL1331 is a group wide risk-stratified, randomized Phase III study to test whether incorporation of
blinatumomab into the treatment of patients with childhood B-Lymphoblastic Leukemia (B-ALL) at first
relapse will improve disease free survival. Blinatumomab is being tested in this population based on its
demonstrated safety profile and single agent activity (induction of MRD-negative remissions in children
with multiple relapsed refractory B-ALL).
AALL1331 risk stratification is determined based on site of relapse [marrow versus isolated extramedullary
(IEM)], time to relapse and minimal residual disease (MRD) status following a uniform first block of
chemotherapy. High risk (HR) and intermediate risk (IR) patients will be eligible for randomization to
either a control arm with two additional blocks of chemotherapy, or an experimental arm with two blocks
of blinatumomab. Both arms will proceed to protocol-specified hematopoietic stem cell transplant (HSCT)
that includes a rapid taper of immune suppression for patients with residual disease and no graft vs. host
disease (GVHD). Low risk (LR) patients will be eligible for randomization to either a control arm with two
blocks of chemotherapy followed by continuation and maintenance chemotherapy, or an experimental arm
with one block of chemotherapy, 2 blocks of blinatumomab, each followed by continuation and a third
additional block of blinatumomab followed by maintenance.
AALL1331 includes correlative laboratory studies to refine risk stratification, identify new targets for
therapy, identify biomarkers to predict response to chemotherapy and blinatumomab, and to link host
polymorphisms with various disease characteristics and toxicities.
Pre-treatment Evaluation
Block 1
Evaluation 1 1
HR/IR LR
Treatment Failure@
Arm C Arm D
Arm A Arm B
Block 2^
Blinatumomab
Evaluation 22 Block 3 (1st cycle)
1.1.1 To compare disease free survival (DFS) of HR and IR relapse B-ALL patients who
are randomized following Induction Block 1 chemotherapy to receive either two
intensive chemotherapy blocks or two 5-week blocks of blinatumomab (HR/IR
Randomization).
1.1.2 To compare DFS of LR relapse B-ALL patients who are randomized following
Block 1 chemotherapy to receive either chemotherapy alone or chemotherapy plus
blinatumomab (LR Randomization).
1.2.1 To compare overall survival (OS) of HR and IR relapse B-ALL patients who are
randomized following Induction Block 1 chemotherapy to receive either two
intensive chemotherapy blocks or two 5-week blocks of blinatumomab (HR/IR
Randomization).
1.3.1 To compare the rates of MRD ≥ 0.01% at the end of Block 2 and Block 3 for HR
and IR relapse B-ALL patients in HR/IR randomization.
1.3.2 To estimate, for treatment failure (TF) patients not previously receiving
blinatumomab, the hematologic complete remission rate (CR), rate of MRD
< 0.01%, and proportion able to proceed to hematopoietic stem cell transplant
(HSCT) in CR after treatment with blinatumomab.
1.3.3 To assess the feasibility and safety of rapid taper of immune suppression for the
subset of HSCT patients with MRD ≥ 0.01% pre- and/or post-HSCT with no acute
graft versus host disease (aGVHD).
2.0 BACKGROUND
2.1 Overview
First relapse of childhood B-ALL is a vexing clinical problem with high rates of subsequent
relapse and death with current treatment approaches.1 This trial will determine if
blinatumomab can reduce rates of second relapse, and improve survival. If successful, these
interventions could become standard of care for this disease. Additionally, success in
relapsed B-ALL will identify interventions that can subsequently be studied in patients
with newly diagnosed B-ALL in order to reduce rates of first relapse.
This study also is a paradigm shift away from the non-randomized historical control
outcome comparisons pursued in recent COG trials for patients with early relapse of B-
ALL (ADVL04P2 and AALL07P1). Those trials were designed to screen promising new
agents for possible activity in order to identify which agents should be tested further in
more definitive trials. Because of the remarkable efficacy of blinatumomab in adult trials2-
5
and in the Phase I portion of COG AALL1121,6 this study will directly test the efficacy
of blinatumomab in two risk groups of relapsed B-ALL in a randomized fashion.
This trial also seeks to enhance accrual to relapse childhood ALL trials within COG. Prior
to this trial, COG has used a fragmented approach that has required institutions to open a
different protocol for each of the many subsets of first relapse ALL. This trial will address
this issue by studying first relapse B-ALL in a single, comprehensive protocol.
Importantly, this study will establish an infrastructure for systematic studies of the
classification and biology of relapsed childhood B-ALL. Due to the fragmented nature of
previous trials and poor accrual, these types of studies have not been possible in the COG
until now.
This fragmented approach has been associated with problems in meeting accrual
goals and/or completion of studies in a timely and efficient manner. A major
obstacle to accrual is the reluctance of COG sites to open all of the relapse studies.
Since relapsed ALL is significantly less common than newly diagnosed ALL, the
annual accrual to each individual study is expected to be low enough at most COG
sites that it may not justify the regulatory burden involved in opening several
studies. This is exacerbated for the HR group by the limited accrual/duration of
each study, since individual COG sites may be unlikely to enroll any patients
during the conduct of such a study. For example, major frontline COG ALL trials
are typically open at 190-200 sites. In contrast the HR relapse trials have been open
in only about 100 sites, and the IR and LR trials at 125 to 150 sites.
The level of single agent activity seen with blinatumomab has not been seen in
recent Phase I ALL studies outside the use of tyrosine kinase inhibitors (TKI) in
patients with Philadelphia chromosome positive (Ph+) ALL, thus supporting a
Phase III randomized clinical trial with DFS endpoints. AALL1331 uses a risk-
stratified approach to test whether incorporating blinatumomab into treatment of
first relapse B-ALL will reduce rates of second relapse and improve DFS. If
successful, inclusion of blinatumomab could become standard of care for this
disease. Additionally, success in relapsed B-ALL will provide additional rationale
to test blinatumomab in newly diagnosed B-ALL patients in order to reduce rates
of first relapse.
Neurological adverse events have been described with blinatumomab and will be
closely monitored on this study. One potential concern regards the safety of
combining blinatumomab with IT chemotherapy. In the current Phase I/II pediatric
study cited above, IT methotrexate or IT triples are included prior to Cycle 1, at
Day 15 of Cycle 1 and at Day 29 of each cycle. No unusual or increased CNS side
effects have been seen in this setting.10
Cytokine release syndrome (CRS) has also been described. This is more prevalent
in patients with higher leukemia burden, however can occur in any patient treated
Late B-ALL marrow relapse patients have excellent end-reinduction CR2 rates
(> 95%),7 but long term EFS is suboptimal at about 50 – 60%. For these late B-
ALL marrow relapse patients, end-Block 1 MRD has been identified in multiple
studies to be a strong prognostic factor.7,12-14 In AALL01P2, for example, the 5-yr
EFS for late marrow/MRD-negative patients (< 0.01% by flow at end-Block 1,
occurring in about 50 % of patients) was 57% vs. 24% for those with MRD
≥ 0.01%.7 Thus, AALL1331 will use MRD response to stratify late pre-B marrow
relapse patients into those for whom HSCT is or is not indicated. We will use an
MRD threshold of 0.1% at end-Block 1 for this HSCT allocation (see Table 3
below). This is based on data from AALL01P2 and the ongoing AALL0433 trial,
in which MRD data (end Blocks 1, 2 and 3) have been collected for a large cohort
(~150) of late B-ALL marrow relapse patients. The AALL0433 cohort is
especially informative since the post-Block 3 therapy given in the context of
AALL0433 is uniform (this is not true for AALL01P2). Recent analyses of MRD
and outcome from patients enrolled on COG AALL0433 with BM or combined
relapse and available end Induction MRD data (n=175) showed 3-yr. EFS 80.2 ±
5.2% vs. 40.9 ± 8.4%, and 3-yr. OS of 87.1 ± 4.3% vs. 56.2 ± 9.0% for those below
(n=112) or above (n=63) an MRD cutoff of 0.1%.15 Thus far, about 62% of late
BM relapse patients enrolled in AALL0433 attain MRD < 0.1% by end Block 1.
Summarily, for late B-ALL marrow relapse patients, post-Block 1 treatment will
be risk-stratified, based on end-Block 1 MRD. MRD analysis will be performed
centrally in Dr. Borowitz’s laboratory at Johns Hopkins using flow cytometry and
a cutoff for MRD positivity of ≥ 0.1% will be used. Patients with end-Block 1
MRD levels of ≥ 0.1% will be classified as IR and will be eligible to participate in
HR/IR randomization. Those with end-Block 1 MRD levels of < 0.1% will be
classified as LR and will be eligible to participate in LR Randomization (see
Table 3 below).
For B-ALL IEM relapses, expected outcomes with chemotherapy and radiation for
those relapsing at least 18 months from diagnosis are similar to those for late
marrow LR patients.16,17 It is generally agreed that HSCT is not indicated for B-
ALL late IEM relapses. An exception to this will be patients found to have
detectable minimal marrow disease at study entry that does not clear to < 0.1% by
the end of Block 1, based on published literature that minimal marrow disease in
the setting of extramedullary relapse is associated with increased risk of
subsequent marrow relapse.18,19 Thus, B-ALL late IEM relapses that have MRD
< 0.1% (or uninterpretable MRD results) at end Block 1 will be classified as LR
and will be eligible to participate in LR Randomization. B-ALL late IEM/end
Block 1 MRD ≥0.1% relapses will be classified as IR and will be eligible to
participate in HR/IR randomization.
All other CNS3 relapse patients that meet IR or HR criteria will proceed to total
body irradiation (TBI) based HSCT as described in Section 14.5. In addition to the
TBI, these patients will also receive a protocol-specified cranial radiation boost as
part of their HSCT preparative regimen.
2.2.9 Additional non-randomized Pilot Intervention for Very High Risk Subset of HSCT
Recipients based on MRD and aGVHD
Relapse rates for patients with relapsed B-ALL who are MRD+ immediately prior
to HSCT or are MRD+ in the first 100 days post HSCT are exceedingly high,
particularly if there is no aGVHD.30-33 Table 7 below shows the relative risk of
relapse in the recently closed COG ASCT0431 trial according to MRD (using
0.01% as a threshold) and aGVHD status by Day +55.33
in Europe and the US have shown that patients defined as high risk for relapse
based upon increasing recipient chimerism (i.e., increased percentage of recipient
DNA markers) can successfully undergo withdrawal of immune suppression (IS)
without excessive toxicity, and their survival is improved. Out of 46 patients with
increasing recipient chimerism (a group at very high risk for relapse), Bader et al
intervened in 31 patients with immune suppression (IS) withdrawal, donor
lymphocyte infusions (DLI), or both. Three-year EFS was 37% vs. 0% in the
intervention vs. non-intervention groups (p < 0.001).34 Of the 31 patients, 11
received IS withdrawal alone; 2 relapsed and 1 died of TRM with 8/11 surviving.
Three, 2, and 2 patients developed Grades 1, 2, and 3 aGHVD respectively, with
no Grade IV or chronic GVHD. A smaller portion of patients responded to DLI
(4/17) or a combination of both (1/3). Follow up publications by the Bader group
and American groups have shown that patients defined at high risk for relapse by
chimerism can safely undergo immune withdrawal with a survival benefit to the
patient.35,36 It is reasonable, therefore, to hypothesize that IS withdrawal may
provide benefit, and it is necessary to do this in a standardized manner in the
context of this trial. In addition, understanding the proportion of high risk patients
who can successfully discontinue IS is vital to future trial planning, as many
interventions to prevent relapse under consideration require patients to be on no or
minimal IS.
Please see Appendix XIII for detailed CTEP Registration Procedures for Investigators
and Associates, and Cancer Trials Support Unit (CTSU) Registration Procedures
including: how to download site registration documents; requirement s for site
registration, submission of regulatory documents and how to check your site’s
registration status.
Study centers can check the status of their registration packets by querying the
Regulatory Support System (RSS) site registration status page of the CTSU
members’ web site by entering credentials at https://www.ctsu.org. For sites under
the CIRB initiative, IRB data will automatically load to RSS.
Note: Sites participating on the NCI CIRB initiative and accepting CIRB
approval for the study are not required to submit separate IRB approval
documentation to the CTSU Regulatory Office for initial, continuing or
amendment review. This information will be provided to the CTSU Regulatory
Office from the CIRB at the time the site’s Signatory Institution accepts the CIRB
approval. The Signatory site may be contacted by the CTSU Regulatory Office or
asked to complete information verifying the participating institutions on the study.
Other site registration requirements (i.e., laboratory certifications, protocol-
specific training certifications, or modality credentialing) must be submitted to the
CTSU Regulatory Office or compliance communicated per protocol instructions.
All site staff will use OPEN to enroll patients to this study. It is integrated with the
CTSU Enterprise System for regulatory and roster data and, upon enrollment,
initializes the patient position in the Rave database. OPEN can be accessed at
https://open.ctsu.org or from the OPEN tab on the CTSU members’ side of the
website at https://www.ctsu.org.
• All eligibility criteria have been met within the protocol stated timeframes.
• All patients have signed an appropriate consent form and HIPAA
authorization form (if applicable).
Note: The OPEN system will provide the site with a printable confirmation of
registration and treatment information. Please print this confirmation for your
records.
Further instructional information is provided on the CTSU members' web site
OPEN tab or within the OPEN URL (https://open.ctsu.org). For any additional
questions contact the CTSU Help Desk at 1-888-823-5923 or
[email protected].
See Section 3.1.6 below for details regarding randomization and treatment
assignment for patients.
3.1.4 Timing
Informed consent: Except for administration of intrathecal chemotherapy, informed
consent/parental permission MUST be signed before protocol therapy begins. See
Section 3.1.5 for summary of time points to obtain informed consent.
Study enrollment: Study enrollment must take place no later than five (5) calendar
days after beginning protocol therapy. If study enrollment takes place before
starting protocol therapy, the date that protocol therapy is projected to start must
be no later than five (5) calendar days after enrollment.
Eligibility studies: Patients must meet all eligibility criteria prior to the start of
protocol therapy or enrollment, whichever occurs first. All clinical and laboratory
studies to determine eligibility must be performed within 7 days prior to
enrollment.
Risk assignment according to the criteria outlined in Section 3.3 will occur prior
to submission of End-Block 1 callback. Consenting IR and HR patients will be
randomized according to HR/IR Randomization upon submission of callback.
Consenting LR patients will be randomized according to LR Randomization upon
submission of callback. Patients deemed Treatment Failure at Evaluation 1 who
are eligible for and consent to Salvage Therapy will be assigned to Salvage
Therapy upon submission of End-Block 1 callback.
Patients on Arm A who are deemed Treatment Failure at Evaluation 2 and are
eligible for and consent to Salvage Therapy will be assigned to Salvage
Therapy upon submission of End Block 2 callback
3.1.7 Callbacks
Callback is performed using the Oncology Patient Enrollment Network (OPEN).
Population for
Callback Timing Purpose
Callback
End Block 1 Evaluation 1* Consenting LR Randomize LR
Callback patients not off patients to either of
(ALL protocol therapy 2 treatment arms:
patients after completion of Arm C (Control
continuing to Block 1 therapy. Arm) or
post-Block 1 Arm D
therapy) (Experimental Arm).
Treatment Failure
patients who are Assign patients to
eligible for and Salvage Therapy
consent to Salvage (Blinatumomab-S)
Therapy
See Section 7.1 for required studies to be obtained prior to starting protocol therapy.
INCLUSION CRITERIA
3.2.1 Age
Patients ≥1 year and < 31 years of age at the time of relapse will be eligible.
3.2.2 Diagnosis
First relapse of B-ALL with or without extramedullary disease.
Please refer to Section 3.3 for definitions of relapse and criteria for risk classification.
3.2.3.1 No waiting period for patients who relapse while receiving standard
Maintenance therapy
3.2.3.5 Stem cell transplant or rescue: Patient has not had a prior stem cell
transplant or rescue.
3.2.3.7 Patient has not received prior relapse-directed therapy (i.e., this
protocol is intended as the INITIAL treatment of first relapse).
Maximum Serum
Age
Creatinine (mg/dL)
Male Female
1 to < 2 years 0.6 0.6
2 to < 6 years 0.8 0.8
6 to < 10 years 1 1
10 to < 13 years 1.2 1.2
13 to < 16 years 1.5 1.4
≥ 16 years 1.7 1.4
The threshold creatinine values in this Table were derived from the Schwartz
formula for estimating GFR utilizing child length and stature data published
by the CDC.37
3.2.5.2 Adequate liver function defined as a direct bilirubin < 3.0 mg/dL.
3.2.6.5 Patients with known optic nerve and/or retinal involvement are not
eligible. Patients who are presenting with visual disturbances should
have an ophthalmologic exam and, if indicated, an MRI to determine
optic nerve or retinal involvement.
3.2.6.10 Patients who are pregnant since fetal toxicities and teratogenic effects
have been noted for several of the study drugs. A pregnancy test is
required for female patients of childbearing potential.
3.2.6.11 Sexually active patients of reproductive potential who have not agreed
to use an effective contraceptive method for the duration of their study
participation.
3.2.6.12
a.) Patients with pre-existing significant central nervous system
pathology that would preclude treatment with blinatumomab,
including: history of severe brain injury, dementia, cerebellar
disease, organic brain syndrome, psychosis, coordination
/movement disorder, or autoimmune disease with CNS involvement
are not eligible. Patients with a history of cerebrovascular
ischemia/hemorrhage with residual deficits are not eligible.
(Patients with a history of cerebrovascular ischemia/hemorrhage
remain eligible provided all neurologic deficits have resolved)
3.2.7.1 All patients and/or their parent or legal guardian must sign a written
informed consent.
3.2.7.2 All institutional, FDA, and NCI requirements for human studies must be
met.
3.3 Definitions
Definitions of Relapse
RELAPSE:
Any recurrence of disease whether in marrow or extramedullary. Relapse should be
confirmed by pathology examination of appropriate tissue.
ISOLATED BONE MARROW RELAPSE: Patients with an M3 marrow at any point after
achieving remission without involvement of the CNS and/or testicles. Every effort should
be made to confirm morphologic relapse using flow cytometry, FISH and/or cytogenetics.
CNS RELAPSE: Positive cytomorphology and WBC ≥ 5/µL OR clinical signs of CNS
leukemia such as facial nerve palsy, brain/eye involvement, or hypothalamic syndrome that
are, in the opinion of the investigator, more likely due to recurrent CNS leukemia than to
alternative causes (e.g., viral infection or chemotherapy toxicity). If any CSF evaluation shows
positive cytomorphology and WBC < 5/µL, a second CSF evaluation is recommended within
2-4 weeks. While identification of a leukemic clone in CSF by flow cytometry (TdT, CD19,
CD10, etc.) or FISH for diagnostic karyotypic abnormality may be useful, definitive evidence
of CNS involvement (i.e. WBC ≥ 5/µL OR clinical signs of CNS leukemia) is required for the
diagnosis of a CNS relapse. Note that AALL1331 excludes patients with known optic nerve
and/or retinal involvement (Section 3.2.5.4).
CNS Status:
NOTE: in this protocol “CNS-positive” means meeting the criteria for CNS Relapse
(whether isolated CNS or combined relapse) at the time of AALL1331 study entry as listed
in the Definitions of Relapse above. CNS 2 status first noted at the time of enrollment for
concurrent relapse at another site (i.e. bone marrow) will not be considered ‘CNS-positive’.
CNS 1: In cerebral spinal fluid (CSF), absence of blasts on cytospin preparation, regardless
of the number of white blood cells (WBCs).
CNS 2: In CSF, presence < 5/µL WBCs and cytospin positive for blasts, or ≥ 5/µL WBCs
but negative by Steinherz/Bleyer algorithm:
CNS 2a: < 10/µL RBCs; < 5/µL WBCs and cytospin positive for blasts;
CNS 2b: ≥ 10/µL RBCs; < 5/µL WBCs and cytospin positive for blasts; and
CNS 2c: ≥ 10/µL RBCs; ≥ 5/µL WBCs and cytospin positive for blasts but negative
by Steinherz/Bleyer algorithm (see below).
CNS3: In CSF, presence of ≥ 5/µL WBCs and cytospin positive for blasts and/or clinical
signs of CNS leukemia:
CNS 3a: < 10/µL RBCs; ≥ 5/µL WBCs and cytospin positive for blasts;
CNS 3b: ≥ 10/µL RBCs, ≥ 5/µL WBCs and positive by Steinherz/Bleyer algorithm
(see below);
CNS 3c: Clinical signs of CNS leukemia (such as facial nerve palsy, brain/eye
involvement or hypothalamic syndrome).
A patient with CSF WBC ≥ 5/µL blasts, whose CSF WBC/RBC is 2X greater than the blood
WBC/RBC ratio, has CNS disease at diagnosis. Example: CSF WBC = 60/µL; CSF RBC =
1500/µL; blood WBC = 46000/µL; blood RBC = 3.0 X 106/µL:
M2 Marrow: 5% - 25% blasts in a bone marrow aspirate with at least 200 cells counted.
M3 Marrow: > 25% blasts in a bone marrow aspirate with at least 200 cells counted.
* The timing of relapse is measured from the date of initial diagnosis to the date of
relapse
Complete Remission
Complete remission (CR) is defined as an M1 marrow (< 5% blasts) with no evidence of
circulating blasts or extramedullary disease and with peripheral count recovery, defined as
absolute neutrophil count higher than or equal to 500/µL and platelet count higher than or
equal to 50,000/µL.
Hospitalization:
Hospitalization is STRONGLY recommended during the first 3 days of the first
blinatumomab Cycle and the first 2 days of subsequent blinatumomab cycles in case of a
cytokine reaction.
CNS Leukemia
All patients with CNS3 involvement at the time of relapse will receive intrathecal triples
(ITT) instead of IT MTX. During Block 1 these patients will receive two additional ITT
doses and during Block 2 they will receive one additional ITT dose. For LR patients with
CNS3 involvement, AALL1331 will further intensify CNS-directed therapy by replacing
lower dose oral MTX (25 mg/m2 every 6 hours x 4 doses) with intermediate dose IV MTX
(1 g/m2 over 24 hours) during the each of the 2 continuation phases. All of these patients
will then receive 1800 cGy cranial radiation and concurrent chemotherapy (including high
dose dexamethasone) between and the first and second 12 week blocks of Maintenance. IR
and HR patients will proceed to TBI-based HSCT and these patients will also receive a
protocol-specified cranial radiation boost as part of their HSCT preparative regimen.
Testicular Leukemia
Patients with suspected isolated testicular relapse or equivocal testicular enlargement with
concurrent bone marrow/CNS relapse must have a testicular biopsy performed at baseline.
Patients with persistent or equivocal testicular enlargement at the end of Block 1 must have
a testicular biopsy. Patients with persistent testicular leukemia at the end of Block 1 are
eligible to continue on study and will receive 2400 cGy of testicular radiation during either
Block 2 chemotherapy (for LR patients and HR/IR patients randomized to the control arm)
or during blinatumomab (for HR/IR patients randomized to the experimental arm and TF.)
Patients with extramedullary testicular leukemia that has resolved by end Block 1 will NOT
receive testicular irradiation, with the exception of that which may be used in the HSCT
preparative regimen for HR/IR patients.
HSCT
All patients enrolled on this protocol, their parents and full siblings should undergo
HLA tissue typing as soon as possible after diagnosis of relapse, and should have
transplantation consultation and initiation of a donor search. For HR/IR patients,
and for TF patients who achieve CR with Salvage Therapy (Blinatumomab-S), the
goal of this protocol is to move to transplant within 2 weeks of recovery from the last
block/cycle of therapy prior to transplant. Because unrelated donor acquisition can
take 8-12 weeks, centers must expedite the donor search process to meet this deadline.
Patient Pill Diary
It is recommended that patients use a Patient Pill Diary to keep track of oral medications.
See Section 5.0 for Dose Modifications based on Toxicities and Appendix I for
Supportive Care Guidelines.
IMPORTANT NOTE:
In order to continue to receive protocol therapy, patients will need to begin
post-Block 1 therapy within a specified time frame (see below). If a patient
has residual severe non-hematologic toxicities from Block 1 that are likely to
preclude beginning post-Block 1 therapy within this time frame, please DO
NOT complete end-Block 1 Callback for the patient. This is important to
minimize the number of patients who are randomized/assigned but are
unable to receive post-callback treatment.
required). Treatment must begin no later than 14 days after risk assignment for
patient to continue to receive protocol therapy.
Treatment failures Post-Block 1 (see Section 3.3. for definition) have the option
to receive up to two 5 week cycles of Blinatumomab (Sections 4.7, 4.8 and
APPENDIX II-F, and II-G respectively) and continue on to HSCT after achieving
CR.
• TF patients with residual CNS disease (CNS2/3) regardless of marrow status
should be removed from protocol therapy.
• TF patients with M3 marrow disease and no residual CNS disease (CNS1)
should proceed to Blinatumomab-S Cycle 1 without awaiting count recovery
(no later than 5 days after callback).
See Section 5.0 for Dose Modifications based on Toxicities and Appendix I for
Supportive Care Guidelines.
Dosing should be based on actual BSA.
Timing of ID MTX:
• For patients beginning Block 2 with M1 marrow, it is recommended to await
count recovery to an ANC ≥ 500/µL and platelets ≥ 50,000/µL prior to
beginning Day 8 IV methotrexate.
• Patients beginning Block 2 with M2 marrow should proceed without awaiting
count recovery.
• To continue to receive protocol therapy, all patients must begin Day 8 IV
methotrexate no later than 14 days after risk assignment.
Leucovorin: PO/IV
Day: 10, 11
Dose: 15 mg/m2/dose every 6 hours beginning 48 hrs after the START of ID
MTX infusion.
• If 48 hr methotrexate level is ≤ 0.5 μM, do not give more than two
doses of leucovorin (48 and 54 hours).
• If MTX level at 48 hours is > 0.5 μM, then continue hydration and
leucovorin rescue at 15 mg/m2/dose po/IV every 6 hours until MTX
levels are < 0.1 μM.
See Section 5.8 for ID MTX/LCV rescue and infusion guidelines.
• See Sections 7.1 and 13.0 for details on specimen acquisition, handling and
shipping.
Leucovorin: PO/IV
Days: 24, 25
Dose: 15 mg/m2/dose beginning 48 hrs after the START of ID MTX infusion.
• If 48 hr methotrexate level is ≤ 0.5 μM, do not give more than two doses of
leucovorin (48 and 54 hours).
• If MTX level at 48 hours is > 0.5 μM, then continue hydration and leucovorin
rescue at 15 mg/m2/dose po/IV every 6 hours until MTX levels are < 0.1 μM.
See Section 5.8 for ID MTX/LCV rescue and infusion guidelines.
See Section 5.0 for Dose Modifications based on Toxicities and Appendix I for
Supportive Care Guidelines.
Dexamethasone: PO/IV
Day: 1
Dose: Prior to day 1 therapy -
• A single dose of 10 mg/m2/dose will be administered 6 to 12 hours prior
to initiation of the blinatumomab infusion.
• A single dose of 5 mg/m2/dose will be administered within 30 minutes
prior to the start of the blinatumomab infusion
If using tablets, adjust dose upward to the nearest 0.25 mg. Oral solutions
are acceptable and intravenous preparations may be used on a temporary
basis, if needed
Methotrexate: Intrathecal (IT) - CNS1/2 PATIENTS ONLY
Day: 15, 29
Age-based dosing:
Age (yrs) Dose
1 – 1.99 8 mg
2 – 2.99 10 mg
3 – 8.99 12 mg
≥9 15 mg
• Two (2 mL) of bone marrow will be collected and shipped to the flow
cytometry laboratory for MRD determination
• See Sections 7.1 and 13.0 for details on specimen handling and shipping.
The Therapy Delivery Map (TDM) for the Blinatumomab Block: Cycle 2 (Arm B) is in
APPENDIX II-E.
See Section 5.0 for Dose Modifications based on Toxicities and Appendix I for
Supportive Care Guidelines.
Dexamethasone: PO/IV
Day: 1
Dose: Prior to day 1 therapy -
• A single dose of 10 mg/m2/dose will be administered 6 to 12 hours
prior to initiation of the blinatumomab infusion.
• A single dose of 5 mg/m2/dose will be administered within 30 minutes
prior to the start of the blinatumomab infusion
If using tablets, adjust dose upward to the nearest 0.25 mg. Oral solutions
are acceptable and intravenous preparations may be used on a temporary
basis, if needed.
Dexamethasone: PO/IV
Dose: Prior to day 1 therapy -
• A single dose of 10 mg/m2/dose will be administered 6 to 12 hours
prior to initiation of the blinatumomab infusion.
• A single dose of 5 mg/m2/dose will be administered within 30 minutes
prior to the start of the blinatumomab infusion
If using tablets, adjust dose upward to the nearest 0.25 mg. Oral solutions
are acceptable and intravenous preparations may be used on a temporary
basis, if needed.
The schedule below describes treatment for the 2nd cycle only. See Experimental Design
Schema.
The Therapy Delivery Map (TDM) for the Blinatumomab-S: Cycle 2 is in APPENDIX II-
G.
See Section 5.0 for Dose Modifications based on Toxicities and Appendix I for
Supportive Care Guidelines.
Dexamethasone: PO/IV
Dose: Prior to day 1 therapy -
• A single dose of 10 mg/m2/dose will be administered 6 to 12 hours prior
to initiation of the blinatumomab infusion.
All patients enrolled on this protocol and their full siblings should undergo
HLA tissue typing as soon as possible after diagnosis of relapse. All high-risk
patients should have transplantation consultation and initiation of a donor
search early while undergoing their initial Block 1 therapy. It is recommended
that all intermediate-risk patients also undergo transplantation consultation
as soon as possible after Block 1 therapy, so that a donor search can be
initiated immediately if they are noted to be MRD+ after initial induction. The
goal of this protocol is to move to transplantation within 2 weeks of recovery
from the final consolidation cycle or completion of Salvage Therapy
(Blinatumomab-S). As unrelated donor acquisition can take 8-12 weeks, to
meet this deadline centers must expedite the search process. Please see Section
7.1c for details of required evaluations.
Patients going on to HSCT will undergo an extensive pre-transplant
evaluation to assess remission status, assure adequate organ system function,
and document freedom from active viral, bacterial, and fungal infection.
Patients should begin their preparative regimen no later than 2 weeks after
obtaining the BM aspiration scheduled after recovery from their final
consolidation cycle or completion of Salvage Therapy (Blinatumomab-S). If
transplantation is delayed by 14 days for donor timing or other reasons,
patients may receive up to 6 weeks of bridging maintenance therapy as
described in Section 4.10, Appendix II-H. For patients who receive bridging
therapy, a repeat BM assessment will be needed to document remission and
MRD status within 14 days of starting the HSCT preparative regimen.
Patients who are unable to begin the transplant preparative regimen within 8
weeks of initial BM assessment are off protocol therapy.
Patients will receive 1 of 3 designated preparative regimens
(cyclophosphamide/total body irradiation (TBI) [cy/TBI] alone or with thiotepa or
etoposide). Fludarabine will be added to cy/TBI for cord blood recipients. GVHD
prophylaxis varies slightly according to stem cell source.
Patients who are high risk for relapse (MRD+ pre-HSCT with no evidence of
GVHD by Day +55, or with + MRD post BMT) are eligible for early taper of
immune suppression. The intent of the protocol will be for patients to undergo TBI-
based myeloablative conditioning. If patients are not eligible for a myeloablative
regimen and either do not undergo HSCT or undergo a reduced intensity
procedure, limited data will be collected regarding post transplant survival and
relapse.
In the recent COG ASCT0431 trial some variability was allowed in lung shielding
with total body irradition. Review of ASCT0431 data showed that lung irradiation
doses above 800 cGy were significantly associated with an increased risk of
treatment-related mortality and relapse, largely due to toxicity (Pulsipher and Lu,
unpublished data November 2014). Accordingly, AALL1331 mandates lung
shielding with total body irradiation to limit the lung dose to less than 800 cGy
(See Section 14.0 for Radiation Therapy Guidelines).
1. Infections
Patients with uncontrolled fungal, bacterial or viral infections are
excluded.
Patients acquiring fungal disease during induction therapy may
proceed if they have a significant response to antifungal therapy with
no or minimal evidence of active disease remaining by CT evaluation.
For children who are unable to cooperate for PFTs, the criteria are: no
evidence of dyspnea at rest, no exercise intolerance, and not requiring
supplemental oxygen therapy.
3. Performance Status
Patients must have a performance status corresponding to ECOG
scores of 0, 1, or 2. Use Karnofsky for patients > 16 years of age and
Lansky for patients ≤ 16 years of age. Please refer to performance
status scale at:
https://members.childrensoncologygroup.org/_files/protocol/Standar
d/PerformanceStatusScalesScoring.pdf
b. Patients who are MRD- (< 0.01%) within 2 weeks of beginning HSCT
preparative regimen but are found to be MRD+ (≥ 0.01%) on peri-
engraftment (Day +28-55) or Day +100 marrow who do not have any
evidence of aGVHD by Day +55 (peri-engraftment group) or Day
+100 (Day +100 group)
1800 cGy (unless alternative TBI dose given in which case the dose will be 1920
cGy). If the patient is ≤ 3 years at the time of transplant, centers may elect not to
give a boost because of developmental concerns. If the patient has CNS1 or CNS2
status at relapse, no boost will be given.
BM Recipient
Tacrolimus IV 0.02 mg/kg/day Begin Day -3 Target
(TAC) cont infusion concentration
8-12 ng/mL
Methotrexate IV 5 mg/m2 sib +1, +3, &+6
(MTX) donor 10mg/m2 sibs
URD^ +1, +3, +6, +11
URD
Cord Recipient
Cyclosporine A IV Varies by age, Begin Day -3 Target
(CYA) adjusted to concentration
target 200-400 ng/mL
Mycophenylate IV ≥50 kg: 1000 Begin Day -3
mofetil (MMF) mg q8h
< 50kg:
15mg/kg q8h
*
All drugs may be switched to PO after engraftment when patients are able to tolerate and
absorb PO medications.
^ URD, unrelated donor
The target serum trough concentration for tacrolimus is 8-12 ng/mL. Dose
adjustments are based on clinical judgment of the treating physician after
considering clinical toxicity, serum levels, GVHD, concomitant drug use and the
rate of rise or decline of the serum level.
The FDA has determined that a REMS (Risk Evaluation and Mitigation Strategy)
program is necessary to ensure the benefits of mycophenolate outweigh the risks
of first trimester pregnancy loss and congenital malformations associated with
mycophenolate use during pregnancy. Mycophenolate REMS is a program to tell
doctors, nurses, pharmacists and patients about the risks of taking mycophenolate
during pregnancy. Providers and patients (females of child-bearing potential) are
required to enroll in the program. The program also contains important information
about patient education and reporting details for pregnancy. Providers and patients
are required to sign acknowledgement forms prior to initiating therapy. Please refer
to the website for additional information (https://www.mycophenolaterems.com/).
If ≤ Grade 1 acute GVHD, MMF will be discontinued by Day +30. For patients
with > Grade 1 acute GVHD, the MMF taper will be per institutional protocols.
Patients may transition to PO formulations at a 1:1 IV to PO conversion when
clinically appropriate. Refer to Section 5.13 for dose adjustments based on serum
drug levels.
4.9.8.1 Taper for patients who are MRD positive pre-HSCT with no aGVHD by
Day +55 and for patients who are MRD positive > 0.01% by flow cytometry at
their peri-engraftment BM test (approx. Day +30)
Drug Stem Cell Source Taper Schedule
Tacrolimus Matched Sibling Taper already started by Day +42. At Day +55
BM/PBSC increase rate of taper to complete taper by Day
+70
Tacrolimus Unrelated Donor Start Day+55 off by Day +100
BM/PBSC
Cyclosporine Unrelated Cord Start Day+55 off by Day +100
Mycophenylate Unrelated Cord Continue until Day +45 or 7 days after
Mofetil engraftment, whichever day is later. Stop
without a taper.
4.9.8.2 Taper for patients who are MRD negative pre- HSCT with no aGVHD by
Day +55 who are MRD positive > 0.01% by flow cytometry at their Day +100 test.
Drug Stem Cell Source Taper Schedule
Tacrolimus Matched Sibling Taper already started by Day +42 and pt
BM/PBSC should be off immune suppression. Stop
tacrolimus without taper if still on medication.
Tacrolimus Unrelated Donor Taper over 2-3 weeks as soon as MRD report
BM/PBSC is received.
Cyclosporine Unrelated Cord Taper over 2-3 weeks as soon as MRD report
is received.
Mycophenylate Unrelated Cord
Mofetil Should be off this medication.
Mercaptopurine: PO
Days: 1-42
Dose: 75 mg/m²/dose*
Other Considerations:
• *See Section 5.9 for suggested starting dose based on TPMT status (if status
is known)
• Administer in the evening on an empty stomach (at least 1 hour before or
two (2) hours after food or drink except water).
• The liquid or tablet formulation may be used. If using tablets, adjust daily
dose using the dosing nomogram in Appendix III to attain a weekly
cumulative dose as close to 525 mg/m2/week as possible
Methotrexate: PO
Days: 1, 8, 15, 22, 29, 36
Dose: 20 mg/m2/dose
Administer on an empty stomach (at least 1 hour before or 2 hours after food or
drink except water).
The therapy delivery map for Continuation 1 and Continuation 2 is in APPENDIX II-I.
See Section 5.0 for Dose Modifications based on Toxicities and Appendix I
for Supportive Care Guidelines.
Mercaptopurine: PO
Days: 1-42
Dose: 75 mg/m²/dose*
Other Considerations:
• *See Section 5.9 for suggested starting dose based on TPMT status, if
known
• Administer in the evening on an empty stomach (at least 1 hour before
or two (2) hours after food or drink except water).
• The liquid or tablet formulation may be used. If using tablets, adjust
daily dose using the dosing nomogram in Appendix III to attain a
weekly cumulative dose as close to 525 mg/m2/week as possible
Methotrexate: PO
Days: 8, 15, 29, 36
Dose: 20 mg/m2/dose
Administer on an empty stomach (at least 1 hour before or 2 hours after food
or drink except water).
Leucovorin: PO
Day: 24
Dose: 10 mg/m2/dose for 2 doses 6 hours apart beginning 48 hrs after the
START of Day 22 Methotrexate
Leucovorin: PO/IV
Days: 24, 25
Dose: 15 mg/m2/dose every 6 hours beginning 48 hrs after the START of ID
MTX infusion.
• If 48 hr methotrexate level is ≤ 0.5 μM, do not give more than two doses of
leucovorin (48 and 54 hours).
• If MTX level at 48 hours is > 0.5 μM, then continue hydration and
leucovorin rescue at 15 mg/m2/dose po/IV every 6 hours until MTX levels
are < 0.1 μM.
•
Thioguanine: PO
Days 43-49.
Dose: 40 mg/m2/dose/once daily
Other Considerations:
• Administer in the evening on an empty stomach (at least 1 hour before or two
(2) hours after food or drink except water).
• Adjust daily dose using the dosing nomogram in Appendix IV to attain a
weekly cumulative dose as close to 280 mg/m2/week as possible.
• An oral suspension can also be compounded for patients who cannot swallow
pills. The compounded oral suspension is recommended for patients with a
BSA between 0.27 and 0.48 m2. (see Section 6.18)
Cytarabine: IV/SQ
Days: 44-47, 51-54
Dose: 50 mg/m2/dose
All other patients receive Maintenance-Post Cycle 1 treatment (Section 4.14, Appendix
II-L) immediately following Maintenance Cycle 1.
Mercaptopurine: PO
Days: 1-84
Dose: 75 mg/m²/dose*
Other Considerations:
• *See Section 5.9 for suggested starting dose based on TPMT status, if
known
• Administer in the evening on an empty stomach (at least 1 hour before
or two (2) hours after food or drink except water).
• The liquid or tablet formulation may be used. If using tablets, adjust
daily dose using the dosing nomogram in Appendix III to attain a
weekly cumulative dose as close to 525 mg/m2/week as possible
Methotrexate: PO
Days: 8, 15, 22, 29, 36, 43, 50, 57, 64, 71, 78
Dose: 20 mg/m2/dose
NOTE: CNS3 and Isolated CNS Relapse ONLY are to be given Chemoradiation (see
Section 4.13, APPENDIX II-K) between Maintenance Cycle 1 and subsequent
Maintenance cycles (Maintenance-Post Cycle 1). All other patients receive
Maintenance-Post Cycle 1 immediately following Maintenance Cycle 1.
See Section 5.0 for Dose Modifications based on Toxicities and Appendix I
for Supportive Care Guidelines.
Mercaptopurine: PO
Days: 1-84
Dose: 75 mg/m²/dose*
Other Considerations:
• *See Section 5.9 for suggested starting dose based on TPMT status
(if status is known)
• Administer in the evening on an empty stomach (at least 1 hour
before or two (2) hours after food or drink except water).
• The liquid or tablet formulation may be used. If using tablets, adjust
daily dose using the dosing nomogram in Appendix III to attain a
weekly cumulative dose as close to 525 mg/m2/week as possible
Methotrexate: PO
Days: 8, 15, 22, 29, 36, 43, 50, 57, 64, 71, 78
Dose: 20 mg/m2/dose
The Therapy Delivery Map (TDM) for the Blinatumomab Block: Cycle 1 (Arm D) is in
APPENDIX II-M.
See Section 5.0 for Dose Modifications based on Toxicities and Appendix I
for Supportive Care Guidelines.
Dexamethasone: PO/IV
Day: 1
Dose: Prior to day 1 therapy -
• A single dose of 10 mg/m2/dose will be administered 6 to 12 hours prior
to initiation of the blinatumomab infusion.
See Section 5.0 for Dose Modifications based on Toxicities and Appendix I
for Supportive Care Guidelines.
Dexamethasone: PO/IV
Day: 1
Dose: Prior to day 1 therapy -
• A single dose of 10 mg/m2/dose will be administered 6 to 12 hours prior
to initiation of the blinatumomab infusion.
• A single dose of 5 mg/m2/dose will be administered within 30 minutes
prior to the start of the blinatumomab infusion
If using tablets, adjust dose upward to the nearest 0.25 mg. Oral solutions
are acceptable and intravenous preparations may be used on a temporary
basis, if needed
4.16.4 Following Blinatumomab Block Cycle 2:
LR B-ALL patients randomized to the experimental arm (Arm D) will receive
Continuation 2- (Section 4.11, APPENDIX II-I)
Note: Premedication with antihistamines to decrease the risk of overt allergy symptoms is
strongly discouraged since anti-histamine use may mask the appearance of systemic
allergy. Systemic allergy is frequently associated with the presence of asparaginase
neutralizing antibodies, which render asparaginase therapy ineffective. In the event of
severe systemic or recurrent local allergic reaction, Erwinia chrysanthemi asparaginase
(Erwinaze®), which is FDA-approved for this indication, should be substituted.
Anaphylaxis
Discontinue pegaspargase if the patient develops Grade 3 anaphylaxis as defined by
CTCAE v4.0 (symptomatic bronchospasm, with or without urticaria, parenteral
intervention indicated; allergy-related edema/angioedema; hypotension). If this occurs,
Erwinia asparaginase should be substituted.
Erwinia asparaginase has a shorter half-life and is associated with a shorter duration of
asparagine depletion than native E. coli asparaginase, with “head-to-head” comparisons of
Erwinia and E. coli asparaginase, using the same dose and schedule for both preparations,
demonstrating a superior outcome, favoring E. coli asparaginase.67,68 Pegaspargase has a
longer half-life and is associated with more prolonged asparagine depletion than native E.
coli asparaginase, but the largest randomized trial comparing weekly native to bi-weekly
pegaspargase wasn’t powered to detect a difference in outcome.69 Current COG trials have
adopted pegaspargase as the preparation of choice, based on the results of CCG 1962.70
COG AALL07P2 showed that Erwinia asparaginase was well tolerated and achieved nadir
serum asparaginase activity at both 48 and 72 hours after dosing that was similar to that
achieved with pegaspargase. Based on these and other data, the FDA initially approved
Erwinia asparaginase for use following allergy to pegaspargase, with a dose of Erwinia
25,000 IU/m2 x 6 doses IM on a Monday/Wednesday/Friday schedule substituted for a
single dose of pegaspargase. In December 2014, the FDA expanded its approval to include
intravenous as well as intramuscular administration.
The dose modification guidelines for ALL trials recommend the substitution for
replacement of Erwinia asparaginase for either native or pegaspargase utilizing the
following schedule:
To replace a dose of intravenous pegaspargase that was discontinued during the infusion
due to an allergic reaction, the following recommendations may be used to guide patient
care.
In the event that a pegaspargase infusion is discontinued for an allergic reaction, regardless
of amount received, substitution with Erwinia asparaginase should begin approximately 48
hours after pegaspargase has been discontinued and preferably to coincide with the
recommended Monday/Wednesday/Friday administration schedule detailed above in
patients who are clinically stable. Up to 6 doses of Erwinia asparaginase may be
administered, as tolerated, to replace the incomplete intravenous pegaspargase dose. Of
note, Erwinia asparaginase is recommended only for pegaspargase hypersensitivity
reactions, and not for pancreatitis, hepatitis, coagulation abnormalities, or other non-
hypersensitivity toxicities associated with pegaspargase. To best suit the needs of each
individual patient, additional modifications to these recommendations may be made at the
discretion of the treating physician.
Coagulopathy: If symptomatic, hold asparaginase until symptoms resolve, then resume with
the next scheduled dose. Consider factor replacement (FFP, cryoprecipitate, factor VIIa). Do
not withhold dose for abnormal laboratory findings without clinical symptoms.
Centers may elect to discontinue pegaspargase and switch to erwinia asparaginase based
upon laboratory evidence of silent inactivation of asparaginase activity in the absence of
clinical symptoms of hypersensitivity at their discretion.
5.2 Blinatumomab
The most frequent serious adverse events noted in patients treated with
blinatumomab to date are disorders of the nervous system, both peripheral and
central, and systemic cytokine release syndrome (CRS). Both categories of events are
more likely to occur within the first week of treatment with blinatumomab, and both
categories of events are usually reversible and able to be managed with attentive
supportive care.
AEs related to blinatumomab that require treatment interruption (according to table below)
and do not resolve to CTCAE ≤ Grade 1 within 14 days will require permanent
discontinuation of blinatumomab treatment. If the patient is eligible to continue protocol
therapy (chemotherapy and/or HSCT), then the patient may, at the discretion of the
investigator and family, continue to receive protocol therapy. Otherwise, the patient will
be off protocol therapy.
In the case that the AE(s) DO resolve within 14 days, blinatumomab treatment may
resume at a reduced dose of 5 µg/m2/day to complete the 28 day course (not counting the
duration of treatment interruption). NOTE: For Grade 4 Nervous System/Psychiatric and
thromboembolic AEs, blinatumomab must be permanently discontinued.
For patients who had experienced a ≥ Grade 2 Neurologic Systems and Psychiatric AE
related to blinatumomab, no dose escalation beyond 5 µg/m2/day will be permitted for
subsequent cycles. For patients who experienced other AEs related to blinatumomab,
subsequent cycles will begin at the reduced dose of 5 µg/m2/day, but may escalate to 15
µg/m2/day after 7 days if there are no significant blinatumomab-related AEs.
Table: Dose modifications for Adverse Events (AE) Possibly, Probably or Definitely Related to
Blinatumomab:
Supportive Escalation to
Restart allowed
Care (in 15 mcg/m2/day
Category: AE AE Stop (with premeds) if Restarting dose
addition to after 7 days in
(CTCAE v4.03) Grade Infusion? Gr 1 within 14 (mcg/m2/day)
institutional subsequent
days?
guidelines)* cycle allowed?
Nervous system/ 1 N - - -
Psychiatric1: 2, 3 Y DEX, CNS Y 5 N
All (other than seizure) 4 Y DEX, CNS N - -
1,2,3 Y SZ, DEX, CNS Y 5 N
Nervous system: Seizure
4 Y SZ, DEX, CNS N
2
Immune system : Cytokine
release syndrome, allergic 1 N - - -
reaction, anaphylaxis and
General: Infusion related 2,3 Y DEX Y 5 Y
reaction 4 Y DEX N - -
Blood and lymphatic
system3: Disseminated 1,2 N - - -
intravascular coagulation,
hemolysis, hemolytic
uremic syndrome, 3,4 Y Y 5 Y
thrombotic
thrombocytopenic purpura
Blood and lymphatic
system4: All others
(lymphopenia, neutropenia, 1,2,3,4 N - - -
anemia, thrombocytopenia,
etc.)
1 N - - -
Vascular: Thromboembolic
2, 3 Y Y 5 Y
event
4 Y N - -
Investigations5,6,
Metabolism and Nutrition:
All
(if not considered 1,2,3,4 N - - -
clinically relevant or
responding to routine
medical management)
1,2 N - - -
Investigations5,6,
Metabolism and Nutrition:
All
3,4 Y Y 5 Y
(if clinically relevant and
not responding to routine
medical management)
1,2 N - - -
All other AE
3,4 Y Y 5 Y
Table Footnotes:
1
Most AEs in the psychiatric disorders category are unlikely to be caused by blinatumomab and
generally require supportive care rather than dose modification or discontinuation of
blinatumomab (e.g., Insomnia, Depression, Anxiety). Psychiatric AEs that may reflect underlying
central nervous system toxicity (e.g., Confusion, Delirium, Hallucinations, Psychosis) are of greater
interest, particularly if accompanied by other AEs in the nervous system disorders category.
2
Close monitoring of fluid status by intake and output should be undertaken for the first week of
blinatumomab infusion. Efforts to keep patients balanced between intake and output should be
maintained, even if diuretic therapy (furosemide or similar) is needed to do this. Careful attention to
fluid status may prevent deterioration from capillary leak, however even with meticulous attention
some patients will experience pulmonary edema and require more aggressive respiratory support.
Treating physicians should use their clinical judgment and institutional standards for whatever
supportive care measures are needed during this period of time.
3
In the first days of treatment, transient DIC-like pictures may develop. Because patients are at risk
for capillary leak syndrome and cytokine release syndrome, appropriate supportive care with
dexamethasone (described above), blood products and factors (packed red cells, platelets,
cryoprecipitate, fresh frozen plasma), vitamin K, and/or albumin should be considered according to
institutional standards of care. Particularly in the first week of infusion, when the risk of capillary
leak and cytokine release is more prominent, appropriate use of blood products and factors is
preferred if laboratory indications suggest the need for replacement, as large volumes of crystalloid
fluids tend to exacerbate the capillary leak.
4
In the first days of treatment, a rapid transient drop in platelets, neutrophils and/or hemoglobin
may be observed. These effects are not necessarily cytokine-mediated. Counts typically recover to
baseline during treatment, and usually within two weeks of starting blinatumomab. Transfusion of
blood and platelets should be performed according to appropriate institutional standards.
5
In the first days of treatment, transient increases in transaminases up to over 1000 U/L may develop.
These have generally returned to baseline in the 1st week of treatment.
6
Decrease in serum immunoglobulins have been observed in patients treated with blinatumomab.
Intravenous immunoglobulin should be administered according to institutional standards, but is
recommended for any patient with a total IgG level below 400. Immunoglobulin must not be
administered through the line through which blinatumomab is actively being infused.
be administered if seizures develop, and continued throughout the blinatumomab infusion. Anti-convulsant
therapy should be considered starting at least 24-48 hours prior to any subsequent blinatumomab infusions,
and continuing for the remainder of those treatment cycles. Diagnostic measures to exclude potential
infectious causes should be conducted once the patient has stabilized (i.e., a lumbar puncture to evaluate
for bacterial, viral or fungal sources should be performed). Any identified pathology should be treated as
clinically appropriate.
CNS: A daily finger-nose-finger or writing sample test is recommended according to age-appropriate
activities for patients. In adults treated with blinatumomab, it has been found that a daily handwriting
sample can often predict future nervous system toxicity before the clinical toxicity develops. In case of a
change in finger-nose-finger or handwriting test it is recommended to start dexamethasone on the schedule
above to prevent
5.3 Cyclophosphamide
Hematuria: Omit in the presence of macroscopic hematuria. If there is a history of previous
significant hematuria, hydrate before cyclophosphamide until specific gravity is < 1.010
and hydrate at 125 mL/m2/hr for 24 hours after dose. Monitor for adequate urine output as
per institution guidelines. Give IV mesna at a total dose that is 60% of the
cyclophosphamide dose divided to 3 doses (e.g., if the cyclophosphamide dose is 1000
mg/m2, the total mesna dose is 600 mg/m2 or 200 mg/m2/dose). Give the first mesna dose
15 minutes before or at the same time as the cyclophosphamide dose and repeat 4 and
8 hours after the start of cyclophosphamide. This total daily dose of mesna can also be
administered as IV continuous infusion. The continuous infusion should be started 15-30
minutes before or at the same time as cyclophosphamide and finished no sooner than
8 hours after the end of cyclophosphamide infusion.
Renal Dysfunction: If creatinine clearance or radioisotope GFR is < 10 mL/min/1.73 m2,
reduce dose of cyclophosphamide by 50%. Prior to dose adjustment of cyclophosphamide, the
creatinine clearance should be repeated with good hydration.
Myelosuppression: Do not interrupt high dose (3000 mg/m2/dose) ARAC, once started, for
uncomplicated myelosuppression; do hold for proven or presumed serious infection and do
not make up missed doses during Block 3.
Once Continuation 1 and 2 have started, do not interrupt for uncomplicated
myelosuppression; do hold for proven or presumed serious infection.
Renal Dysfunction: Adequate renal function (defined as creatinine within normal range) is
required for the administration of high dose (3000 mg/m2/dose) ARAC. Creatinine
clearance (CrCl) should be measured for patients with elevated creatinine or suspected
renal insufficiency. For CrCl < 60 mL/min/1.73 m2, hold pending recovery and omit if
recovery requires > 3 weeks.
5.5 Anthracycline-Mitoxantrone
Cardiac Toxicity: Discontinue for clinical or echocardiographic evidence of
cardiomyopathy (SF < 27% or EF < 50%) or Grade 3-4 left ventricular systolic dysfunction
(LVSD) per CTCAE version 4.0.
Note: use the following updated term to report decreases in the EF or SF: cardiac
disorders-others
Hyperbilirubinemia:42
Direct Bili % Dose Reduction
< 1.2 mg/dL Full dose
1.2 – 3.0 mg/dL 50%
3.1 – 5.0 mg/dL 75%
> 5.0 mg/dL Withhold dose and administer next scheduled dose if toxicity has
resolved. Do not make up missed doses.
Extravasation:
In the event of an extravasation, discontinue the IV administration of the drug and
institute appropriate measures to prevent further extravasation and damage according to
institutional guidelines. Also see
https://members.childrensoncologygroup.org/_files/disc/Nursing/extravasationguidelines.
pdf for COG guidelines.
5.6 Etoposide
Allergic Reaction: Premedicate with diphenhydramine (1-2 mg/kg slow IV push,
maximum dose is 50 mg). If symptoms persist, add hydrocortisone 100-300 mg/m2.
Continue to use premedication before etoposide in future. Also consider substituting an
equimolar amount of etoposide phosphate, in the face of significant allergy and/or
hypotension. Etoposide phosphate is a water soluble prodrug that does not contain
polysorbate 80 and polyethyleneglycol, the solubilizing agent in etoposide that may induce
allergic reactions and hypotension. Etoposide phosphate is rapidly converted to etoposide
in vivo and provides total drug exposure, as represented by AUC (0-infinity), that is
statistically indistinguishable from that measured for etoposide at equimolar doses.
Renal Insufficiency: If renal function decreases, adjust etoposide as follows: CrCl 10-50
mL/min/1.73 m2, decrease dose by 25%; if CrCl < 10 mL/min/1.73 m2, decrease dose by
50%.
The following guidelines are offered for consideration following an acute event, but it must
be recognized that there are little data to support these approaches or any others. Thus the
treating physician must evaluate the patient and, with the family, make the best possible
decision with respect to the relative risk and benefit of continued therapy.
Following an acute neurotoxic event, a history and physical exam should guide the
differential diagnosis. A neurology consult may be of value and should be considered.
Seizures and other transient events may be linked to fever, infection, encephalitis,
meningitis, hypertension, electrolyte disturbance, hypoglycemia, trauma, intracranial
hemorrhage or thrombosis, narcotic withdrawal, illicit drug use, or other causes in addition
to the direct side effects of chemotherapy. Appropriate laboratory studies may include, but
are not limited to, blood cultures, a CBC, electrolytes, including glucose, calcium,
magnesium and phosphorus, renal and liver function studies and/or an examination of the
CSF. Imaging studies may include a CT scan and/or an MRI. The CT is commonly normal,
in the absence of stroke, but if calcifications are present, this finding may be indicative of
a more severe mineralizing leukoencephalopathy.52 MRI abnormalities may be
pronounced, but transient. Posterior reversible encephalopathy may be present on MR with
extensive diffusion abnormalities, but these do not appear to correlate with subsequent
demyelination or gliosis.53-55 Additional studies, including MR angiography and/or
venogram should be considered, if clinically indicated (e.g., focal deficits).
Many acute events, seizures or episodes of transient hemiparesis, are temporally related to
the administration of intrathecal therapy, commonly 9 to 11 days after the IT
administration.56 For patients who return to their “pre-event” status, without residual
deficits of physical or neurologic exam, there are few data to support or guide therapeutic
interventions. It is reasonable to hold the next dose of IT therapy, or, substitute IT Ara-C
for 1 dose of IT MTX, or triple IT therapy. It is also reasonable to include leucovorin rescue
at a dose of 5 mg/m2 q 12 hrs x 2 doses beginning 48 hours after the LP. This pattern of
rescue was associated with a clear diminution in the incidence of acute neurotoxicity in
one case series.56 There have been questions about potential interference of leucovorin with
the efficacy of the IT MTX, but there are little data to support or refute this position.
Moreover, the administration 48 hours later would minimize any potential interference. If
the event does not recur, resumption of standard therapy should be considered, following
1 modified or omitted IT dose. In the face of multiply recurrent events, or evidence of
progressive encephalopathy, another evaluation is warranted and the treating physician
may consider a more prolonged or definitive change in therapy. These decisions are
extremely difficult and may hinge on an individual’s view of the importance of quality of
life versus an increase in the risk of relapse. Since the greatest impact of CNS prophylaxis
occurs early in therapy, the timing of these events may also influence clinical decisions.
Cranial radiation has been suggested as an alternative to continued IT therapy though much
of the literature on long-term neurocognitive dysfunction supports a more deleterious effect
from CRT than IT therapy.57-60 Dramatic deviations from protocol recommended therapy
might result in the child being taken off protocol therapy.
The use of dextromethorphan (DM) has been suggested as a neuroprotectant, capable of
preventing NMDA mediated neurotoxicity without prohibitive toxicity. Low dose therapy
has been recommended, in part, based on data suggesting that DM is concentrated in brain
relative to serum. However, the literature on the use of DM supports a tight dose response
relationship, with the likelihood of sparing an initially unaffected area, following ischemic
damage, linked to dose, in both clinical trials and animal models of CNS ischemia.61-64 At
doses thought to be therapeutic, side effects have included nystagmus, nausea and
vomiting, distorted vision, ataxia, and dizziness. In addition, Hollander et al 65 have raised
concerns about the potential deleterious effects of long-term NMDA receptor blockade on
memory because hippocampal long-term potentiation is dependent on the activation of the
NMDA receptor. Thus in the absence of a clinical trial there are few data to support the
addition of DM.
Leucovorin rescue: 15 mg/m2 PO/IV at 48 and 54 hrs after the start of the MTX
infusion. If 48 hr methotrexate level is ≤ 0.5 μM, then only two doses of leucovorin
are administered (at 48 and 54 hours). If MTX level at 48 hours is > 0.5 μM, then
continue hydration and leucovorin rescue at 15 mg/m2/dose po/IV every 6 hours
until MTX levels are < 0.1 μM.
Hour 48: Check plasma methotrexate level at 48 hours after start of the
methotrexate infusion. If the level is ≤ 0.5 μM, then do not give more than two
doses of leucovorin (48 and 54 hours). If MTX level at 48 hours is > 0.5 μM, then
continue hydration and leucovorin rescue at 15 mg/m2/dose po/IV every 6 hours
until MTX levels are < 0.1 μM.
For MTX levels that exceed these expected values modify the rescue regimen
as noted below and increase hydration to 200 mL/m2/hr. Monitor urine pH to
assure a value ≥ 7.0 and monitor urine output to determine if volume is ≥ 80% of
the fluid intake, measured every 4 hours. If serum creatinine rises significantly, at
any time point (> 100% in 24 hours), assure appropriate urine pH and urine volume
as above and consider glucarpidase If urine output fails to continue at 80% of the
fluid intake, consider furosemide or acetazolamide. Regardless of urine output,
also consider glucarpidase (carboxypeptidase G2) (see below).
NOTE: For patients who have markedly delayed MTX clearance secondary to
renal dysfunction, consider using glucarpidase (carboxypeptidase G2,
Voraxaze).66,67 To obtain supplies of glucarpidase in the US contact the Voraxaze
24-hour Customer Service line at 855-786-7292. Additional information can be
found at http://www.btgplc.com/products/specialty-pharmaceuticals/voraxaze
regarding product availability through ASD Healthcare, Cardinal, and McKesson.
Canadian sites should contact McKesson at (877) 384-7425 for further
information. Sites in Australia and New Zealand should contact Hospira at 1300-
046-774 (local) or [email protected]. Patients requiring
glucarpidase rescue will remain on study.
It is essential that patients are NOT co-prescribed the following medicines which
reduce MTX excretion: NSAIDS, aspirin, ciprofloxacin, co-trimoxazole,
penicillin, probenecid, omeprazole.
Liver Dysfunction: Samples for the determination of ALT value must be drawn
within 72 hours, PRIOR to a course of intravenous MTX. Blood samples for ALT
should not be drawn following the start of MTX infusions as MTX causes
significant short term elevation in ALT levels.
ALT IV MTX
< 10 X ULN Continue with therapy as scheduled
10 – 20 X ULN Continue with therapy as scheduled for 1 cycle
10 – 20 X ULN for 2 Discontinue TMP/SMX*
consecutive cycles Hold therapy until ALT < 10 X ULN, then
resume at full doses at point of interruption.
Do not skip doses.
> 20 X ULN Hold therapy until ALT < 10 X ULN, then
resume at full doses at point of interruption.
Do not skip doses.
> 20 X ULN for > 2 weeks Evaluate with AST, Bili, Alkaline phosphatase,
PT, albumin, total protein, and hepatitis A, B,
C, CMV, and EBV serologies.
Consider liver biopsy before additional therapy
given. Notify Study Chair.
* Please see COG Supportive care Guidelines at:
https://members.childrensoncologygroup.org/prot/reference_materials.asp for TMP/SMX
substitutions.
Mucositis: For Grade 3-4 mucositis, withhold IV MTX until resolved. Increase
leucovorin rescue following the next course from 2 to 4 doses on a q6 hr schedule. If
subsequent course is not associated with Grade 3-4 mucositis, attempt to decrease the
number of leucovorin doses to 2. If mucositis recurs despite the extended leucovorin,
decrease the dose of MTX by 25%, increase hydration to 200 mL/m2/hr and continue
increased leucovorin as above. Should subsequent courses be well tolerated, use a
stepwise approach to resuming a standard approach to drug delivery. Consider
culturing lesions for herpes simplex if mucositis persists or recurs.
Myelosuppression: All chemotherapy should be held for ANC < 750/µL and
platelets < 75 000/µL.
See Section 5.13 for dose modifications when MTX is used post HSCT.
b) ANC ≥ 500/µL and < 750/µL and and/or platelets ≥ 50 000/µL and < 75 000/µL:
Reduce dose to 50% of original dose until ANC recovers to ≥ 750/µL and platelets
recover to ≥ 75 000/µL. Increase dose approximately every 2 weeks, first to 75%
of the original dose and then to full dose, provided ANC remains ≥ 750/µL and
platelets remain ≥ 75 000/µL.
c) ANC < 500/µL and/or platelets < 50 000/µL:
Discontinue dose until ANC is ≥ 750/µL and platelets are ≥ 75 000/µL. Restart
mercaptopurine and/or MTX at 50% of the original dose on the same day the
counts recover. Increase to 75% and then 100% of the original dose at 2-week
intervals provided ANC remains ≥ 750/µL and platelets remain ≥ 75 000/µL.
Consider a marrow evaluation in the face of persistent or prolonged neutropenia.
Prolonged cytopenia is defined as ANC < 750/µL and/or platelets < 75 000/µL after
withholding therapy for > 2 weeks. Perform a bone marrow examination after 2 weeks of
withholding chemotherapy, if no recovery is apparent. If monocyte count is increasing or
viral myelosuppression is clinically suspected, the bone marrow examination may be
postponed for 1-2 weeks and omitted if ANC and platelets fully recover by the 4th week
after therapy is withheld.
During Maintenance:
If neutrophil count falls below 500/µL or if platelet count falls below 50 000/µL during
Maintenance, MP and MTX will be held until recovery above these levels. For the first drop
in ANC or platelets, resume chemotherapy (both MP and MTX) at the same dose the patient
was taking prior to the episode of myelosuppression. If neutrophil count falls below 500/µL
or if platelet count falls below 50 000/µL for a second (or greater) time, discontinue doses of
MP and MTX until ANC is ≥ 750/µL and platelets are ≥ 75 000/µL. Restart both MP and
MTX at 50% of the dose prescribed at the time the medication was stopped. Then continue to
increase to 75% and then 100% of the dose prescribed prior to stopping the medication at 2-4
week intervals provided ANC remains ≥ 750/µL and platelets remain ≥ 75 000/µL. May
increase both 6-MP and MTX simultaneously.
If neutrophil count falls below 500/µL or if platelet count falls below 50 000/µL on > 2
occasions during Maintenance, perform thiopurine pharmacology testing as described below.
Should therapy be withheld for myelosuppression or elevated transaminase, do not “make up”
that week. Resume therapy at the correct point, chronologically.
Liver Dysfunction:
For increase in hepatic transaminases (SGPT/ALT or SGOT/AST) to greater than 5x ULN
consistent with Grade 3 toxicity, obtain total bilirubin. Monitor SGPT/ALT or SGOT/AST
and total bilirubin weekly during Continuation and every 4 weeks during Maintenance as long
as transaminases remain over 5x ULN.
If either of these occurs, hold MTX and monitor labs as above, weekly. Restart at full dose
therapy when the transaminase is less than 5x ULN, if bilirubin is normal. If liver dysfunction
persists, consider a trial period with MTX but without MP, especially if red cell MP
methylated derivatives are elevated. Also consider liver biopsy.
Exclude infectious hepatitis (A, B, C) for persistent (> 1 month) elevations in SGPT/ALT or
SGOT/AST above 5x ULN.
For dose modifications when MTX is given for GVHD prophylaxis see Section 5.13.3.
There are now CLIA certified tests for TPMT genotype and phenotype, and for thiopurine
metabolites (6-methyl mercaptopurine [6-MMP] and 6-TGN) measurements. Only 3 SNPs
constitute well over 90% of the inactivating mutations in the gene, based on studies in
numerous racial and ethnic groups worldwide. 68,73-76 Thus, the genotyping test has a low
false negative rate, and may be preferable to TPMT phenotype testing in cases where a
history of red cell transfusions would potentially confound assessments of RBC TPMT
activity. When the genotyping result is coupled with a phenotyping test for TPMT or with
thiopurine metabolite concentrations in erythrocytes, the reliability of the tests will be even
greater. Moreover, metabolite levels can provide an index of patient compliance with
thiopurine therapy.
- If the patient is homozygous wild-type (high activity) for TPMT, then discontinue
TMP/SMX and use pentamidine or dapsone. For modifications of the oral MP and
MTX see the beginning of this section.
5.10 Steroids (Dexamethasone)
Hypertension: Dose should not be reduced. Sodium restriction and anti-hypertensives
should be employed in an effort to control hypertension. Avoid calcium channel blockers
due to their potential prohemorrhagic effect.
Hyperglycemia: Dose should not be reduced for hyperglycemia. Rather, insulin therapy
should be employed to control the blood glucose level.
Pancreatitis: Do not modify dose for asymptomatic elevations of amylase and/or lipase.
Discontinue steroids, except for stress doses, in the presence of hemorrhagic pancreatitis or
severe pancreatitis.
Osteonecrosis (ON): Do not modify corticosteroid therapy for osteonecrosis (also referred to
as avascular necrosis) prior to Maintenance therapy. Consider omitting Maintenance steroid
for osteonecrosis Grade 1 (clinically asymptomatic, radiographic finding only). Omit
Maintenance steroid for osteonecrosis Grade 2 or greater, and notify study chair. Consider
resuming Maintenance steroid after 6 months if joint symptoms have resolved and if MRI
findings have significantly improved or normalized.
Varicella: Steroids should be held during active infection except during Induction. Do not hold
during incubation period following exposure.
Severe infection: Do not hold or discontinue steroids during Induction without serious
consideration, as this is a critical period in the treatment of ALL. Later in therapy, one may
consider holding steroid until patient achieves cardiovascular stability, except for “stress
doses.”
Severe psychosis: Dexamethasone dose may be reduced by 50% for severe psychosis. If
symptoms persist, consider switching to an equivalent dose of prednisone.
5.11 PO 6-Thioguanine (TG)
Continuation:
Oral TG will be held for suspected or proven serious infection.
For severe and/or unexpected myelosuppression, evaluate for TPMT activity as described
in Section 5.9.
5.12 Vincristine
PLEASE USE “BALIS” SCALE FOR GRADING NEUROPATHY (See text box
below)
Hyperbilirubinemia77,78:
Direct Bili Dose reduction
< 3.1 mg/dL Full dose (maximum dose: 2 mg),
3.1- 5.0 mg/dL 50% of calculated dose (maximum dose: 1 mg),
5.1-6.0 mg/dL 75% of calculated dose (maximum dose: 0.5 mg),
> 6.0 mg/dL Withhold dose and administer next scheduled dose if toxicity has
resolved.
Do not make up missed doses.
Extravasation:
In the event of an extravasation, discontinue the IV administration of the drug and
institute appropriate measures to prevent further extravasation and damage according to
institutional guidelines. Also see
https://members.childrensoncologygroup.org/_files/disc/Nursing/extravasationguidelines.
pdf for COG guidelines.
• Grade 2: Weakness that alters fine motor skills (buttoning shirt, coloring, writing or
drawing, using eating utensils) or gait without abrogating ability to perform these
tasks.
• Grade 3: Unable to perform fine motor tasks (buttoning shirt, coloring, writing or
drawing, using eating utensils) or unable to ambulate without assistance.
• Grade 4: Paralysis.
5.13.2 Cyclosporine
Cyclosporine commonly causes mild/moderate hypertension and less commonly
kidney or liver dysfunction, TAM, and neurological changes associated with
significant hypertension. When trough levels are kept in the therapeutic range and
patients receive adequate hydration and magnesium replacement, most of these
side effects can be minimized. Hypertension should be managed with single or
combination antihypertensive therapy. Cyclosporine should be held for severe
toxicities thought to be related to its administration (significant neurological
changes/malignant hypertension, TAM, kidney failure, etc.). Other immune
suppressive medications may be substituted if cyclosporine is not tolerated
5.13.3 Methotrexate
The most common acute side-effects of methotrexate include delay of count
recovery, worsened mucositis, and kidney and liver damage (can contribute to
VOD). Toxicity is directly related to the length of exposure to the drug. While
methotrexate is generally excreted rapidly, delayed excretion of methotrexate
occurs with decreased renal function and in circumstances where patients have
third-space fluid collections (pulmonary effusions, ascites, joint effusions, etc.).
The attending transplant physician should assess each patient prior to delivery of
each dose and decide whether full dose methotrexate should be administered.
Guidelines for modification of methotrexate dosing are listed in the tables below.
For significant third spacing (ascites, effusions, significant edema or weight gain
> 5-10% above baseline) consider dose reductions of 50% and leucovorin rescue.
5.13.7 Dose Adjustment of Chemotherapy for Patients Whose Weight Exceeds > 125%
Ideal Body Weight (IBW)
Chemotherapy given during the preparative regimen (thiotepa, cyclophosphamide,
and etoposide) will be dosed based on actual weight for patients ≤ 125% IBW.
Those > 125% IBW will be dosed based upon adjusted ideal body weight as
follows:
Adjusted ideal body weight = IBW + 0.25 (Actual weight – IBW).
The following formulas for pediatric and adult IBW calculations are
recommended, but IBW may be calculated according to institutional standard
operating procedures (SOPs).
list of reported and/or potential adverse events (AE) associated with an agent using a
uniform presentation of events by body system. In addition to the comprehensive list, a
subset, the Specific Protocol Exceptions to Expedited Reporting (SPEER), appears in a
separate column and is identified with bold and italicized text. This subset of AEs (SPEER)
is a list of events that are protocol specific exceptions to expedited reporting to NCI (except
as noted below). Refer to the 'CTEP, NCI Guidelines: Adverse Event Reporting
Requirements'
http://ctep.cancer.gov/protocolDevelopment/electronic_applications/docs/aeguidelines.pd
f for further clarification. Frequency is provided based on 486 patients. Below is the
CAEPR for Blinatumomab (AMG 103).
NOTE: Report AEs on the SPEER ONLY IF they exceed the grade noted in parentheses
next to the AE in the SPEER. If this CAEPR is part of a combination protocol using
multiple investigational agents and has an AE listed on different SPEERs, use the lower of
the grades to determine if expedited reporting is required.
1
This table will be updated as the toxicity profile of the agent is revised. Updates will be
distributed to all Principal Investigators at the time of revision. The current version can
be obtained by contacting [email protected]. Your name, the name of the
investigator, the protocol and the agent should be included in the e-mail.
2
Blinatumomab (AMG 103) is known to cause a variety of adverse events associated with
coagulopathy which may include: Activated partial thromboplastin time prolonged,
Disseminated intravascular coagulation, Fibrinogen decreased, INR increased,
Investigations - Other (blood fibrinogen increased), Investigations - Other (fibrin D dimer
increased), Investigations - Other (activated partial thromboplastin time shortened),
Investigations - Other (antithrombin III decreased), Investigations - Other (coagulation
factor XII level decreased), Investigations - Other (coagulation factor XIII level
increased), Investigations - Other (haptoglobin decreased), Investigations - Other (protein
S decreased), Platelet count decreased, and Thromboembolic events.
3
Symptoms of cytokine release syndrome (CRS) and/or allergic reaction may include
chills, fever, fatigue, flushing, bronchospasm, and hypotension. In some cases,
disseminated intravascular coagulation (DIC), capillary leak syndrome (CLS), and
hemophagocytic lymphohistiocytosis/macrophage activation syndrome (HLH/MAS)
have been reported in the setting of CRS.
4
Symptoms of hepatic dysfunction may include Alanine aminotransferase increased,
Alkaline phosphatase increased, Aspartate aminotransferase increased, Blood bilirubin
increased, and GGT increased under the INVESTGATIONS SOC.
5
Infection includes all 75 sites of infection under the INFECTIONS AND
INFESTATIONS SOC.
6
Acute kidney injury (acute renal failure) is associated with increased creatinine levels.
7
Tumor lysis syndrome is defined as a massive overload of potassium, phosphate, uric
acid, plus hypocalcemia, potentially causing lethal cardiac arrhythmias and/or renal
failure.
8
Blinatumomab (AMG103) is known to cause a variety of nervous system disorders which
may include: Ataxia, Dizziness, Cognitive disturbance, Concentration impairment,
Depressed level of consciousness, Dysphagia, Dysarthria, Dysesthesia, Encephalopathy,
Facial nerve disorder, Headache, Memory impairment, Paresthesia, Peripheral sensory
neuropathy, Seizure, Somnolence, Syncope, Transient ischemic attacks, Tremor, Voice
alteration, Nervous system disorders - Other (allodynia), Nervous Systems disorders -
Other (cerebellar syndrome), Nervous system disorders - Other (dysgraphia), Nervous
system disorders - Other (epilepsy), Nervous system disorders - Other (facial palsy),
Nervous system disorders - Other (hemiparesis), Nervous system disorders - Other
(hypertonia), Nervous system disorders - Other (pleocytosis), and Nervous system
disorders - Other (polyneuropathy). Additionally, symptoms of some nervous system
disorders are adverse events under the PSYCHIATRIC DISORDERS SOC and may
include: Confusion, Agitation, Anxiety, Hallucinations, Personality change, and
Psychosis.
9
Rash includes rash, rash maculo-papular, erythema, erythematous rash, generalized rash,
exanthema, allergic dermatitis, and palmar-plantar erythrodysesthesia syndrome.
Adverse events also reported on blinatumomab (AMG 103) trials but with the
relationship to blinatumomab (AMG 103) still undetermined:
Note: Blinatumomab (AMG 103) in combination with other agents could cause an
exacerbation of any adverse event currently known to be caused by the other agent, or the
combination may result in events never previously associated with either agent
Store intact vials of blinatumomab and the IV solution stabilizer of blinatumomab refrigerated
at 2° – 8°C (36° – 46°F). Protect from light. Shelf life stability studies of the intact vials of
blinatumomab and stabilizer solution are on-going. The stability of the final prepared IV
solution is 8 days when stored refrigerated at 2° – 8°C. For storage prior to administration,
the prepared infusion solution must be kept at 2° C to 8° C ( 36°F to 46°F). The total storage
and administration time must not exceed 8 days. Once at room temperature, discard the IV
bag after 48 hours.
Preparation
Only trained staff may prepare the blinatumomab IV solution. Each site’s standard
procedure for compounding blinatumomab must be in compliance with the USP < 797 >
guidelines (ISO Class 5 or better). Use aseptic technique and prepare blinatumomab IV
solution under a qualified biological safety cabinet.
The final IV solution must be prepared in the following sequential order (do not
deviate from this order; refer to the table below for volume details):
1. Reconstitute blinatumomab lyophilized powder (30.3 mcg/vial)
a. Add 3 mL of Sterile Water for Injection (SWFI) to the vial to yield
3.07 mL of blinatumomab at a final concentration of 9.87
mcg/mL.
b. Rotate the vial to dissolve all powder. Do not shake.
c. The stability of the reconstituted vial is 4 hours at room
temperature (22°C – 27°C) or 24 hours refrigerated at 2° – 8°C.
2. Add the appropriate amount of 0.9% NaCl into the IV bag
3. Add the IV solution stabilizer for blinatumomab to the IV bag
4. Add the calculated dose (mL) of blinatumomab into the solution in the IV
bag
a. Rotate the IV bag to mix the solution thoroughly. Do not shake.
Avoid foaming the IV bag.
b. Visually inspect for floating particles or discoloration of the IV
solution. If floaters or discoloration is present, do not use the
prepared solution.
c. The total volume of blinatumomab IV solution will account for the
volume of the IV infusion set for the inpatient or outpatient setting.
Guidelines for Administration: See Treatment and Dose Modifications sections of the
protocol.
Monitor patients for cytokine release syndrome, tumor lysis syndrome, and infusion
reaction. Refer to protocol for specific recommendation. Monitor patients for psychiatric
events such as confusion, disorientation, and cognitive attention disturbances. Patients
should not drive or operate dangerous machinery while receiving blinatumomab.
Supplier
Blinatumomab and the solution stabilizer for blinatumomab is supplied by Amgen, Inc
and distributed by the Division of Cancer Treatment and Diagnosis (DCTD), NCI.
Agent Accountability
Agent Inventory Records:
The investigator, or a responsible party designated by the investigator, must maintain a
careful record of the inventory and disposition of all agents received from DCTD using the
NCI Drug Accountability Record Form (DARF). See the CTEP home page at
http://ctep.cancer.gov for the Procedures for Drug Accountability and Storage and to obtain
a copy of the DARF and Clinical Drug Request form.
Agent Returns:
Investigators/Designees must return unused DCTD supplied investigational agent to the
NCI clinical repository as soon as possible when: the agent is no longer required because
the study is completed or discontinued and the agent cannot be transferred to another
DCTD sponsored protocol; the agent is outdated or the agent is damaged or unfit for use.
Regulations require that all agents received from the DCTD, NCI be returned to the DCTD,
NCI for accountability and disposition. Return only unused vials/bottles. Do NOT return
opened or partially used vials/bottles unless specifically requested otherwise in the
protocol. See the CTEP web site for Policy and Guidelines for Investigational agent
Returns at: http://ctep.cancer.gov/protocolDevelopment/default.htm#agents_drugs.
The appropriate forms may be obtained at:
http://ctep.cancer.gov/forms/docs/return_form.pdf
Effective asparaginase levels have been defined as activity of ≥ 0.1 International Units per
mL. Clinical trials with asparaginase Erwinia chrysanthemi demonstrated that 100% of
patients achieved effective asparaginase levels at 48 and 72 hours (n=35 and n=13,
respectively) following the third total dose when given on a Monday, Wednesday, Friday
schedule using the IM route of administration. In a multicenter study characterizing the
pharmacokinetic profile of 25,000 International Units/m2 Erwinaze® given intravenously
over one hour on the same dosing schedule of Monday, Wednesday, Friday for 2
consecutive weeks, 83% (20/24) and 43% (9/21) of evaluable patients achieved an
asparaginase activity level of ≥ 0.1 International Units/mL at 48 post-dose 5 and 72 hours
post-dose 6, respectively.79 No formal drug interaction studies have been performed with
asparaginase Erwinia chrysanthemi.
Toxicity:
Common Occasional Rare
Happens to 21-100 Happens to 5-20 children Happens to < 5 children
children out of every 100 out of every 100 out of every 100
Immediate: Allergic reactions, Local injection site
Within 1-2 days of anaphylaxis, urticaria reactions, fever
receiving drug
Prompt: Pancreatitis, glucose
Within 2-3 weeks, intolerance,
prior to the next thrombosis, hemorrhage,
course transient ischemic attack,
disseminated intravascular
coagulation,
hyperbilirubinemia,
alanine aminotransferase
increased, aspartate
aminotransferase
increased, hyperglycemia,
hyperammonemia,
vomiting, nausea,
abdominal pain,
headache, diarrhea, seizure
Unknown Fetal toxicities and teratogenic effects of L-asparaginase have been noted in
Frequency and animals. It is unknown whether the drug is excreted in breast milk. Adequate,
Timing: well-controlled studies of asparaginase Erwinia chrysanthemi have NOT
been conducted. It is not known whether asparaginase Erwinia chrysanthemi
will cause fetal harm or affect the ability to reproduce. It is not known if
asparaginase Erwinia chrysanthemi is excreted into breast milk. The use of
asparaginase Erwinia chrysanthemi should be avoided in pregnant or
lactating patients.
(L) Toxicity may also occur later.
Guidelines for Administration: See Treatment and Dose Modifications sections of the
protocol.
For intramuscular administration, the contents of each vial should be reconstituted by slowly
adding 1 mL or 2 mL of sterile, preservative-free NS to the inner vial wall. The final
concentration is 10,000 International Units per mL when using 1 mL for reconstitution or
5,000 International Units per mL when using 2 mL for reconstitution. Gently mix or swirl the
contents to dissolve the contents of the vial. Do not shake or invert the vial. The resulting
solution should be clear and colorless. Discard if any particulate matter or protein aggregates
are visible. Withdraw the appropriate dosing volume into a polypropylene syringe
within 15 minutes of reconstitution. Polycarbonate luer-lok syringes from B-D (1 mL) are
also acceptable (personal communication, EUSA Pharma). Discard any unused drug; do not
save or use any unused drug remaining in the vial.
For intravenous use, infuse Erwinia asparaginase in 100 mL of normal saline over 1-2 hours.
Do not infuse other intravenous drugs through the same intravenous line while infusing
Erwinia asparaginase.
Drug Ordering:
In the United States, asparaginase Erwinia chrysanthemi (ErwinazeTM) is distributed by
Accredo Health Group, Inc. An institutional contract with Accredo is required in order to
purchase the product. Accredo’s contact information is as follows:
CANADIAN SITES
Asparaginase Erwinia chrysanthemi is commercially available in Canada. Canadian sites
may purchase the Canadian commercial supply from EUSA via CGF Pharmatech,
Montreal, Quebec, a subsidiary of EUSA (order desk phone: 1-514-343-0344 or 1-866-
343-0344, fax: 1-514-343-0340). CGF requests that a site fax a Purchase Order number.
There is no special fax order form. Shipments are sent Monday to Wednesday only and
usually arrive at the site within 48-72 hours.
Special precautions:
1. Pegaspargase is contraindicated with a history of severe pancreatitis with any prior
asparaginase therapy. Caution should be used if serious thrombosis or hemorrhagic
events have occurred with any prior asparaginase therapy (see Section 5.1)
2. Pegaspargase may affect coagulation factors and predispose to bleeding and/or
thrombosis. Caution should be used when administering any concurrent anticoagulant
therapy.
3. Suggested monitoring during and after administration: Because pegaspargase is long
acting, hypersensitivity reactions may not appear for hours after drug administration.
Monitor vital signs, for signs of fever, chills, or acute allergic reactions including
anaphylaxis. Have medications to treat hypersensitivity reactions readily available at
each administration (e.g., epinephrine, IV corticosteroids, antihistamines). Consider
prescribing an EpiPen® for home use.
Toxicity:
Common Occasional Rare
Happens to 21-100 Happens to 5-20 children Happens to < 5 children
children out of every 100 out of every 100 out of every 100
Immediate: Anorexia, nausea & Abdominal discomfort, Transient blurred vision,
Within 1-2 vomiting diarrhea nasal stuffiness with
days of (acute and delayed) rapid administration,
receiving drug arrhythmias (rapid
infusion), skin rash,
anaphylaxis, SIADH
Prompt: Leukopenia, alopecia, Thrombocytopenia, Cardiac toxicity with
Within 2-3 immune suppression anemia, hemorrhagic high dose (acute – CHF
weeks, prior to cystitis (L) hemorrhagic myocarditis,
the next course myocardial necrosis) (L),
hyperpigmentation, nail
changes, impaired wound
healing, infection
secondary to immune
suppression
1
Delayed: Gonadal dysfunction: Amenorrhea Gonadal dysfunction:
Any time later azoospermia or ovarian failure1 (L),
during therapy oligospermia (prolonged interstitial pneumonitis,
or permanent)1 (L) pulmonary fibrosis2 (L)
Late: Secondary malignancy
Any time after (ALL, ANLL, AML),
completion of bladder carcinoma (long
treatment term use > 2 years),
bladder fibrosis
Unknown Frequency and Timing: Fetal toxicities and teratogenic effects of cyclophosphamide
(alone or in combination with other antineoplastic agents) have been noted in humans. Toxicities
include: chromosomal abnormalities, multiple anomalies, pancytopenia, and low birth weight.
Cyclophosphamide is excreted into breast milk. Cyclophosphamide is contraindicated during
breastfeeding because of reported cases of neutropenia in breast fed infants and the potential for
serious adverse effects.
1
Dependent on dose, age, gender, and degree of pubertal development at time of treatment.
2
Risk increased with pulmonary chest irradiation and higher doses.
(L) Toxicity may also occur later.
Guidelines for Administration: See Treatment and Dose Modifications sections of the
protocol.
Supplier: Commercially available from various manufacturers. See package insert for
further information.
Toxicity:
Common Occasional Rare
Happens to 21-100 Happens to 5-20 Happens to < 5 children
children out of every 100 children out of every 100 out of every 100
Immediate: Hypertension (L), Headache (L), nausea Anaphylaxis,
Within 1-2 days immunosuppression (L) and vomiting, diarrhea angioedema, seizure (L),
of receiving drug arrhythmia, chest pain,
fever, facial flushing
Prompt: Tremor (L), renal Hypomagnesemia (L), Confusion (L),
Within 2-3 dysfunction (acute with hyperlipidemia (L) somnolence (L),
weeks, prior to decrease in GFR, insomnia, depression (L),
the next course impaired urinary anxiety, dizziness, rash,
concentrating ability, and urticaria, acne,
sodium retention) hyperkalemia,
encephalopathy,
hemolytic-uremic
syndrome, cardiac failure,
MI, leukopenia (L),
anemia,
thrombocytopenia,
increased creatinine,
infections,
hyperbilirubinemia (L),
hepatic insufficiency
Delayed: Hirsutism (L) Gingival hyperplasia (L) Tinnitus, vestibular
Any time later disorder, cholelithiasis,
during therapy, cataracts (L),
excluding the gynecomastia, chronic
above conditions renal dysfunction
Late: Lymphoproliferative
Any time after disorders, skin
completion of malignancies
treatment
Unknown Fetal toxic effects of cyclosporine have been noted in animals. Of the reported
Frequency and outcomes of 116 pregnancies in women receiving cyclosporine during
Timing: pregnancy, 90% of whom were transplant patients, and most of whom received
cyclosporine throughout the entire gestational period, the only consistent
patterns of abnormality were premature birth (gestational period of 28 to 36
weeks) and low birth weight for gestational age. Cyclosporine is excreted in
human milk; nursing should be avoided.
(L) Toxicity may also occur later.
Unknown Frequency Fetal toxicities and teratogenic effects of cytarabine have been noted in humans. It
and Timing: is unknown whether the drug is excreted in breast milk.
Toxicity: (Intrathecal)
Common Occasional Rare
Happens to 21-100 Happens to 5-20 children Happens to < 5 children
children out of every 100 out of every 100 out of every 100
Immediate: Nausea, vomiting, fever, Arachnoiditis Rash, somnolence,
Within 1-2 days of headache meningismus,
receiving drug convulsions, paresis
Prompt: Myelosuppression,
Within 2-3 weeks, ataxia
prior to the next
course
Delayed: Necrotizing
Any time later leukoencephalopathy,
during therapy, paraplegia, blindness (in
excluding the above combination with XRT
condition & systemic therapy)
Formulation:
Cytarabine for Injection is available in vials of 100 mg, 500 mg, 1 g, and 2 g containing a
sterile powder for reconstitution. It is also available at a 20 mg/mL concentration with
benzyl alcohol (25 mL per vial) or as a preservative free solution (5 mL, 50 mL per vial),
and at a 100 mg/mL concentration with benzyl alcohol (20 mL vial) or as preservative free
solution (20 mL vial). Hydrochloric acid and/or sodium hydroxide may be added to adjust
the pH. Store at 25°C (77°F); excursions permitted to 15°-30°C (59°-86°F). Cytarabine
solutions should be protected from light.
Guidelines for Administration: See Treatment and Dose Modifications sections of the
protocol.
IV Infusion:
Reconstitute the lyophilized powder with Bacteriostatic Water for Injection or NS injection.
Solution containing bacteriostatic agent should not be used for the preparation of doses
> 200 mg/m2. May be further diluted with dextrose or sodium chloride containing solutions.
May give by IV push injection, by IV infusion, or by continuous infusion.
Low Dose (≤ 200 mg/m²/dose): For administration by IV push, reconstitute to a
concentration of 20-100 mg/mL.
High Dose (≥ 1000 mg/m²/dose): Administer steroid eye drops (dexamethasone or
prednisolone), 2 drops each eye q6h beginning immediately before the first dose and
continuing 24 hours after the last dose. If patient does not tolerate steroid eye drops,
administer artificial tears on a q2-4 hour schedule.
Stability: When reconstituted with Bacteriostatic Water for Injection, cytarabine is stable for
48 hours at room temperature. Solutions reconstituted without a preservative should be used
immediately. Discard if solution appears hazy. Diluted solutions in D5W or NS are stable
for 8 days at room temperature; however, the diluted cytarabine should be used within 24
hours for sterility concerns.
Injection:
Dexamethasone Sodium Phosphate Solution for Injection is available as 4 mg/mL (1 mL,
5 mL, and 30 mL vials) and 10 mg/mL (1 mL and 10 mL vial sizes). Vials are available in
multi-dose vials as well as unit of use vials and syringes. Inactive ingredients vary depending
on manufacturer but include creatinine, sodium citrate, sodium hydroxide to adjust pH,
Water for Injection, sodium sulfite, bisulfite and metabisulfite, methyl and propyl paraben,
benzyl alcohol, and EDTA.
Guidelines for Administration: See Treatment and Dose Modifications section of the
protocol.
Dexamethasone Sodium Phosphate for Injection may be given IV, or IM undiluted. For IV
use, it may be further diluted in dextrose or saline containing solutions. Avoid using benzyl
alcohol-containing dexamethasone solutions in neonates. Diluted solutions that contain no
preservatives should be used within 24 hours, but maintain stability for at least 14 days in
PVC bags at room temperature protected from light.
Supplier:
Commercially available from various manufacturers. See package insert for further
information.
55% of the dose is excreted in the urine as etoposide in 24 hours. The mean renal clearance
of etoposide is 7 to 10 mL/min/m² or about 35% of the total body clearance over a dose
range of 80 to 600 mg/m². Etoposide, therefore, is cleared by both renal and non renal
processes, i.e., metabolism and biliary excretion. The effect of renal disease on plasma
etoposide clearance is not known. Biliary excretion appears to be a minor route of etoposide
elimination. Only 6% or less of an intravenous dose is recovered in the bile as etoposide.
Metabolism accounts for most of the non renal clearance of etoposide.
The maximum plasma concentration and area under the concentration time curve (AUC)
exhibit a high degree of patient variability. Etoposide is highly bound to plasma proteins
(~94%), primarily serum albumin. Pharmacodynamic studies have shown that etoposide
systemic exposure is related to toxicity. Preliminary data suggests that systemic exposure
for unbound etoposide correlates better than total (bound and unbound) etoposide. There
is poor diffusion into the CSF < 5%.
Etoposide phosphate is a water soluble ester of etoposide that is rapidly and completely
converted to etoposide in plasma. Pharmacokinetic and pharmacodynamic data indicate
that etoposide phosphate is bioequivalent to etoposide when it is administered in molar
equivalent doses.
Toxicity:
Common Occasional Rare
Happens to 21-100 Happens to 5-20 children Happens to < 5 children
children out of every 100 out of every 100 out of every 100
Immediate: Nausea, vomiting Anorexia Transient hypotension
Within 1-2 days during infusion;
of receiving anaphylaxis (chills, fever,
drug tachycardia, dyspnea,
bronchospasm,
hypotension)
Prompt: Myelosuppression Thrombocytopenia, Peripheral neuropathy,
Within 2-3 (anemia, diarrhea, abdominal pain, mucositis, hepatotoxicity,
weeks, prior to leukopenia), alopecia asthenia, malaise, rashes chest pain,
next course and urticaria thrombophlebitis,
congestive heart failure,
Stevens-Johnson
Syndrome, exfoliative
dermatitis
Delayed: Dystonia, ovarian failure,
Any time later amenorrhea, anovulatory
during therapy cycles, hypomenorrhea,
onycholysis of nails
Late: Secondary malignancy
Any time after (preleukemic or leukemic
completion of syndromes)
treatment
Unknown Fetal toxicities and teratogenic effects of etoposide have been noted in animals
Frequency and at 1/20th of the human dose. It is unknown whether the drug is excreted in breast
Timing: milk.
Guidelines for Administration: See Treatment and Dose Modifications sections of the
protocol.
Etoposide:
Dilute etoposide to a final concentration ≤ 0.4 mg/mL in D5W or NS. Etoposide infusions
are stable at room temperature for 96 hours when diluted to concentrations of 0.2 mg/mL;
stability is 24 hours at room temperature with concentrations of 0.4 mg/mL. The time to
precipitation is highly unpredictable at concentrations > 0.4 mg/mL. Use in-line filter
during infusion secondary to the risk of precipitate formation. However, the use of an in-
line filter is not mandatory since etoposide precipitation is unlikely at concentrations of
0.1-0.4 mg/mL. Do not administer etoposide by rapid intravenous injection. Slow rate
of administration if hypotension occurs.
Leaching of diethylhexyl phthalate (DEHP) from polyvinyl chloride (PVC) bags occurred
with etoposide 0.4 mg/mL in NS. To avoid leaching, prepare the etoposide solution as close
as possible, preferably within 4 hours, to the time of administration or alternatively as per
institutional policy; glass or polyethylene-lined (non-PVC) containers and polyethylene-
lined tubing may be used to minimize exposure to DEHP.
Etoposide Phosphate:
Reconstitute the 100 mg vial with 5 or 10 mL of Sterile Water for Injection, D5W, NS,
Bacteriostatic Water for Injection with Benzyl Alcohol, or Bacteriostatic Sodium Chloride
for Injection with Benzyl Alcohol for a concentration equivalent to 20 mg/mL or
10 mg/mL etoposide equivalent (22.7 mg/mL or 11.4 mg/mL etoposide phosphate),
respectively. Use diluents without benzyl alcohol for neonates and infants < 2 years of
age or patients with hypersensitivity to benzyl alcohol.
Supplier:
Commercially available from various manufacturers. See package insert for more detailed
information.
CANADIAN SITES
Etopophos® (etoposide phosphate) is not commercially available in Canada. Sites may
purchase and import the USA commercial supply from Bristol Laboratories via an
International Distributor (Pharma Exports LLC, phone: 1-412-885-3700, fax: 1-412-885-
8022, email: [email protected]) under the authority of the protocol’s No Objection Letter
(NOL). Drug Accountability Log (DAL) must record Lot #’s and expiry dates of shipments
received and doses dispensed. Sites may use their own DAL as long as it complies with
all elements of ICH GCP and Division 5 of the Food and Drugs Act. Each site is responsible
for the procurement (import +/- purchase) of Etoposide Phosphate (Etopophos). Sites may
import and manage a single clinical trial supply for multiple protocols as long as each
protocol has an NOL and the protocol the patient is registered on is recorded on the DAL.
Toxicity:
Common Occasional Rare
Happens to 21-100 Happens to 5-20 children out Happens to < 5 children
children out of every 100 of every 100 out of every 100
Immediate: Local irritation at the Allergic reactions (more
Within 1-2 days of injection site, headache common with IV
receiving drug administration than
subq):skin (rash,
urticaria, facial edema),
respiratory (wheezing,
dyspnea) and
cardiovascular
(hypotension,
tachycardia), low grade
fever
Prompt: Mild to moderate Increased: alkaline Splenomegaly, splenic
Within 2-3 weeks, medullary bone pain phosphatase, lactate rupture, rash or
prior to the next dehydrogenase and uric acid, exacerbation of pre-
course thrombocytopenia existing skin rashes,
sickle cell crises in
patients with SCD,
excessive leukocytosis,
Sweet’s syndrome
(acute febrile
neutrophilic
dermatosis)
Delayed: Cutaneous vasculitis,
Anytime later ARDS
during therapy
Late: MDS or AML (confined
Anytime after to patients with severe
completion of chronic neutropenia and
treatment long term
administration)
Unknown Fetal toxicities and teratogenic effects of filgrastim in humans are unknown.
Frequency and Conflicting data exist in animal studies and filgrastim is known to pass the
Timing: placental barrier. It is unknown whether the drug is excreted in breast milk.
Do not dilute with saline-containing solutions at any time; precipitation will occur.
Supplier: Commercially available from various manufacturers. See package insert for
further information
Phase I studies in humans have demonstrated that within several minutes after intravenous
infusion, fludarabine phosphate is converted to the active metabolite, 2-fluoro-ara-A and
becomes undetectable. Therefore, pharmacokinetics studies have focused on 2-fluoro-ara-
A. Fludarabine phosphate 25 mg/m2 infused intravenously over 30 minutes to adult cancer
patients, showed a moderate accumulation of 2-fluoro-ara-A. During a 5-day treatment
schedule, 2-fluoro-ara-A plasma trough levels increased by a factor of about 2.
Fludarabine is widely distributed. The volume of distribution at steady state (Vss) reported
after daily administration of 25mg/m2 for 5 days to adults averaged at 96-98 L/m2. Tissue
distribution studies in animals indicate that the highest concentrations of the drug are in
liver, kidney, and spleen. Although the extent to which fludarabine and/or its metabolites
distribute into the CNS in humans has not been determined to date, severe neurologic
toxicity (e.g., blindness, coma) has been reported in patients receiving the drug, particularly
in high dosages. There is evidence from animal studies that fludarabine distributes into the
CNS and that a toxic metabolite (2-fluoroadenine, possibly formed by bacteria in the GI
tract), can be absorbed systematically via enterohepatic circulation and distributed into
CSF. According to in vitro data, about 19-29% of fludarabine is bound to plasma proteins.
or triphasic; however, reported terminal elimination half-lives have been similar. In adult
cancer patients receiving fludarabine 25 mg/m2 as a 30-minute IV infusion daily for 5 days,
a terminal half-life of about 20 hours was reported. In a limited number of pediatric
patients, the plasma concentration profile of fludarabine exhibited both monoexponential
and biexponential decay, with a mean t1/2 of 10.5 hours in patients with monoexponential
elimination and a t1/2 of 1.2-1.4 and 12.4-19 hours, respectively, in patients with
biexponential elimination.
Renal clearance accounts for about 40% of the total body clearance of fludarabine. Renal
elimination appears to become more important at high dosages of the drug. The dose of
fludarabine needs to be adjusted in patients with moderate renal impairment.
The use of fludarabine in combination with pentostatin is not recommended due to the risk
of severe pulmonary toxicity.
Toxicity:
Common Occasional Rare
Happens to 21- Happens to 5-20 Happens to < 5 subjects out of every
100 subjects out subjects out of every 100
of every 100 100
Immediate: Fever, fatigue, Edema including Anaphylaxis, tumor lysis syndrome,
Within 1-2 weakness, pain, peripheral edema, dehydration*
days of nausea, vomiting, chills, rash, diarrhea,
receiving drug anorexia, cough, rhinitis, diaphoresis,
dyspnea malaise, abdominal
pain, headache, back
pain, myalgia,
stomatitis, flu-like
syndrome
Prompt: Myelosuppressio Weight loss, Sinusitis, dysuria, opportunistic
Within 2-3 n (anemia, gastrointestinal infections and reactivation of latent
weeks, prior to neutropenia, bleeding, viral infections like Epstein-Barr virus
next course thrombocytopeni hemoptysis, (EBV), herpes zoster and John
a), infection paresthesia, allergic Cunningham (JC) virus (progressive
(urinary tract pneumonitis, multifocal leukoencephalopathy
infection, herpes bronchitis, [PML])L, EBV associated
simplex infection, pharyngitis, visual lymphoproliferative disorder,
pneumonia, upper disturbance, hearing panycytopenia (can be prolonged),
respiratory) loss, hyperglycemia pulmonary hypersensitivitya (dyspnea,
cough, hypoxia, interstitial pulmonary
infiltrate), pulmonary toxicity (acute
respiratory distress syndrome
[ARDS], pulmonary fibrosis,
pulmonary hemorrhage, respiratory
distress, respiratory failure),
pericardial effusion, skin toxicity
(erythema multiforme, Stevens-
Johnson syndrome, toxic epidermal
necrolysis, pemphigus), liver failure,
renal failure, hemorrhage, transfusion-
associated graft-versus-host disease
has occurred following transfusion of
nonirradiated blood products,
phlebitis*, sleep disorder*, cerebellar
Guidelines for Administration: See Treatment and Dose Modifications sections of the
protocol.
Supplier: Commercially available from various manufacturers. See package insert for
further information.
1
May be enhanced by systemic therapy such as high dose methotrexate or cytarabine and/or cranial
irradiation.
(L) Toxicity may also occur later.
Guidelines for Administration: See Treatment and Dose Modifications sections of the
protocol.
For intrathecal administration, dilute each agent with 5-10 mL preservative free NS, lactated
ringers or Elliot’s B solution or as per institutional standard of practice. The volume of CSF
removed should be equal to at least half the volume delivered.
Doses
Patient Age (MTX/Hydrocortisone/ Recommended 10% CSF
CSF Volume *
(years) Ara-C) volume volume
Of note: Larger volumes approximating at least 10% of the CSF volume, isovolumetric
delivery, with the patient remaining prone after the procedure may facilitate drug distribution.
These procedures have not been validated in clinical trials. They are allowed but not mandated
for patients on COG studies.
Intrathecal triples are stable in NS for 24 hours at 25ºC but contain no preservative and should
be administered as soon as possible after preparation.
Supplier: Commercially available from various manufacturers. See package insert for
further information.
6.11 LEUCOVORIN CALCIUM (LCV, Wellcovorin®, citrovorum factor, folinic acid) NSC
#003590
The oral form of leucovorin is available as 5 mg, 10 mg, 15 mg, and 25 mg tablets. Inactive
ingredients vary depending on manufacturer but tablet formulations may include: corn
starch, dibasic calcium phosphate, magnesium stearate, pregelatinized starch, lactose,
microcrystalline cellulose, and sodium starch glycolate.
Guidelines for Administration: See Treatment and Dose Modifications sections of the
protocol.
Injection:
Because of the calcium content of the leucovorin solution, no more than 160 mg of
leucovorin should be injected intravenously per minute (16 mL of a 10 mg/mL solution per
minute). IV leucovorin and sodium bicarbonate are incompatible.
Oral:
Oral leucovorin should be spaced evenly (e.g., every six hours) throughout the day and
may be taken without regard to meals. Doses > 25 mg should be given IV due to the
saturation of absorption.
Leucovorin should not be administered < 24 hours after intrathecal injections that contain
methotrexate unless there are special circumstances.
Supplier: Commercially available from various manufacturers. See package insert for
further information.
Toxicity:
Incidence Toxicities
Common
(>20% of Anemia, neutrophil count decreased, white blood cell decreased, platelet count decreased
patients)
Occasional
(4 - 20% of Anorexia, diarrhea, nausea, vomiting, erythematous rash, malaise, oligospermia
patients)
NOTE: the concentration of the commercially available suspension (20 mg/mL) and the
compounded suspension (50 mg/mL) are NOT the same; doses should be prescribed in the
milligrams required, not mL.
Guidelines for Administration: See Treatment and Dose Modifications sections of the
protocol.
Do not give oral mercaptopurine with food or milk. Concurrent milk products can decrease
absorption and mercaptopurine effect is enhanced if given at bedtime on an empty stomach.
If allopurinol is also given, the oral dose of mercaptopurine should be reduced by 67-75%.
Patients with severe myelosuppression should have their thiopurine S-methyltransferase
(TPMT) status and/or their thiopurine metabolite concentrations evaluated, so that the dose
of mercaptopurine can be reduced in patients with a TPMT defect. Patients with the rare
homozygous deficient TPMT phenotype may tolerate only 1/10th to 1/20th the average
mercaptopurine dose. TPMT testing and thiopurine metabolite measurements are
commercially available.
Suspension:
For children unable to swallow the tablets whole, a 50 mg/mL oral suspension can be
compounded. The suspension is prepared by crushing 50 mercaptopurine 50 mg tablets in a
mortar and adding 8.5 mL sterile water for irrigation. The mixture is triturated to form a
smooth paste. Next, 16.5 mL simple syrup (pH=7) are added with continuous mixing and
finally cherry syrup (pH=7.1) is added to a total volume of 50 mL. The suspension is stable
in amber glass bottles at room temperature (19ºC -23ºC) for up to 5 weeks. The suspension
should be shaken well before each use. Procedures for proper handling and disposal of
cytotoxic drugs should be used when preparing the suspension. (Aliabadi HM, Romanick
M, Desai S et al. Effect of buffer and antioxidant on stability of mercaptopurine suspension.
Am J Heath-Syst Pharm. 65:441-7, 2008.)
Supplier: The tablets are commercially available from various manufacturers. In the
United States, the commercially available oral suspension is available through AnovoRx
Distribution, LLC (1-888-470-0904). See package insert for further information. PLEASE
NOTE there is a difference in the concentration of the commercially available (20
mg/mL) and extemporaneously compounded (50 mg/mL) oral suspensions.
Toxicity:
Common Occasional Rare
Happens to 21-100 Happens to 5-20 children Happens to < 5 children out
children out of every 100 out of every 100 of every 100
Immediate: Transaminase elevations Nausea, vomiting, Anaphylaxis, chills, fever,
Within 1-2 days of anorexia dizziness, malaise,
receiving drug drowsiness, blurred vision,
acral erythema, urticaria,
pruritus, toxic epidermal
necrolysis, Stevens-Johnson
Syndrome, tumor lysis
syndrome, seizures1,
photosensitivity
Prompt: Myelosuppression, Alopecia, folliculitis, acne,
Within 2-3 weeks, stomatitis, gingivitis, renal toxicity (ATN,
prior to the next photosensitivity, fatigue increased creatinine/BUN,
course hematuria), enteritis, GI
ulceration and bleeding,
acute neurotoxicity1
(headache, drowsiness,
aphasia, paresis, blurred
vision, transient blindness,
dysarthria, hemiparesis,
decreased reflexes)
diarrhea, conjunctivitis
Delayed: Learning disability1 (L) Pneumonitis, pulmonary
Any time later fibrosis (L), hepatic fibrosis
during therapy, (L), osteonecrosis (L),
excluding the leukoencephalopathy1 (L),
above conditions pericarditis, pericardial
effusions,
hyperpigmentation of the
nails
Late: Progressive CNS
Any time after the deterioration1
completion of
therapy
Unknown Methotrexate crosses the placenta. Fetal toxicities and teratogenic effects of
Frequency and methotrexate have been noted in humans. The toxicities include: congenital defects,
Timing: chromosomal abnormalities, severe newborn myelosuppression, low birth weight,
abortion, and fetal death. Methotrexate is excreted into breast milk in low
concentrations.
1
May be enhanced by HDMTX and/or cranial irradiation.
(L) Toxicity may also occur later.
Toxicity:
Oral administration: Food or milk delays absorption and reduces peak concentration.
Methotrexate for oral use should preferentially be given on an empty stomach, 1 hour
before or 2 hours after food or milk and at the same time each day. Methotrexate injection
diluted in water can be used for oral administration (Marshall PS, Gertner E. Oral
administration of an easily prepared solution of injectable methotrexate diluted in water: a
comparison of serum concentrations vs methotrexate tablets and clinical utility. J
Rheumatol 23:455-8, 1996).
For IM/IV use: Powder for injection: Dilute 1000 mg vial with 19.4 mL of preservative
free SWFI, D5W or NS to a 50 mg/mL concentration. The powder for injection may be
further diluted in NS or dextrose containing solutions to a concentration of ≤ 25mg/mL for
IV use.
The 25 mg/mL solution may be given directly for IM administration or further diluted in
Saline or Dextrose containing solutions for IV use. Do not use the preserved solution for
high dose methotrexate administration due to risk of benzyl alcohol toxicity.
Methotrexate dilutions are chemically stable for at least 7 days at room temperature but
contain no preservative and should be used within 24 hours. Diluted solutions especially
those containing bicarbonate exposed to direct sunlight for periods exceeding 4 hours
should be protected from light.
High dose methotrexate requires alkalinization of the urine, adequate hydration and
leucovorin rescue. Avoid probenecid, penicillins, cephalosporins, aspirin, proton pump
inhibitors, and NSAIDS as renal excretion of MTX is inhibited by these agents.
For intrathecal administration, dilute with 5-10 mL preservative free NS, lactated Ringer’s,
or Elliot’s B solution as per institutional standard of practice. The volume of CSF removed
should be equal to at least half the volume delivered.
Note: When IT therapy and ID MTX are scheduled for the same day, deliver the IT therapy
within 6 hours of the beginning of the IV MTX infusion (hour -6 to +6, with 0 being the
start of the MTX bolus).
For IT administration, use the preservative free formulation. The volume to be given IT
should be in the range of 5-10 mL. The volume of CSF removed should be equal to at least
half the volume delivered (see drug monograph).
Note: Larger volumes approximating at least 10% of the CSF volume, isovolumetric delivery,
with the patient remaining lying down after the procedure may facilitate drug distribution.
These procedures have not been validated in clinical trials. They are allowed but not mandated
for patients on COG studies.
Of Note: Larger volumes approximating at least 10% of the CSF volume, isovolumetric
delivery, with the patient remaining prone after the procedure may facilitate drug distribution.
These procedures have not been validated in clinical trials. They are allowed but not mandated
for patients on COG studies.
Diluted methotrexate for intrathecal administration is stable for 24 hours at 25ºC but contains
no preservative and should be administered as soon as possible after preparation.
Supplier: Commercially available from various manufacturers. See package insert for
further information.
Toxicity:
Common Occasional Rare
Happens to 21- Happens to 5- Happens to
100 children out of 20 children out of < 5 children out of
every 100 every 100 every 100
Immediate: Nausea, vomiting, Abdominal pain, back Anaphylaxis,
Within 1-2 days of diarrhea, fever, pain, headache, angioedema, cardiac
receiving drug anorexia, green blue phlebitis, constipation arrhythmias1
discoloration of the (bradycardia), seizures,
urine and/or sclera extravasation reactions
rare but if occur can
lead to: (erythema,
swelling, pain, burning
and/or blue
discoloration of the
skin and rarely tissue
necrosis), tumor lysis
Prompt: Myelosuppression (L),Transient elevation of Rash, conjunctivitis,
Within 2-3 weeks, mucositis /stomatitis,
LFTs, pruritus with (GI) hemorrhage,
prior to the next course immunosuppression, desquamation of the interstitial pneumonitis
alopecia, fatigue skin due to progressive
dryness
Delayed: Amenorrhea, menstrual Cardiotoxicity CHF, hepatotoxicity
Any time later during disorders, temporary (decreased LVEF)2 (L)
therapy reduction in sperm
count
Late: Secondary malignancy
Any time after
completion of
treatment
Unknown Frequency Fetal toxicities and teratogenic effects of mitoxantrone have been noted
and Timing: in animals. Toxicities include: low birth weight and prematurity.
Mitoxantrone is excreted in human milk and significant concentrations
(18 ng/mL) have been reported for 28 days after the last administration.
1Rarely clinically significant.
2Risk increases with chest radiation and prior anthracycline dosage
(L) Toxicity may also occur later.
Guidelines for Administration: See Treatment and Dose Modifications sections of the
protocol.
Mitoxantrone must be diluted prior to injection. DO NOT GIVE IV PUSH. The dose of
mitoxantrone should be diluted in at least 50 mL or to a concentration ≤ 0.5 mg/mL with
either NS or D5W. The dilution is stable at room temperature for 48 hours with no loss of
potency. Admixture with heparin may result in precipitation. Mitoxantrone is an irritant:
Care should be taken to avoid extravasation; the use of a central line is suggested. If it is
known or suspected that subcutaneous extravasation has occurred, it is recommended that
intermittent ice packs be placed over the area of extravasation and that the affected
extremity be elevated. Because of the progressive nature of extravasation reactions, the
area of injection should be frequently examined and surgery consultation obtained early if
there is any sign of a local reaction.
Supplier: Commercially available. See package insert for more detailed information.
MMF has been used in a variety of solid organ and hematopoietic stem cell transplant
settings for the prevention of acute rejection. MMF is a prodrug that after oral
administration is rapidly hydrolyzed, primarily by the liver, to the biologically active
metabolite mycophenolic acid. MPA is metabolized principally by glucuronyl transferase
to form the pharmacologically inactive phenolic glucuronide of MPA (MPAG). In vivo,
MPAG is converted to MPA via enterohepatic recirculation. Mycophenolic acid inhibits
nucleic acid synthesis and produces a potent, noncompetitive, and reversible inhibition of
inosine monophosphate dehydrogenase (IMPDH), blocking the de novo synthesis of
guanosine nucleotides without being incorporated into DNA. Both T and B lymphocytes
rely on this de novo pathway for purine synthesis. As a result, the proliferative responses
of T and B lymphocytes to both mitogenic and allospecific stimulation are inhibited. Other
rapidly dividing cell lines are capable of recycling purine nucleotides via the "salvage"
pathway, which is not blocked by mycophenolic acid.
In vitro and in vivo studies have demonstrated the ability of mycophenolic acid to block
proliferative responses of T and B lymphocytes, inhibit antibody formation and the
generation of cytotoxic T-cells, and suppress antibody formation by B lymphocytes.
Mycophenolic acid prevents the glycosylation of lymphocyte and monocyte glycoproteins
that are involved in intercellular adhesion of these cells to endothelial cells, and may inhibit
Mycophenolate mofetil can cause fetal harm when administered to a pregnant woman. Use
of MMF during pregnancy is associated with an increased risk of first trimester pregnancy
loss and an increased risk of congenital malformations, especially external ear and other
facial abnormalities including cleft lip and palate, and anomalies of the distal limbs, heart,
esophagus, and kidney. According to the package labeling, the National Transplantation
Pregnancy Registry (NTPR) presents data on 33 MMF-exposed pregnancies in 24
transplant patients. Of these, there were 15 spontaneous abortions and 18 live-born infants.
Four of the 18 infants had structural malformations (22%). In postmarketing data (collected
1995-2007) of 77 women exposed to systemic MMF during pregnancy, 25 had spontaneous
abortions and 14 had a malformed infant or fetus. Six of 14 malformed offspring had ear
abnormalities. Because these postmarketing data are reported voluntarily, it is not always
possible to reliably estimate the frequency of particular adverse outcomes. These
malformations seen in offspring were similar to findings in animal reproductive toxicology
studies. In animal reproductive toxicology studies, there were increased rates of fetal
resorptions and malformations in the absence of maternal toxicity. Female rats and rabbits
received MMF doses equivalent to 0.02 to 0.9 times the recommended human dose for
renal and cardiac transplant patients, based on body surface area conversions. In rat
offspring, malformations included anophthalmia, agnathia, and hydrocephaly. In rabbit
offspring, malformations included ectopia cordis, ectopic kidneys, diaphragmatic hernia,
and umbilical hernia. If this drug is used during pregnancy, or if the patient becomes
pregnant while taking this drug, the patient should be apprised of the potential hazard to
the fetus.
Women of childbearing potential should have a negative serum or urine pregnancy test
with a sensitivity of at least 25 mIU/mL within 1 week prior to beginning therapy
(manufacturer’s recommendation). MMF should not be initiated until a negative pregnancy
test report is obtained. Women of childbearing potential taking MMF must receive
contraceptive counseling and use effective contraception. It is recommended by the
manufacturer that the patient begin using two chosen methods of contraception 4 weeks
prior to starting MMF, unless abstinence is the chosen method. She should continue
contraceptive use during therapy and for 6 weeks after stopping MMF. Patients should be
aware that MMF reduces blood levels of the hormones in the oral contraceptive pill and
could theoretically reduce its effectiveness.
Inactive ingredients in the 250 mg capsules include the following: croscarmellose sodium,
magnesium stearate, povidone (K-90) and pregelatinized starch. The capsule shells contain
black iron oxide, FD&C blue #2, gelatin, red iron oxide, silicon dioxide, sodium lauryl
sulfate, titanium dioxide, and yellow iron oxide.
Inactive ingredients in the 500 mg tablets include black iron oxide, croscarmellose sodium,
FD&C blue #2 aluminum lake, hydroxypropyl cellulose, hydroxypropyl methylcellulose,
magnesium stearate, microcrystalline cellulose, polyethylene glycol 400, povidone (K-90),
red iron oxide, talc, and titanium dioxide; may also contain ammonium hydroxide, ethyl
alcohol, methyl alcohol, n-butyl alcohol, propylene glycol, and shellac.
Inactive ingredients in the powder for oral suspension include aspartame, citric acid
anhydrous, colloidal silicon dioxide, methylparaben, mixed fruit flavor, sodium citrate
dihydrate, sorbitol, soybean lecithin, and xanthan gum.
Guidelines for Administration: See Treatment and Dose Modifications sections of the
protocol.
Preparation:
To prepare the oral suspension, add 47 mL of water to the bottle and shake well for
approximately 1 minute. Add another 47 mL of water to the bottle and shake well for an
additional minute. The final concentration is 200 mg/mL of mycophenolate mofetil. Avoid
inhalation or direct contact with skin or mucous membranes of the dry powder or the
constituted suspension. If such contact occurs, wash thoroughly with soap and water; rinse
eyes with water.
To prepare the intravenous injection, reconstitute the contents of each vial with 14 mL of
D5W. Dilute the contents of a vial with D5W to a final concentration of 6 mg/mL. Each
vial is vacuum-sealed; if a lack of vacuum is noted during preparation, the vial should not
be used.
Stability:
Oral formulations should be stored at 25°C (77°F); excursions are permitted to 15°C to
30°C (59°F to 86°F). Protect from moisture and light. Once reconstituted, the oral solution
may be stored at room temperature or under refrigeration. Do not freeze. The mixed
suspension is stable for 60 days.
Store intact vials and diluted solutions at 25°C (77°F); excursions permitted to 15°C to
30°C (59°F to 86°F). Do not freeze. Begin infusion within 4 hours of reconstitution.
Administration:
Oral formulations of MMF should be administered on an empty stomach to avoid
variability in MPA absorption. The oral solution may be administered via a nasogastric
tube (minimum 8 French, 1.7 mm interior diameter). Do not mix the oral suspension with
other medications. Some products may contain phenylalanine; refer to the package labeling
for additional details.
The intravenous solution should be given over at least 2 hours. Do not administer by rapid
or bolus injection.
Supplier:
Commercially available from various manufacturers. See package insert for further
information.
enzyme could not be detected in the urine. The half-life is independent of the dose
administered, disease status, renal or hepatic function, age, or gender. In a study of newly
diagnosed pediatric patients with ALL who received either a single intramuscular injection
of pegaspargase (2500 IU/m2), E. coli L-asparaginase (25000 IU/m2), or Erwinia
(25000 IU/m2), the plasma half-lives for the three forms of L-asparaginase were:
5.73 ± 3.24 days, 1.24 ± 0.17 days, and 0.65 ± 0.13 days respectively. The plasma half-life
of pegaspargase is shortened in patients who are previously hypersensitive to native
L-asparaginase as compared to non-hypersensitive patients. L-asparaginase is cleared by
the reticuloendothelial system and very little is excreted in the urine or bile. Cerebrospinal
fluid levels are < 1% of plasma levels.
Toxicity:
Common Occasional Rare
Happens to 21-100 children Happens to 5-20 children Happens to < 5 children out
out of every 100 out of every 100 of every 100
Immediate: Allergic reactions (total Allergic reactions Anaphylaxis,
Within 1- likelihood of local, and or (total likelihood of hyper/hypotension,
2 days of systemic reaction local, and or systemic tachycardia, periorbital
receiving especially if previous reaction if no previous edema, chills, fever,
drug hypersensitivity reaction to hypersensitivity dizziness, dyspnea,
native asparaginase), pain reaction to native bronchospasm, lip edema,
at injection site, weakness, asparaginase), rash arthralgia, myalgia,
fatigue, diarrhea urticaria, mild
nausea/vomiting,
abdominal pain, flatulence,
somnolence, lethargy,
headache, seizures (L),
hyperuricemia
Prompt: Hyperammonemia (L), Hyperglycemia, Hemorrhage (L), DIC,
Within 2- coagulation abnormalities abnormal liver thrombosis, anorexia,
3 weeks, with prolonged PTT, PT function tests, weight loss, CNS ischemic
prior to the and bleeding times pancreatitis (L), attacks, edema, azotemia
next course (secondary to decreased increased serum and decreased renal
synthesis of fibrinogen, lipase/amylase function, mild leukopenia,
AT-III & other clotting granulocytopenia,
factors) (L) thrombocytopenia,
pancytopenia, hemolytic
anemia, infections (sepsis
with/without septic shock,
subacute bacterial
endocarditis [SBE], URI),
CNS changes including
irritability, depression,
confusion, EEG changes,
hallucinations, coma and
stupor, paresthesias,
hypertriglyceridemia,
hyperlipidemia, Parkinson-
like syndrome with tremor
and increase in muscular
tone, hyperbilirubinemia,
chest pain
Delayed: Renal failure, urinary
Any time frequency, hemorrhagic
later during cystitis, elevated creatinine
therapy and BUN, fatty liver
deposits, hepatomegaly,
liver failure
Unknown Animal reproduction studies have not been conducted with pegaspargase. It is not
Frequency known whether pegaspargase can cause fetal harm when administered to a pregnant
and woman or can affect reproduction capacity. However, fetal toxicities and teratogenic
Timing: effects of asparaginase have been noted in animals. It is unknown whether the drug
is excreted in breast milk.
(L)Toxicity may also occur later.
Guidelines for Administration: See Treatment and Dose Modifications sections of the
protocol.
For IM administration: the volume at a single injection site should be limited to 2 mL. If
the volume to be administered is greater than 2 mL, multiple injection sites should be used.
Have available during and after the infusion: antihistamine, epinephrine, oxygen, and IV
corticosteroids. Observe patient for ONE hour after administration for signs of
hypersensitivity reactions.
CANADIAN SITES
Pegaspargase is not commercially available in Canada. Sites may purchase and import the
USA commercial supply directly from Sigma Tau Pharmaceuticals under the authority of
the protocol’s No Objection Letter (NOL). The Canadian Senior Medical Officer (SMO)’s
office is responsible for coordinating the “Fax Back” for all lot numbers and expiry dates
to Health Canada’s Biologics and Genetic Therapies Directorate for approval for use in
Canada on behalf of all Canadian sites. A list of approved lot numbers is distributed to all
Canadian sites by the SMO’s office. If an unapproved lot is received from Sigma Tau
Pharmaceuticals, quarantine the lot and contact the COG Canada Regulatory Affairs Office
at 1-780-492-7064. Note: Pegaspargase may have orders placed and Drug Accountability
Toxicity:
Common Occasional Rare
Happens to 21- Happens to 5- Happens to < 5 children out
100 children out of 20 children out of of every 100
every 100 every 100
Immediate: Headache (L), Chest pain Anaphylaxis with the
Within 1-2 days hypertension (L), nausea, injection, allergic reaction,
of receiving drug vomiting, anorexia, hypotension, asthma,
immunosuppression (L), dyspnea, increased cough,
diarrhea, constipation, flu like syndrome, pleural
fever effusion, seizure (L),
tachycardia, angina
Prompt: Tremor (L), ), renal Alopecia, dizziness, Dyspepsia, dysphagia,
Within 2- dysfunction (acute with elevated LFTs, UTI, gastritis, esophagitis,
3 weeks, prior to decrease in GFR, peripheral edema, flatulence, CNS
the next course impaired urinary rash, pruritus, abnormalities (confusion
concentrating ability, and hyperlipidemia, (L), somnolence (L),
sodium retention), hypercholesteremia depression (L), anxiety,
elevated creatinine/BUN, anxiousness, abnormal
anemia, insomnia, dreams, emotional labiality,
asthenia, pain hallucinations, psychosis,
(abdominal, back, pain), hypertonia, incoordination,
hyperglycemia, neuropathy, nervousness
hypomagnesemia (L), encephalopathy),
hyper/hypokalemia (L), coagulation disorder,
hypophosphatemia, leukopenia (L),
paresthesia thrombocytopenia,
polycythemia, anemia,
leukocytosis, infections
(bacterial, fungal, viral –
sepsis, cellulites, fungal
dermatitis, herpes simplex,
sinusitis, pharyngitis,
abscess, pneumonia,
bronchitis, peritonitis),
hyperbilirubinemia (L),
thrombosis, phlebitis,
arthralgia, myalgia,
electrolyte abnormalities
Delayed: Acne, exfoliative dermatitis,
Any time later skin discoloration,
during therapy, photosensitivity reaction,
excluding the skin ulcer, delayed wound
above conditions healing, hirsutism
(hypertrichosis) (L), gingival
hyperplasia, abnormal
vision, amblyopia, ear pain,
otitis, tinnitus, GI
hemorrhage, GI perforation,
cholelithiasis, cholestatic
jaundice, chronic renal
dysfunction, renal failure,
post-transplant diabetes
mellitus (L), myocardial
hypertrophy, elevated liver
Injection:
Tacrolimus Injection must be diluted with NS or D5W before use to a concentration
between 0.004 mg/mL and 0.02 mg/mL. Diluted infusion solution should be stored in glass
or polyethylene containers and should be discarded after 24 hours. The polyoxyethylated
castor oil contained in the concentrate for intravenous infusion can cause phthalate
stripping from PVC. It is strongly recommended that glass bottles and non-PVC tubing
be used to minimize patient exposure to DEHP. Due to the chemical instability of
tacrolimus in alkaline media, tacrolimus injection should not be mixed or co-infused with
solutions of pH 9 or greater (e.g., ganciclovir or acyclovir).
Monitor closely for an acute allergic reaction for the first 30 minutes and at frequent
intervals thereafter.
Oral:
Administer at a consistent time of day and at consistent intervals with regard to meals.
Tacrolimus may be given with food as long as it is given the same way each time; however;
administration with food significantly decreases the rate and extent of absorption.
Grapefruit juice should be avoided during the entire course of tacrolimus administration.
Supplier: Commercially available from various manufacturers. See package insert for
further information.
Peak levels occur 2 to 4 hours after oral administration with a median half-life is about
90 minutes (range: 25-240 minutes). Very little unchanged drug is excreted renally.
Toxicity:
Common Occasional Rare
Happens to 21- Happens to 5- Happens to < 5 children out of
100 children out of 20 children out of every 100
every 100 every 100
Immediate: Anorexia, nausea, Urticaria, rash, hyperuricemia
Within 1-2 days of vomiting, diarrhea,
receiving drug malaise
Prompt: Myelosuppression Toxic hepatitis (L), increased
Within 2-3 weeks, SGOT (AST)/SGPT (ALT),
prior to next course ataxia, mucositis
Delayed: Hepatic fibrosis(L), sinusoidal
Anytime later during obstruction syndrome (SOS,
therapy formerly VOD) (L),
hyperbilirubinemia
Unknown Frequency Fetal toxicities and teratogenic effects of thioguanine have been noted in animals.
and Timing: It is unknown whether the drug is excreted in breast milk.
(L) Toxicity may also occur later.
For patients unable to swallow tablets, a 20 mg/mL oral suspension may be compounded.
Crush fifteen (n=15) 40 mg tablets in a mortar and reduce to a fine powder. Add 10 mL
methylcellulose 1% in incremental proportions and mix to a uniform paste. Transfer to a
graduated cylinder, rinse mortar with simple syrup, and add quantity of simple syrup
sufficient to make 30 mL. Dispense in an amber glass bottle and label "shake well" and
"refrigerate". If methylcellulose is not available, substitute 15 mL of Ora-Plus in place of
the methylcellulose and qs with Ora-Sweet (in place of simple syrup) to a final volume of
30 mL. Both preparations are stable for 63 days at 19°C – 23°C. (Aliabadi HM, Romanick
M, Somayah V, et al. Stability of compounded thioguanine oral suspensions. Am J Health
Syst Pharm 2011;68:1278. Dressman JB, Poust RI. Stability of Allopurinol and Five
Antineoplastics in Suspension. Am J Hosp Pharm 1983;40(4):616-8.)
Guidelines for Administration: See Treatment and Dose Modifications sections of the
protocol.
Supplier: Commercially available. See package insert for more detailed information.
Toxicity:
Common Occasional Rare
Happens to 21-100 children Happens to 5-20 children Happens to < 5 children
out of every 100 out of every 100 out of every 100
Immediate: Nausea, vomiting, anorexia, Pain at the injection site, Anaphylaxis, laryngeal
Within 1-2 days fatigue, weakness dizziness, headache, edema, wheezing, hives
of receiving blurred vision, abdominal
drug pain, contact dermatitis,
rash
Prompt: Myelosuppression; at At higher doses in Febrile reaction,
Within 2- higher doses in conditioning regimens conjunctivitis, dysuria,
3 weeks, prior to conditioning regimens for for BMT: urinary retention
next course BMT: mucositis, encephalopathy
esophagitis (inappropriate behavior,
confusion, somnolence),
increased liver
transaminases, increased
bilirubin,
hyperpigmentation of the
skin (bronzing effect)
Delayed: Gonadal Alopecia, secondary
Anytime later dysfunction/infertility, malignancy
during therapy, azoospermia, amenorrhea
excluding the
above
conditions
Unknown Fetal and teratogenic toxicities: Carcinogenic and teratogenic effects of thiotepa
Frequency and have been noted in animal models at doses ≤ to those used in humans. It is not
Timing: known if thiotepa is excreted into human breast milk.
(L) Toxicity may also occur later.
Guidelines for Administration: See Treatment and Dose Modifications sections of the
protocol.
Reconstitute Thiotepa for Injection with 1.5 mL of Sterile Water for Injection resulting in
a drug concentration of approximately 10 mg/mL. (As per manufacturer’s information:
Actual content per vial 15.6 mg; withdrawable amount 14.7 mg/1.4 mL; approximate
reconstituted concentration: 10.4 mg/mL). When reconstituted with Sterile Water for
Injection, solutions of thiotepa should be stored at refrigerated temperatures 2°-8°C (36°-
46°F) protected from light and used within 8 hours. The reconstituted solution is hypotonic
and should be further diluted with Sodium Chloride Injection (0.9% NaCl) prior to use.
Thiotepa at a concentration of 1-5mg/mL in 0.9% NaCl is stable for 24 hours at room
temperature. At concentration of 0.5mg/mL it is stable for only one hour and stability
decreases significantly at concentrations of less than 0.5mg/mL. Therefore, solutions
diluted to 0.5mg/mL should be used immediately
In order to eliminate haze, filter solutions through a 0.22 micron filter [Polysulfone
membrane (Gelman’s Sterile Aerodisc®, Single Use) or triton-free mixed ester of
cellulose/PVC (Millipore’s MILLEX®-GSFilter Unit)] prior to administration. Filtering
does not alter solution potency. Reconstituted solutions should be clear. Solutions that
remain opaque or precipitate after filtration should not be used.
When thiotepa is given in bone marrow transplant doses, bath the patient and change linen
frequently (≥ 2 baths/day) to avoid the contact dermatitis and discoloration of the skin that
is seen with high dose.
CANADIAN SITES
Thiotepa is not commercially available in Canada. Sites may purchase and import thiotepa
through the following two routes:
Commercial supply may be purchased from the USA through Bedford Laboratories via an
International Distributor (Pharma Exports LLC; Contact: Pete Bigley, phone: 1-412-885-
3700, fax: 1-412-885-8022, email: [email protected] or [email protected])
under the authority of the protocol’s No Objection Letter (NOL).
Commercial supply may also be purchased through Adienne Pharma & Biotec, Bergama,
Italy (tel: +39.035.402.872, fax: +39.035.264.207, email: [email protected])
under the authority of the protocols’ No Objection Letter (NOL). They have available a
commercial 15mg and 100mg vial. Consult the product monograph for appropriate
reconstitution and infusion preparation instructions.
NOTE: A “Fax Notification” of the lot number and expiry date of each lot is no longer
required by Health Canada (effective: January 2011). Canadian sites must keep a drug
accountability log recording manufacturer, lot number and expiry date of all vials of
thiotepa. Separate accountability logs must be kept for different manufacturers and
strengths. Thiotepa may have orders placed and Drug Accountability Logs maintained on
a multiple protocol basis (Multiple Protocol—Imported Therapeutic) as long as each
protocol has a NOL and the DAL clearly indicates on which protocol the subject is
registered.
Toxicity:
Common Occasional Rare
Happens to 21-100 children Happens to 5-20 children Happens to < 5 children
out of every 100 out of every 100 out of every 100
Immediate: Jaw pain, headache Extravasation (rare) but if
Within 1-2 days occurs = local ulceration,
of receiving drug shortness of breath, and
bronchospasm
Prompt: Alopecia, constipation Weakness, abdominal Paralytic ileus, ptosis,
Within 2- pain, mild brief diplopia, night blindness,
3 weeks, prior to myelosuppression hoarseness, vocal cord
the next course (leukopenia, paralysis, SIADH, seizure,
thrombocytopenia, defective sweating
anemia)
Delayed: Loss of deep tendon Peripheral paresthesias Difficulty walking or
Any time later reflexes including numbness, inability to walk;
during therapy tingling and pain; sinusoidal obstruction
clumsiness; wrist drop, syndrome (SOS, formerly
foot drop, abnormal gait VOD) (in combination);
blindness, optic atrophy;
urinary tract disorders
(including bladder atony,
dysuria, polyuria, nocturia,
and urinary retention);
autonomic neuropathy
with postural hypotension;
8th cranial nerve damage
with dizziness, nystagmus,
vertigo and hearing loss
Unknown Fetal toxicities and teratogenic effects of vincristine (either alone or in
Frequency and combination with other antineoplastic agents) have been noted in humans. The
Timing: toxicities include: chromosome abnormalities, malformation, pancytopenia,
and low birth weight. It is unknown whether the drug is excreted in breast milk.
The World Health Organization, the Institute of Safe Medicine Practices (United States)
and the Safety and Quality Council (Australia) all support the use of minibag rather than
syringe for the infusion of vincristine. The delivery of vincristine via either IV slow push
or minibag is acceptable for COG protocols. Vincristine should NOT be delivered to the
patient at the same time with any medications intended for central nervous system
administration. Vincristine is fatal if given intrathecally.
Supplier: Commercially available from various manufacturers. See package insert for
more detailed information.
Note: Medication errors have occurred due to confusion between vinCRIStine and
vinBLAStine. VinCRIStine is available in a liposomal formulation (vinCRIStine sulfate
liposomal injection, VSLI, Marqibo®). The conventional and liposomal formulations are
NOT interchangeable; use of the liposomal formulation is not permitted in this trial.
7.1b LR Patients
STUDIES TO BE Baseline Block 1 Block 2 Block 3, Blinatumomab Maintenance and
OBTAINED blocks, Continuation post-therapy
Hx/PE with VS/Wt X start of start of phase* every 28 days
(BSA) phase
CBC/diff/plts X weekly weekly weekly every 28 days
Bilirubin, ALT, X weekly weekly weekly every 28 days
creatinine, BUN
Local Bone Marrow X1 end of end of
(BM) Evaluation phase phase
Bone Marrow (BM) for end of end of
central flow MRD2 phase+ phase++
Bone Marrow (BM) for X2,7
Immunophenotyping
CSF cell count and X with with with each IT with each IT
cytospin each IT each IT
Absolute lymphocyte At end of each 12
count with T and B week maintenance
subset quantification cycle, and every 3
months after
completion of
therapy for 1 year
Peripheral Blood for Blinatumomab Cycle 1:Day
Pharmacokinetics (PK) 6
2 and Day 14
Peripheral Blood for • Prior to (Hour 0) start of Prior to start of
Immunogenicity first blinatumomab Maintenance
infusion (Cycle 1) Cycle 1 therapy
• End of Cycle 2$
Echocardiogram X
Pregnancy Test3 X
Testicular Biopsy X4 X5
1 BM evaluation to confirm relapse and/or detect marrow disease in presumed isolated extramedullary relapse patients should
include morphology, immunophenotyping & cytogenetics/FISH. Cytogenetic/FISH analysis must be performed at a COG
approved cytogenetics lab and cases will be reviewed retrospectively by the COG Cytogenetics Committee. See Section
13.2 for details.
2 See Section 13.3 for details on shipping and handling
3 Female patients of childbearing potential require a negative pregnancy test prior to starting treatment; sexually active patients
7.1c HSCT
Observation Prior to 1-2 3 mo 6 mo 9 mo 1 yr. 2 3 4 5 At
transplant weeks Post- Post- Post- Post- yrs. yrs. yrs. yrs. subsequent
(Tx) after Tx Tx Tx Tx Post- Post- Post- Post- relapse
ANC> Tx Tx Tx Tx
500/µL
Bone X+++ X* X**
Marrow
(BM) for
central
MRD1
CSF cell X X
count and
cytospin
CBC, X X X X X X X X
differential,
platelets
BUN, X
Creatinine
LFT X X X X X X X
Pulmonary X
function test
Performance X X X X X X X X X X X
score
1 See
Section 13.3 for details on shipping and handling; (see Section 7.2)
+++pre-HSCT evaluation
*Day 30 (25-30)
**Day100 (+/- 3 weeks)
This table only includes evaluations necessary to answer the primary and secondary aims. Obtain other studies as
indicated for good clinical care.
Peripheral blood:
• Prior to (Hour 0) start of first blinatumomab
infusion
• During (Hour 6, Hour 12, Day 2, Day 7, Day 14,
Day 21) first blinatumomab infusion
•
* For cases with a limited amount of tissue available for analysis, please prioritize specimen for
tissue banking.
7.3 At Relapse
Patients who relapse and have consented to cell banking at time of enrollment should
have samples of bone marrow sent to the Molecular Reference Laboratory for cell
banking (see Section 13.1.)
7.4 Follow-up
See COG Late Effects Guidelines for recommended post treatment follow-up:
http://www.survivorshipguidelines.org/
Note: Follow-up data are expected to be submitted per the Case Report Forms (CRFs)
schedule.
8.0 CRITERIA FOR REMOVAL FROM PROTOCOL THERAPY AND OFF STUDY
CRITERIA
Patients who are off protocol therapy are to be followed until they meet the criteria for Off
Study (see below). Follow-up data will be required unless consent was withdrawn.
8.2 Off Study Criteria
a) Death.
b) Lost to follow-up.
c) Patient enrollment onto another COG study with tumor therapeutic intent (e.g., at
recurrence).
d) Withdrawal of consent for any further data submission.
e) Tenth anniversary of the date the patient was enrolled on this study.
who are event-free. DFS event date will be set at Day 1 if patients are deemed as
TF after randomization.
Another primary objective (LR Randomization) of AALL1331 is to compare DFS
of LR relapse patients randomized to chemotherapy alone (Control: Block 3,
Continuation 1, Continuation 2 and Maintenance) vs.
blinatumomab/chemotherapy (Experimental: blinatumomab, Continuation 1,
blinatumomab, Continuation 2, blinatumomab, Maintenance). DFS is defined as
time from start of randomization to first event (relapse, second malignant
neoplasm, remission death) or last follow up for those who are event-free.
# randomized on HR/IR
61 24 85
Randomization (per arm)
Rate of not dropping out
63% 75%
prior to end Block 2
# evaluable for end Block 2
38 18 56
MRD testing
Rate of not dropping out
89% 88%
prior to end Block 3
# evaluable for end Block 3
34 16 50
MRD testing
Further, we anticipate that the rates of MRD ≥ 0.01% in the control arm
will be 67% (67% for early and 68% for late) at the end of Block 2 and
47% (43% for early and 55% for late) at the end of Block 3. A two-sample
Fisher’s exact test of proportions (one-sided, alpha = 5%) will be used to
test the hypothesis that the proportion of patients who are MRD+ on each
experimental arm (with blinatumomab) is smaller than the control arm
(without blinatumomab) at the end of Block 2 and 3. The corresponding
power to detect various degrees of reduction in MRD+ rates is shown in
the table below:
2 57 67% 5 8.77%
3 85 100% 7 8.24%
The potential for late adverse effects due to long term depletion of CD19+
normal lymphocytes following blinatumomab treatment is unknown. We
will monitor for lymphocyte recovery and late events related to delayed
recovery. Since HR/IR/TF patients go to HSCT soon after blinatumomab,
it would be difficult to make sense of lymphocyte recovery/late events
after blinatumomab for them. However, the LR patients that are
randomized to +/- blinatumomab and do not proceed to HSCT serve as an
excellent population to monitor for these delayed adverse effects. For all
LR patients, we will collect absolute lymphocyte counts
(ALC)/lymphocyte subset counts (total T and B cells) and data regarding
late infections and progressive multifocal leukoencephalopathy (PML)
after each 12-week maintenance cycle and every 3 months after
completion of therapy for 1 year. The peripheral B-cell recovery rates and
rates of late events by randomized arms will be monitored and compared
9.3.3 Pilot intervention for very high risk subset of HSCT recipients based on MRD and aGVHD
2 24 67% 4 16.67%
3 36 100% 4 11.110%
The intervention will be rejected for excessive toxicity death if the number
of observed toxic deaths is greater than or equal to the corresponding
boundary at any interim analysis. The actual power (based on exact
binomial test) of declaring the intervention too toxic are approximately
88.5%, 71.1%, 41.6% and 10.9% when the true toxic death rate is 17%,
13%, 9% and 5%, respectively.
Table for Grade 3+ GVHD monitoring boundary:
Excessive
# evaluable Pocock boundary of
Looks Information toxicity
patients excessive toxicity
rate
1 12 33% 6 50.00%
2 24 67% 10 41.67%
3 36 100% 13 36.11%
The intervention will be rejected for excessive Grade 3+ GVHD if the
number of observed Grade 3+ GVHD is greater than or equal to the
corresponding boundary at any interim analysis. The actual power (based
on exact binomial test) of declaring the intervention too toxic are
approximately 96.4%, 80.5%, 45.9% and 13.2% when the true rate of
Grade 3+ GVHD is 49%, 41%, 33% and 25%, respectively.
See Appendix XII for guidelines for establishing organ stage and overall
grade of acute GVHD.
9.3.4 Treatment Failure (TF) patients who are non-randomly assigned to blinatumomab
Point estimate and the corresponding exact 95% confidence interval based on
Clopper-Pearson’s method will be provided to estimate the hematologic CR , the
rate of MRD < 0.01%, and the proportion treatment failure patients able to
proceed to hematopoietic stem cell transplant (HSCT).
The toxic death will be closely monitored for TF patients who are non-randomly
assigned to blinatumomab. According to past studies, an approximate 60 (10%) of
enrolled patients will be deemed as TF, and the cumulative toxic death rate is
estimated to be 6%. The safety stopping boundaries to be used for toxic death will
be based on the Pocock-type spending function at one-sided 20% significance
level. The intervention for TF patients will be temporarily closed for detailed
review and possible therapy modifications if the number of toxic deaths is greater
than or equal to that specified in the corresponding table below,
Table for toxic death monitoring boundary:
# evaluable Pocock boundary of Excessive toxic
Looks Information
patients excessive toxicities death rate
1 12 33% 3 25.00%
2 24 67% 4 16.67%
3 36 100% 5 13.89%
4 48 33% 6 12.50%
5 60 67% 7 11.67%
The intervention will be rejected for excessive toxicity death if the number of
observed toxic deaths is greater than or equal to the corresponding boundary at any
interim analysis. The actual power (based on exact binomial test) of declaring the
intervention too toxic are approximately 85.9%, 67.1%, 39.1% and 12.9% when
the true toxic death rate is 15%, 12%, 9% and 6%, respectively.
Accrual Targets
Sex/Gender
Ethnic Category
Females Males Total
Please note: ‘CTCAE v4.0’ is understood to represent the most current version of CTCAE
v4.0 as referenced on the CTEP website (i.e., v4.02 and all subsequent iterations prior to
version 5.0).
11.1 Purpose
Adverse event (AE) data collection and reporting, which are required as part of every
clinical trial, are done to ensure the safety of patients enrolled in the studies as well as those
who will enroll in future studies using similar agents. Certain adverse events must be
reported in an expedited manner to allow for timelier monitoring of patient safety and care.
The following sections provide information about expedited reporting.
Commercial agents are those agents not provided under an IND but obtained instead from
a commercial source. The NCI, rather than a commercial distributor, may on some
occasions distribute commercial agents for a trial.
When a study includes both investigational and commercial agents, the following rules
apply
• Concurrent administration: When an investigational agent is used in
combination with a commercial agent, the combination is considered to be
occurs at any time following treatment with an agent under a NCI IND/IDE since
these are considered to be serious AEs
11.4.3 Death
Reportable Categories of Death
Any death occurring within 30 days of the last dose, regardless of attribution to
the investigational agent/intervention requires expedited reporting within 24 hours.
Any death occurring greater than 30 days after the last dose of the investigational
agent/intervention requires expedited reporting within 24 hours only if it is
possibly, probably, or definitely related to the investigational agent/intervention.
The NCI requires all secondary malignancies that occur following treatment with
an agent under an NCI IND/IDE be reported via CTEP-AERS. Three options are
available to describe the event:
• Leukemia secondary to oncology chemotherapy
• Myelodysplastic syndrome
• Treatment related secondary malignancy
Any malignancy possibly related to cancer treatment (including AML/MDS) must
also be reported via the routine reporting mechanisms outlined in this protocol.
Second Malignancy:
A second malignancy is one unrelated to the treatment of a prior malignancy
(and is NOT a metastasis from the initial malignancy). Second
11.4.5.1 Pregnancy
Patients who become pregnant on study risk intrauterine exposure of the fetus to
agents that may be teratogenic. For this reason, pregnancy needs to be reported in
an expedited manner via CTEP-AERS as
Grade 3 “Pregnancy, puerperium and perinatal conditions - Other
(pregnancy)” under the Pregnancy, puerperium and perinatal conditions SOC.
There is a possibility that the sperm of male patients treated on studies involving
possible teratogenic agents may have been damaged. For this reason, pregnancy
in partners of men on study needs be reported and followed in the same
manner as a patient pregnancy.
Pregnancy needs to be followed until the outcome is known. If the baby is born
with a birth defect or anomaly, then a second CTEP-AERS report is required.
ROUTINE reporting: The AE must be reported only once unless the grade
becomes more severe in a subsequent course. If the grade becomes more severe
the AE must be reported again with the new grade.
EXPEDITED reporting: The AE must be reported only once unless the grade
becomes more severe in the same or a subsequent course.
Note: All expedited AEs (reported via CTEP-AERS) must also be reported via routine
reporting. Routine reporting is accomplished via the Adverse Event (AE) Case Report
Form (CRF) within the study database.
An expedited AE report for all studies utilizing agents under an NCI IND/IDE must be
submitted electronically to NCI via CTEP-AERS at:
https://webapps.ctep.nci.nih.gov/openapps/plsql/gader_accept$.startup.
In the rare situation where Internet connectivity is disrupted, the 24-hour notification is to
be made to the NCI for agents supplied under a CTEP IND by telephone call to 301-897-
7497.
In addition, once Internet connectivity is restored, a 24-hour notification that was phoned
in must be entered into the electronic CTEP-AERS system by the original submitter of the
report at the site.
• Any death occurring within 30 days of the last dose, regardless of attribution to an
agent/intervention under an NCI IND/IDE requires expedited reporting within 24
hours.
• Any death occurring greater than 30 days of the last dose with an attribution of
possible, probable, or definite to an agent/intervention under an NCI IND/IDE requires
expedited reporting within 24 hours.
CTEP-AERS Medical Reporting includes the following requirements as part of the report:
1) whether the patient has received at least one dose of an investigational agent on this
study; 2) the characteristics of the adverse event including the grade (severity), the
relationship to the study therapy (attribution), and the prior experience (expectedness) of
the adverse event; 3) the Phase (1, 2, or 3) of the trial; and 4) whether or not hospitalization
or prolongation of hospitalization was associated with the event.
11.9 Reporting Table for Late Phase 2 and Phase 3 Studies – Table A
Expedited Reporting Requirements for Adverse Events that Occur on Studies under an
IND/IDE within 30 Days of the Last Administration of the Investigational
Agent/Intervention 1
FDA REPORTING REQUIREMENTS FOR SERIOUS ADVERSE EVENTS (21 CFR Part 312)
NOTE: Investigators MUST immediately report to the sponsor (NCI) ANY Serious Adverse Events, whether or
not they are considered related to the investigational agent(s)/intervention (21 CFR 312.64)
An adverse event is considered serious if it results in ANY of the following outcomes:
1) Death.
2) A life-threatening adverse event.
3) Any AE that results in inpatient hospitalization or prolongation of existing hospitalization for ≥ 24 hours. This
does not include hospitalizations that are part of routine medical practice.
4) A persistent or significant incapacity or substantial disruption of the ability to conduct normal life functions.
5) A congenital anomaly/birth defect.
6) Important Medical Events (IME) that may not result in death, be life threatening, or require hospitalization may
be considered serious when, based upon medical judgment, they may jeopardize the patient or subject and may
require medical or surgical intervention to prevent one of the outcomes listed in this definition. (FDA, 21 CFR
312.32; ICH E2A and ICH E6.)
ALL SERIOUS adverse events that meet the above criteria MUST be immediately reported to the NCI via CTEP-
AERS within the timeframes detailed in the table below.
Hospitalization Grade 1 Grade 2 Grade 3 Grade 4 & 5
Timeframes Timeframes Timeframes Timeframes
Resulting in
Hospitalization 7 Calendar Days
≥ 24 hrs 24-Hour Notification
Not resulting in 5 Calendar Days
Hospitalization Not Required 7 Calendar Days
≥ 24 hrs
NOTE: Protocol specific exceptions to expedited reporting of serious adverse events are found in the Specific
Protocol Exceptions to Expedited Reporting (SPEER) portion of the CAEPR. Additional Special Situations as
Exceptions to Expedited Reporting are listed below.
Expedited AE reporting timelines are defined as:
“24-Hour; 5 Calendar Days” - The AE must initially be reported via CTEP-AERS within 24 hours of learning of
the AE, followed by a complete expedited report within 5 calendar days of the initial 24-hour notification.
“7 Calendar Days” - A complete expedited report on the AE must be submitted within 7 calendar days of learning
of the AE.
1SAEs that occur more than 30 days after the last administration of investigational agent/intervention and have an
Table B
Reporting requirements for adverse events experienced by patients on study who
have NOT received any doses of an investigational agent on this study.
CTEP-AERS Reporting Requirements for Adverse Events That Occur During
Therapy With a Commercial Agent or Within 30 Days1
12.1 CDUS
This study will be monitored by the Clinical Data Update System (CDUS). Cumulative
CDUS data will be submitted quarterly to CTEP by electronic means. Reports are due
January 31, April 30, July 31 and October 31. This is not a responsibility of institutions
participating in this trial.
12.2 CTA/CRADA
The agent(s) supplied by CTEP, DCTD, NCI used in this protocol is/are provided to the
NCI under a Collaborative Agreement (CRADA, CTA, CSA) between the Pharmaceutical
Company(ies) (hereinafter referred to as “Collaborator(s)”) and the NCI Division of Cancer
Treatment and Diagnosis. Therefore, the following obligations/guidelines, in addition to
the provisions in the “Intellectual Property Option to Collaborator”
(http://ctep.cancer.gov/industryCollaborations2/intellectual_property.htm) contained
within the terms of award, apply to the use of the Agent(s) in this study:
12.2.1 Agent(s) may not be used for any purpose outside the scope of this protocol, nor can
Agent(s) be transferred or licensed to any party not participating in the clinical study.
Collaborator(s) data for Agent(s) are confidential and proprietary to Collaborator(s)
and shall be maintained as such by the investigators. The protocol documents for
studies utilizing Agents contain confidential information and should not be shared or
distributed without the permission of the NCI. If a copy of this protocol is requested
by a patient or patient’s family member participating on the study, the individual
should sign a confidentiality agreement. A suitable model agreement can be
downloaded from: http://ctep.cancer.gov.
12.2.2 For a clinical protocol where there is an investigational Agent used in combination
with (an)other Agent(s), each the subject of different Collaborative Agreements, the
access to and use of data by each Collaborator shall be as follows (data pertaining to
such combination use shall hereinafter be referred to as "Multi-Party Data”):
a. NCI will provide all Collaborators with prior written notice regarding the
existence and nature of any agreements governing their collaboration with NCI,
the design of the proposed combination protocol, and the existence of any
obligations that would tend to restrict NCI's participation in the proposed
combination protocol.
b. Each Collaborator shall agree to permit use of the Multi-Party Data from the
clinical trial by any other Collaborator solely to the extent necessary to allow
said other Collaborator to develop, obtain regulatory approval or commercialize
its own Agent.
c. Any Collaborator having the right to use the Multi-Party Data from these trials
must agree in writing prior to the commencement of the trials that it will use
the Multi-Party Data solely for development, regulatory approval, and
commercialization of its own Agent.
12.2.3 Clinical Trial Data and Results and Raw Data developed under a Collaborative
Agreement will be made available to Collaborator(s), the NCI, and the FDA, as
appropriate and unless additional disclosure is required by law or court order as
described in the IP Option to Collaborator
(http://ctep.cancer.gov/industryCollaborations2/intellectual_property.htm).
Additionally, all Clinical Data and Results and Raw Data will be collected, used and
disclosed consistent with all applicable federal statutes and regulations for the
protection of human subjects, including, if applicable, the Standards for Privacy of
Individually Identifiable Health Information set forth in 45 C.F.R. Part 164.
12.2.4 When a Collaborator wishes to initiate a data request, the request should first be sent
to the NCI, who will then notify the appropriate investigators (Group Chair for
Cooperative Group studies, or PI for other studies) of Collaborator's wish to contact
them.
12.2.5 Any data provided to Collaborator(s) for Phase 3 studies must be in accordance with
the guidelines and policies of the responsible Data Monitoring Committee (DMC),
if there is a DMC for this clinical trial.
12.2.6 Any manuscripts reporting the results of this clinical trial must be provided to CTEP
by the Group office for Cooperative Group studies or by the principal investigator
for non-Cooperative Group studies for immediate delivery to Collaborator(s) for
advisory review and comment prior to submission for publication. Collaborator(s)
will have 30 days from the date of receipt for review. Collaborator shall have the
right to request that publication be delayed for up to an additional 30 days in order
to ensure that Collaborator’s confidential and proprietary data, in addition to
Collaborator(s)’s intellectual property rights, are protected. Copies of abstracts must
be provided to CTEP for forwarding to Collaborator(s) for courtesy review as soon
as possible and preferably at least three (3) days prior to submission, but in any case,
prior to presentation at the meeting or publication in the proceedings. Press releases
and other media presentations must also be forwarded to CTEP prior to release.
Copies of any manuscript, abstract and/or press release/ media presentation should
be sent to:
Email: [email protected]
For cases with a limited amount of tissue available for analysis, please prioritize
specimen for tissue banking.
This study is designed to provide material for banking for the purpose of performing
retrospective studies to refine risk stratification, identify new targets for therapy, identify
biomarkers to predict response, and to link host polymorphisms with various disease
characteristics and toxicities. See Appendix VI for details.
Please note: The AALL1331 Specimen Transmittal Form should be used. This is a protocol
specific form.
Please Note: The receiving institutions are strongly encouraged to make requests for
sample tubes well in advance of their first patient registration on AALL1331; it will not be
possible to expedite shipping because of prohibitive costs.
c. For BM, use multiple syringes and tubes as necessary. Reposition the BM aspirate needle
at least once during the diagnostic procedure to ensure the maximum quality of BM.
DO NOT SHIP SYRINGES.
d. Label each tube with COG registration number/Biopathology number, patient name and
date of birth, date, institution and type of specimen (bone marrow).
e. Samples are to be shipped at room temperature except for international samples that are
expected to be delayed for more than 48 hours – place a cold pack (not ice pack) in
shipment.
NOTE: For patients who are not having a diagnostic BM for medical reasons, and who
have an absolute blast count of at least 1,000/μL, 2 mL of PB at diagnosis may be submitted
for each 1 mL of required marrow and submitted to the Reference Laboratories instead of
diagnostic BM.
Molecular Laboratory
Julie Gastier-Foster, PhD
COG ALL Reference Laboratory
Nationwide Children’s Hospital
700 Children's Drive, C1961
Columbus, OH 43205
Contact Person: Yvonne Moyer
Phone: (614) 722-2866
Fax: (614) 722-2887
Email: [email protected]
All samples should be mailed by overnight carrier (Federal Express) PRIORITY (delivery
before 10 am). Use the COG FedEx account number, which may be found at:
https://members.childrensoncologygroup.org/_files/reference/FEDEXmemo.pdf.
Notify Laboratory of each Saturday delivery
Required Material:
a) Two original karyotypes of different cells from each abnormal clone or of two normal
cells when no abnormalities are found; full-size metaphase images of the karyotypes
cells.
b) A completed COG Cytogenetics Reporting Form and FISH Form (if done). These
reporting forms are available on the AALL1331 protocol web page. Electronic
submission of the karyotypes/FISH images is required.
Shipping: The review materials on each case should be sent to the COG ALL Cytogenetic
Coordinators within 4 weeks of study entry. If the institution is west of the Mississippi
River, send these materials to Dr. Andrew Carroll (University of Alabama). If the
institution is east of Mississippi River (except Minnesota and Wisconsin, which go to Dr.
Carroll), send all materials to Dr. Nyla Heerema (The Ohio State University).
Please note: The AALL1331 Specimen Transmittal Form should be used. This is a protocol
specific form.
Shipping Media (SM)
Samples for the Reference Laboratories are to be collected in shipping media (SM) in
special 15 mL conical tubes. The SM contains RPMI with EDTA as the anticoagulant.
These tubes will be prepared in the Molecular Reference Laboratory and shipped in
batches to each participating institution, where they can be stored frozen at -20°C until
use. Tubes are stable for 3 months if refrigerated and stable for 1 year if frozen. To
request prepared and pre-packaged sample shipping tubes, order tubes through the
Biopathology Center kit management system (use the link provided on the CRA Web
page).
Please note: The receiving institutions are strongly encouraged to make requests for
sample tubes well in advance of their first patient registration on AALL1331; it will NOT
be possible to expedite shipping because of prohibitive costs.
NOTE: For patients who are not having a diagnostic BM for medical reasons, and who
have an absolute blast count of at least 1,000/μL, 2 mL of PB at diagnosis may be
submitted for each 1 mL of required marrow and submitted to the Reference Laboratories
instead of diagnostic BM.
Please note: Specimens will be shipped to the COG Reference Laboratory at Johns Hopkins
(Eastern) ONLY.
All samples should be mailed by overnight carrier (Federal Express) PRIORITY (delivery
before 10 am). Use the COG FedEx account number, which may be found at:
https://members.childrensoncologygroup.org/_files/reference/FEDEXmemo.pdf.
Immunopharmacologic testing will be performed using blood and bone marrow specimens
in consenting patients treated with blinatumomab during the first exposure to
blinatumomab in an effort to identify biomarkers of blinatumomab response. For details,
refer to Appendix X.
NOTE: All patients will be offered participation in this study at Block 1, prior to treatment
assignment/randomization. Bone marrow samples will be drawn on all consenting patients
at end Block 1. If the patient is subsequently randomized or assigned to treatment with
blinatumomab (Arm B, Arm D, or Salvage Therapy), the Block 1 bone marrow sample will
be analyzed and subsequent blood samples should be obtained. If the patient is randomized
to treatment on the control arm (Arm A or Arm C), the Block 1 bone marrow sample will
be destroyed and no further samples will be collected.
Please note: The AALL1331 Specimen Transmittal Form should be used. This is a protocol
specific form.
• Blood during first blinatumomab cycle (7 samples, 3mL each in sodium heparin tubes)
o Day 1
- 0 hr (just prior to start of blinatumomab infusion)
- 6 hr
- 12 hr
o Day 2
o Day 7
o Day 14
o Day 21
All samples should be mailed by overnight carrier (Federal Express) PRIORITY (delivery
before 10 am). Use the COG FedEx account number, which may be found at:
https://members.childrensoncologygroup.org/_files/reference/FEDEXmemo.pdf.
Saturday deliveries are permissible. If a Saturday delivery is required, please notify Dr.
Brown of the planned shipment via email ([email protected]) PRIOR to the time of
shipment and clearly mark the package “For Saturday Delivery”.
This study is designed to analyze two specific aims: 1) To determine if specific protein
expression profiles, as determined by reverse-phase protein lysate array (RPPA) analysis
and phospho-flow analysis, correlate with therapy response and 2) To determine if
alterations in specific cell stress proteins (such as the UPR) during chemotherapy can
identify low risk patients with “high risk” protein signatures. For details, refer to Appendix
XI.
Consenting patients will have peripheral blood samples (5mL for those ≥ 10kg, 3 mL for
those < 10 kg) collected at 0h (prior to start of Block 1 systemic chemotherapy), and at 2
time points after the initiation of therapy (6 hours (h) and 24h after the first dose of Block
1 systemic chemotherapy) to examine for changes in protein expression patterns. Blood
samples for RPPA using sucrose centrifugation and (if necessary) the lymphoblast
population will be isolated by either bead technology or flow cytometry. Sample collected
for phospho-flow will be analyzed directly from whole blood. Sorted lymphoblasts will be
made into lysates for RPPA analysis prior to freezing. Remaining samples will be frozen
as pellets for protein analysis.
Eligible samples:
Sample should be sent to the Horton lab only if the patient has an initial absolute blast
count of at least 1000 lymphoblasts/µL. To calculate the absolute blast percentage,
multiply the total WBC by the % peripheral blasts:
Example: If the patient has a WBC of 10 and 50% blasts, the absolute blast count is:
(10)(.5)(1000) = 5000/µL
If the initial % blasts is unknown, send samples only if the total WBC is more than 10,000
and notify the Horton lab of the % blast as soon as available (contact info provided below).
Store samples in refrigerator until shipment, and use Thermosafe with ice pack (or
similar Styrofoam shipping containers with sufficient ice packs if these are not available at
your site.) These containers maintain biology samples at a constant temperature and are
strongly recommended for biology sample shipment, particularly in warm weather months.
All samples should be mailed by overnight carrier (Federal Express) PRIORITY (delivery
before 10 am). Use the COG FedEx account number, which may be found at:
https://members.childrensoncologygroup.org/_files/reference/FEDEXmemo.pdf.
The Horton lab can accept Saturday shipments if we are contacted ahead of time.
Please contact Gaye Jenkins or Dr. Horton (832-824-4676 or 832-824-4269) for
alternative address and shipping information for Saturday delivery.
positive for binding, non-neutralizing antibodies and have clinical sequelae that are
considered potentially related to an anti-blinatumomab antibody response may also be
asked to return for additional follow-up testing.
14.3 Timing
14.3.1 All patients who might require irradiation should be seen in consultation by a
radiation oncologist in advance of treatment. The purpose of the consultation is to
participate in planning the sequence of treatment and to determine the need for
extra medullary (cranial, testicular and other) and supplemental irradiation in
conjunction with HSCT.
14.3.2 The timing of TBI and supplemental irradiation will be determined in part by the
TBI fractionation regimen. Supplemental irradiation should precede TBI and be
performed during weekdays.
14.4 Emergency Irradiation
There may be instances when radiation therapy has been administered prior to enrollment
on this protocol because of potentially life-threatening or function-threatening
extramedullary involvement. Although it is not expected that prior treatment would make
total body irradiation contraindicated, it should be considered when describing to the
patient or parent the potential complications of treatment on this protocol.
14.5 Equipment and Methods of Delivery
Treatment Site Photons Electrons
TBI Any Energy
Cranial Irradiation 4 or 6MV
Testicular Irradiation Any Energy Any Energy
Any energy for photons or electrons can be used as long as the skin and target dose
requirements are met.
14.6 Treatment Volumes
14.6.1 Total Body Irradiation
The entire body will be treated including the head and feet in one field. Care should
be taken to insure that the patient is entirely within the 90% isodose line of the
beam and not in the penumbra region.
14.6.2 Cranial Irradiation
The treatment site consists of the entire brain and intracranial subarachnoid volume
as well as the optic nerves and the posterior halves of the optic globes. The caudal
border shall be the C2/3 vertebral interspace.
14.6.3 Testicular Irradiation
The treatment site consists of the testes in the scrotal sac.
14.7.4 Prescribed Dose and Fractionation for Testicular Irradiation (testicular leukemia
at protocol entry)
meninges. Shielding blocks cover the anterior halves of the eyes and
protect the nose and mouth.
• The "RT-2 Radiotherapy Total Dose Record" forms must be completed for cranial
and testicular irradiation.
• If the treatment technique differs from an approved benchmark, a new benchmark
must be completed and approved.
Deviation Unacceptable:
• The dose to the prescription point differs from the protocol specified dose
by >10%.
14.12.3 Deviations for Testicular Irradiation Dose
Variation Acceptable:
• The dose to the prescription point differs from the protocol specified dose
by > 6% but ≤ 10%.
Deviation Unacceptable:
• The dose to the prescription point differs from the protocol specified dose
by > 10%.
NOTE: For blinatumomab-specific supportive care guidelines, please refer to Section 5.2.3.
General Guidelines
Aggressive supportive care improves outcome. The following guidelines are intended to give general
direction for optimal patient care and to encourage uniformity in the treatment of this study population.
Notify Study Chair of any unexpected or unusually severe complications. Please also see the COG
Supportive Care Guidelines at: https://members.childrensoncologygroup.org/prot/reference_materials.asp
Blood Components
Blood products should be irradiated following current FDA guidelines found at:
http://www.fda.gov/OHRMS/DOCKETS/98fr/981218g2.pdf
Investigators in Canadian institutions need to follow the CSA standards for Blood and Blood Components
CAN/CSA-Z902-10 issued in February 2010 and available at: http://www.shopcsa.ca
Platelets
Transfusion with platelets is indicated to correct bleeding manifestations and may be indicated for severe
thrombocytopenia without bleeding particularly prior to an invasive procedure.
Infection Prophylaxis
Patients undergoing transplantation for ALL on this protocol are at high risk for infection. This risk may be
increased due to infections that occur during the intensive chemotherapy most patients will receive prior to
stem cell transplant on this protocol. Centers should be especially mindful of this as they choose anti-
bacterial, anti-fungal, anti-viral, and PCP prophylactic regimens. Aside from well established screening
approaches for CMV, centers may wish to consider routine screening for other viral pathogens such as
Adenovirus or HHV-6. Any prophylaxis regimen chosen must be the same for patients on both arms of the
protocol. Special considerations should be given for antifungal prophylaxis (see below).
Pneumocystis jiroveci
All patients should receive trimethoprim/sulfamethoxazole (TMP/SMX) at a dose of TMP 2.5 mg/kg/dose
(maximum dose 160mg/dose) by mouth twice daily on 2 or 3 sequential days per week. For patients allergic
to or experiencing excessive myelosuppression with TMP/SMX, alternative prophylaxis with dapsone (2
mg/kg/day by mouth, maximum dose 100 mg/day), aerosolized pentamidine (300 mg/q month ≥ 5 years of
age), or atovaquone (4-24 month: 45 mg/kg/day; >24 months: 30mg/kg/day) by mouth may be considered.
For children in whom TMP/SMX, dapsone, atovaquone, and inhaled pentamidine cannot be administered,
IV pentamidine (4 mg/kg/dose IV every 2 to 4 weeks) should be given. (REF: Centers for Disease Control
and Prevention. Guidelines for preventing opportunistic infections among hematopoietic stem cell
transplant recipients: recommendations of CDC, the Infectious Disease Society of America, and the
American Society of Blood and Marrow Transplantation. MMWR 2000; 49(No. RR-10):1-125. Available
at http://www.cdc.gov/mmwr/PDF/rr/rr4910.pdf. Accessed November 24, 2010.) Please also see
https://members.childrensoncologygroup.org/prot/reference_materials.asp for COG Supportive Care
Guidelines.
Varicella Vaccine
May be given to the siblings of patients in remission and stable at the physician’s discretion. Varicella
vaccination is not recommended for ALL patients during therapy.
Gamma globulin
If clinically indicated, IgG levels may be monitored throughout treatment. If the IgG level falls below
age-determined normal levels, IVIG at 400 mg/kg may be administered at the discretion of the
investigator. Note of IVIG administration should be recorded on data form.
Antifungals
Azole antifungal agents (i.e. fluconazole, itraconazole, voriconazole) given concurrently with vincristine
may increase the risk of neurotoxicity. Investigator caution is advised if azole antifungals are used.
Fluconazole and other azoles are expected to increase serum tacrolimus and sirolimus levels. Therefore,
dosages of sirolimus and tacrolimus should be adjusted accordingly. Due to extreme interactions with
sirolimus, voriconazole is contraindicated during sirolimus. Patients on voriconazole prior to transplant
should have another drug substituted for voriconazole prior to starting sirolimus therapy.
Use of G-CSF
G-CSF may be used for severe infections with neutropenia, but routine use is discouraged.
(TMP/SMX), and fungi (amphotericin) pending culture results. If fungal pulmonary disease is documented,
surveillance radiographic imaging studies of the sinuses, abdomen/pelvis and brain are indicated. Surgical
excision of pulmonary lesions should be considered at the discretion of the treating physician. Treatment
of fungal infections with amphotericin B and/or other antifungal agents will be at the discretion of the
treating physician. Azole antifungal agents (i.e. fluconazole, itraconazole, voriconazole) given concurrently
with vincristine may increase the risk of neurotoxicity. Investigator caution is advised if azole antifungals
are used.
Constipation
As vincristine may cause severe constipation, prophylactic stool softeners/laxatives (per investigator‘s
discretion) are recommended during all phases in which VCR is given.
17 ____IU
---
19
22 _____mg* _____mg _____mg b, e*, f
---
5
---
15 ____mg& ____mg& b, f
---
19
22 ____mg b, e#, f
---
29 b,c,d,f,g,h+,i
Following count recovery, HR/IR patients on Arm A receive Block 3 therapy (Section 4.4, APPENDIX II-C). HR/IR Treatment Failures post-Block 2 have the opt
of receiving 2 blocks of Salvage Therapy –(Blinatumomab-S) (Sections 4.7, 4.8, APPENDIX II-F, II-G). LR patients randomized to Arm C receive Block 3 therapy
(Section 4.4., APPENDIX II-C), and LR patients randomized to Arm D receive Blinatumomab Block 1: Cycle 1 therapy (Section 4.15, APPENDIX II-D).
^
All patients with M1 marrow must have ANC ≥ 500/µL and platelets ≥ 50,000/µL prior to beginning Day 8 ID MTX. Day 8 treatment must begin no later than 14
days after risk assignment for patient to continue to receive protocol therapy.
& Await count recovery to ANC ≥ 500/µL and platelets ≥ 50,000/µL prior to beginning Day 15 cyclophosphamide and etoposide.
+ Peripheral blood: End Block 2 (See Section 13.1 for complete details)
*Administer 4 hours after completion of Day 8 IV MTX. (Day 9 or 10)
# CNS3 ONLY
See Section 5.0 for Dose Modifications for Toxicities and Appendix I for Supportive Care Guidelines.
$ Prior to (Hour 0) first blinatumomab infusion (see Section 13.7 & lab manual for details)
&
See lab manual for collection and shipping details
*See recommended vital sign monitoring for Blinatumomab blocks in Section 7.1e
See Section 5.0 for Dose Modifications for Toxicities and Appendix I for Supportive Care Guidelines.
8 ____mg ____mg b, d, e
---
15
---
22 b, e
---
28
29 ____mg ____mg b, c, d, e,f$
30-35 Rest Period
Following count recovery, patients will proceed to HSCT (Section 4.9). See Section 4.10, Appendix II-H for suggested bridging therapy in the event that
HSCT is delayed. Treatment failures will be taken off protocol therapy.
$
In cases where blinatumomab treatment will not continue to Cycle 2, collect sample at end of Cycle 1. See Section 13.7 and lab manual for details
&
*See recommended vital sign monitoring for Blinatumomab blocks in Section 7.1e See lab manual for collection and shipping details
See Section 5.0 for Dose Modifications for Toxicities and Appendix I for Supportive Care Guidelines.
OBTAIN OTHER
STUDIES AS
REQUIRED FOR GOOD
PATIENT CARE
Testicular Radiotherapy: Patients with persistent testicular disease will receive testicular radiation during the 1st cycle of Blinatumomab-S therapy. See Section 14.2 for details of TRT.
7 i+
8 b, d
---
14 g&, i+
15 ____mg b, c, d, i+
---
21 i+
22 b, d
---
28
29 b, d, e, f+++
30-35 Rest Period
See Section 4.7.6 for criteria for proceeding to HSCT (Section 4.9) or to Blinatumomab-S: Cycle 2 (Section 4.8, APPENDIX II-G)
+
Peripheral blood: Prior to (Hour 0) and during (Hour 6, Hour 12, Day 2, Day 7, Day 14, Day 21) first blinatumomab infusion (see Section 13.5 for complete
details).
$ Prior to (Hour 0) first blinatumomab infusion. In cases where blinatumomab treatment will not continue to Cycle 2, collect additional sample at end of Cycle 1. (see Section
13.7 and lab manual for details)
&
See lab manual for collection and shipping details
*See recommended vital sign monitoring for Blinatumomab blocks in Section 7.1e
+++ Pre-HSCT evaluation
See Section 5.0 for Dose Modifications for Toxicities and Appendix I for Supportive Care Guidelines.
8 ____mg b, d, e
---
15 b, e
---
22 b, e
---
29 ____mg b, c, d, e,g$,
f+++
30-35 Rest Period
See Section 4.8.5 for criteria for proceeding to HSCT (Section 4.9) or to Bridging Maintenance Therapy Section 4.10, Appendix II-H. Patients
with M2/M3 marrow after Blinatumomab-S: Cycle 2 will be taken off protocol therapy.
$
In cases where blinatumomab treatment will not continue to Cycle 2, collect sample at end of Cycle 1. See Section 13.7 and lab manual for details
*See recommended vital sign monitoring for Blinatumomab blocks in Section 7.1e
+++ Pre-HSCT evaluation
See Section 5.0 for Dose Modifications for Toxicities and Appendix I for Supportive Care Guidelines.
8 ___mg b,d
---
15 ___mg b,d
----
24 ____mg
25 ____mg
---
29 ___mg b,d
---
36 ___mg b,d
----
42
43^ ___mg ___mg ___mg ___mg ___mg b, c,d
44 ___mg
---
46
47
---
48
49
50 ___mg ___mg b, d
51 ___mg
---
53
54
---
56 b,d
Following Continuation 1, upon count recovery, LR patients randomized to Arm C will receive Continuation 2 therapy (Section 4.11, APPENDIX II-I), LR patients randomized to Arm D will receive
Blinatumomab Block: Cycle 2 therapy (Section 4.16, APPENDIX II-N),
Following Continuation 2, upon count recovery, LR patients randomized to Arm C will receive Maintenance Cycle 1 therapy (Section 4.12, Appendix II-J), LR patients randomized to Arm D will receive
Blinatumomab Block: Cycle 3 therapy (Section 4.16, APPENDIX II-N),
^ Await count recovery to ANC ≥ 500/µL and platelets ≥ 50,000/µL prior to beginning Day 22 and Day 43 therapy. See Section Section 4.11 for details
See Section 5.0 for Dose Modifications for Toxicities and Appendix I for Supportive Care Guidelines.
Version Date: 04/27/15 224
THIS PROTOCOL IS FOR RESEARCH PURPOSES ONLY, SEE PAGE 1 FOR USAGE POLICY AALL1331
8 ______mg
----
15 ______mg
----
22 ______mg
----
36 ______mg
----
43 ______mg
-----
50 ______mg
-----
64 ______mg
-----
71 ______mg
-----
78 ______mg
----
84 a, b, d, e
85 Following Maintenance Cycle 1 therapy, CNS3 patients ONLY will receive CNS directed therapy with chemoradiation (Section 4.13, APPENDIX II-K). All other
patients on Arm C and Arm D will continue with Maintenance Post-Cycle 1 therapy (Section 4.14, APPENDIX II-L)
$ Arm D ONLY. Prior to start of Maintenance therapy for eligible patients (see Section 13.7 and lab manual for details)
See Section 5.0 for Dose Modifications for Toxicities and Appendix I for Supportive Care Guidelines.
Dexamethasone (DEX) PO 5 mg/m²/dose 1-7, 15-21 Total Daily Dose: 10 mg/m2/day, divided a Hx, PE [VS/Wt (BSA)]
BID. b CBC/diff/platelets
c Bilirubin, ALT & creatinine,
VinCRIStine (VCR) IV push over 1 minute+ 1.5 mg/m²/dose 1, 8, 15 + Or infusion via minibag as per
BUN
institutional policy d Absolute lymphocyte count with
T and B subset quantification.2
Maximum dose: 2 mg
Pegaspargase (PEG-ASP) IV over 1-2 hours 2500 International units/m²/dose 1 Administer through the tubing of a freely
infusing solution of D5W or 0.9% NaCl
See Section 7.0 for further details.
Cranial Radiotherapy: Patients with CNS3 and isolated CNS relapse will receive cranial radiation during Maintenance, between Blocks 1 and 2. See
Section 14.1 for details of cXRT. OBTAIN OTHER STUDIES
AS REQUIRED FOR GOOD
PATIENT CARE
Enter Cycle #: _____ Ht _________cm Wt _________kg BSA _________m2
Date Due Date Day DEX. VCR PEG-ASP Studies Comments (Include any held doses, or dose
Given ____mg ____mg ____IU modifications)
Enter calculated dose above and actual dose administered below
1 ________mg ________mg _______IU a, b, c
2
3
4
5
6
7
8 ________mg
---
15 ________mg ________mg
16
17
18
19
20
21 d
22 Following chemoradiation, patients will receive Maintenance Post-Cycle 1 therapy (Section 4.14, APPENDIX II-L) when count parameters are met.
See Section 5.0 for Dose Modifications for Toxicities and Appendix I for Supportive Care Guidelines.
Maintenance Post Cycle 1 is given in 12 week cycles based on dose modifications for low counts and platelets. See Section 4.14 and Section 5.9 for details. This therapy delivery map is on two (2)
pages.
DRUG ROUTE DOSAGE DAYS IMPORTANT NOTES OBSERVATIONS
8 ______mg
----
15 ______mg
----
22 ______mg
----
36 ______mg
----
43 ______mg
-----
50 ______mg
-----
64 ______mg
-----
71 ______mg
-----
78 ______mg
----
84 a, b, d, e
85 Maintenance cycles are repeated in 12 week cycles based on dose modifications for low counts or low platelets until 2 yrs from start of Block 1 therapy for both
male and female patients.
See Section 5.0 for Dose Modifications for Toxicities and Appendix I for Supportive Care Guidelines.
This cycle lasts 5 weeks (35 days) and starts when peripheral counts recover to ANC ≥ 500/µL and platelets ≥ 50 000/µL.. See Section 4.15 for treatment details. This Therapy Delivery Map is on one (1) page.
21 g+
22
---
b, d
28
29 ____mg ____mg b, c, d
30-35 Rest Period
36 Following Blinatumomab Block: Cycle 1, patients receive Continuation 1 therapy (Section 4.11, APPENDIX II-I) when count parameters are
met.
+
Peripheral blood: Prior to (Hour 0) and during (Hour 6, Hour 12, Day 2, Day 7, Day 14, Day 21) first blinatumomab infusion (see Section 13.5 for complete details).
$ Prior to (Hour 0) first blinatumomab infusion (see Section 13.7 and lab manual for details)
&
See lab manual for details
*See recommended vital sign monitoring for Blinatumomab blocks in Section 7.1e
See Section 5.0 for Dose Modifications for Toxicities and Appendix I for Supportive Care Guidelines.
Blinatumomab Block: Cycles 2 & 3; each last 5 weeks (35 days) and starts when peripheral counts recover to ANC ≥ 500/µL and platelets ≥ 50,000/µL. See Section 4.16 for treatment details.
This Therapy Delivery Map is on one (1) page.
DRUG ROUTE DOSAGE DAYS IMPORTANT NOTES OBSERVATIONS
1
Dexamethasone (DEX) PO or IV 10 mg/m²/dose , 6-12 hours prior to 1 Start prior to blinatumomab Obtain with each IT/ITT
blinatumomab infusion, therapy See Section 7.0 for further details.
THEN
5 mg/m2 within 30 minutes of starting OBTAIN OTHER STUDIES AS
Blinatumomab infusion REQUIRED FOR GOOD PATIENT CARE
8 b, c
---
15 b, c
---
22 b, c
---
28
29 b, c,d$
30-35 Rest Period
36 Following Blinatumomab Block Cycle 2: LR B-ALL patients randomized to Arm D will receive Continuation 2 (Section 4.11, APPENDIX II-I) when
count parameters are met.
Following Blinatumomab Block Cycle 3: LR B-ALL patients randomized to Arm D will receive Maintenance Cycle 1 therapy- (Section 4.12, Appendix
II-J) when count parameters are met.
$
Obtain at End Cycle 2. In cases where blinatumomab treatment will not continue to Cycle 2, collect sample at end of Cycle 1. See Section 13.7 and lab manual for details
*See recommended vital sign monitoring for Blinatumomab blocks in Section 7.1e
See Section 5.0 for Dose Modifications for Toxicities and Appendix I for Supportive Care Guidelines.
MERCAPTOPURINE 75 mg/m2
THIOGUANINE 40 mg/m2
*Patients exceeding a BSA of 3 m2 should have their TG doses calculated on actual BSA with no maximum dose.
Body Surface Area Daily Dose (d) for 7 days Cumulative Weekly
(m²)* (1 tablet = 40 mg) Dose
0.27 – 0.3 Use oral compounded suspension
0.31 – 0.34 Use oral compounded suspension
0.35 – 0.38 Use oral compounded suspension
0.39 – 0.41 Use oral compounded suspension
0.42 – 0.45 Use oral compounded suspension
0.46 – 0.48 Use oral compounded suspension
0.49 – 0.54 ½ tab / d x 7 140 mg/wk
0.55 – 0.61 ½ tab / d x 6; 1 tab / d x 1 160 mg/wk
0.62 – 0.68 ½ tab / d x 5; 1 tab / d x 2 180 mg/wk
0.69 – 0.75 ½ tab / d x 4; 1 tab / d x 3 200 mg/wk
0.76 – 0.82 1 tab / d x 4; ½ tab / d x 3 220 mg/wk
0.83 – 0.89 1 tab / d x 5; ½ tab / d x 2 240 mg/wk
0.9 – 0.96 1 tab / d x 6; ½ tab / d x 1 260 mg/wk
0.97 – 1.04 1 tab / d x 7 280 mg/wk
1.05 – 1.11 1 tab / d x 6; 1½ tab / d x 1 300 mg/wk
1.12 – 1.18 1 tab / d x 5; 1½ tab / d x 2 320 mg/wk
1.19 – 1.25 1 tab/ d x 4; 1½ tab / d x 3 340 mg/wk
1.26 – 1.32 1½ tab / d x 4; 1 tab / d x 3 360 mg/wk
1.33 – 1.39 1½ tab / d x 5; 1 tab /d x 2 380 mg/wk
1.4 – 1.46 1½ tab / d x 6; 1 tab / d x 1 400 mg/wk
1.47 – 1.54 1½ tab /day 420 mg/wk
1.55 – 1.61 1½ tab / d x 6; 2 tab / d x 1 440 mg/wk
1.62 – 1.68 1½ tab / d x 5; 2 tab / d x 2 460 mg/wk
1.69 – 1.75 1½ tab / d x 4; 2 tab / d x 3 480 mg/wk
1.76 – 1.82 2 tab / d x 4; 1½ tab / d x 3 500 mg/wk
1.83 – 1.89 2 tab / d x 5; 1½ tab / d x 2 520 mg/wk
1.9 – 1.96 2 tab / d x 6; 1½ tab / d x 1 540 mg/wk
1.97 – 2.04 2 tab / day 560 mg/wk
2.05 – 2.11 2 tab / d x 6; 2½ tab / d x 1 580 mg/wk
2.12 – 2.18 2 tab / d x 5; 2½ tab / d x 2 600 mg/wk
2.19 – 2.25 2 tab / d x 4; 2½ tab / d x 3 620 mg/wk
2.26 – 2.32 2½ tab / d x 4; 2 tab / d x 3 640 mg/wk
2.33 – 2.39 2½ tab / d x 5; 2 tab / d x 2 660 mg/wk
2.4 – 2.46 2½ tab / d x 6; 2 tab / d x 1 680 mg/wk
2.47 – 2.54 2½ tab / d 700 mg/wk
2.55 – 2.61 2½ tab / d x 6; 3 tab / d x 1 720 mg/wk
2.62 – 2.68 2½ tab / d x 5; 3 tab / d x 2 740 mg/wk
2.69 – 2.75 2½ tab / d x 4; 3 tab / d x 3 760 mg/wk
2.76 – 2.82 3 tab / d x 4; 2½ tab / d x 3 780 mg/wk
2.83 – 2.89 3 tab / d x 5; 2½ tab / d x 2 800 mg/wk
2.9 – 2.96 3 tab / d x 6; 2½ tab / d x 1 820 mg/wk
2.97 – 3 3 tab / d x 7 840 mg/wk
2. We are asking you to take part in a research study because you have B-ALL that has relapsed. A
research study is when doctors work together to try out new ways to help people who are sick. In this
study we are trying to learn more about how to treat B-ALL when it has come back after the first time
it was treated. During this study, we want to test whether the relapsed B-ALL responds better to
combination chemotherapy or a new drug called blinatumomab. The treatment that you get will
depend on a process called random assignment. Random assignment is a lot like flipping a coin and
you will have an equal chance of getting both treatment options.
3. Children and teens who are part of this study will be treated with either combination chemotherapy or
a new drug called blinatumomab. It is possible that some children will get radiation to their brain or
testes, depending on where the leukemia is found. Some children will also get a stem cell transplant.
4. Sometimes good things can happen to people when they are in a research study. These good things
are called “benefits.” We hope that a benefit to you of being part of this study is having the leukemia
go away for as long as possible. But we don’t know for sure if there is any benefit of being part of this
study.
5. Sometimes bad things can happen to people when they are in a research study. These bad things are
called “risks.” The risks to you from this study are that you might experience more side effects from
the combination chemotherapy or blinatumomab. Another risk is that the therapy on this study might
not be as effective as other options. Other things may happen to you that we don’t yet know about.
6. Your family can choose to be part of this study or not. Your family can also decide to stop being in
this study at any time once you start. There may be other treatments for your illness that your doctor
can tell you about. Make sure to ask your doctors any questions that you have.
7. We are asking your permission to collect additional blood. We want to see if there are ways to tell
how the cancer will respond to treatment. These samples would be taken when other standard blood
tests are being performed, so there would be no extra procedures. You can still be treated on this
study even if you don't allow us to collect the extra blood samples for research.
1. We have been talking with you about your illness, B-ALL. You have received treatment for this B-
ALL before. After doing tests, we have found that the cancer has come back in your bone marrow,
brain or spinal cord, or testes. B-ALL that has come back after the first treatment is called relapse.
2. We are asking you to take part in a research study because you have B-ALL that has relapsed. A
research study is when doctors work together to try out new ways to help people who are sick. In this
study we are trying to learn more about how to treat B-ALL when it has come back after the first time
it was treated. During this study, we want to test whether the relapsed B-ALL responds better to
combination chemotherapy or a new drug called blinatumomab. The treatment that you get will
depend on a process called random assignment. Random assignment is a lot like flipping a coin and
you will have an equal chance of getting both treatment options.
3. Children and teens who are part of this study will be treated with either combination chemotherapy or
a new drug called blinatumomab. It is possible that some children will get radiation to their brain or
testes, depending on where the leukemia is found. Some children will also get a stem cell transplant.
4. Sometimes good things can happen to people when they are in a research study. These good things
are called “benefits.” We hope that a benefit to you of being part of this study is having the leukemia
go away for as long as possible. But we don’t know for sure if there is any benefit of being part of this
study.
5. Sometimes bad things can happen to people when they are in a research study. These bad things are
called “risks.” The risks to you from this study are that you might experience more side effects from
the combination chemotherapy or blinatumomab. Another risk is that the therapy on this study might
not be as effective as other options. Other things may happen to you that we don’t yet know about.
6. Your family can choose to be part of this study or not. Your family can also decide to stop being in
this study at any time once you start. There may be other treatments for your illness that your doctor
can tell you about. Make sure to ask your doctors any questions that you have.
7. We are asking your permission to collect additional bone marrow. We want to see if there are ways to
tell how the cancer will respond to treatment. These samples would be taken when other standard
tests are being performed, so there would be no extra procedures. You can still be treated on this
study even if you don't allow us to collect the extra samples for research.
a) Rationale
The ability to collect leukemic and normal tissue samples for future research, and to link these samples to
patient characteristics and outcomes, has been critical to recent seminal discoveries regarding the molecular
basis of de novo ALL and relapsed ALL, and the relationship of specific molecular lesions with outcomes
and response to targeted therapies. In the context of this protocol, we will seek consent from participants to
provide material for banking for the purpose of performing retrospective studies to refine risk stratification,
identify new targets for therapy, identify biomarkers to predict response, and to link host polymorphisms
with various disease characteristics and toxicities.
Consenting patients will submit bone marrow and/or peripheral blood samples at study entry and, if
applicable, at second relapse to the central COG Tissue Bank at Nationwide Children’s Hospital for
storage of viably cryopreserved cells and for isolation nucleic acids, including DNA and RNA. Nucleic
acids isolated from relapse specimens will be stored for potential use for genomic profiling, which will
include single nucleotide polymorphism (SNP) arrays, array-based gene expression profiles and/or
quantitative RT-PCR-based gene expression analyses, methylation arrays, and/or comprehensive
sequencing approaches (e.g., RNA-seq, whole exome sequencing), depending on currently available
technologies and funding sources. Banked specimens subsequently will be distributed from the
reference laboratories to the laboratories performing the individual correlative biology studies. Results
from these correlative biology studies may ultimately facilitate (a) enhanced refinement of risk
stratification for patients with relapsed ALL, (b) detection of previously-unsuspected subset specificity
of blinatumomab, and (c) generation of additional hypotheses with respect to the underlying biology of
relapsed ALL.
• Host polymorphism studies
There is substantial evidence that both inherited germline constitutional and somatically-acquired ALL-
specific genomic variation may contribute to variations in response.87-96 Blood samples obtained
during remission are requested specifically for the purpose of providing constitutional (germline) DNA
from each patient. Several efforts are anticipated for study of how genomic variation (SNPs, copy
number variations, DNA methylation and insertions/deletions) in the constitutional DNA may be
associated with phenotypes in ALL. The phenotypes could include those related to probability of cure,
response, adverse events or classification. Techniques for interrogation of DNA variation are constantly
evolving, and thus these techniques vary but may include candidate polymorphism testing (eg, via
Taqman, GoldenGate, or PCR/RFLP assays), candidate gene sequencing (eg, via next-generation
technologies coupled with exon capture) or whole-genome sequencing (eg, via next-generation
technologies, with or without capture).
• Prepare all out-patient infusion bags at the registering/treating NCTN Network institution.
Study subjects should return to the registering/treating institution for all infusion bag
changes.
• For study subjects that cannot return to the registering/treating institution every 48-hours
for infusion bag exchanges, the next preference would be for another NCTN Network
institution that is participating on the trial and is closer to the subject’s home take
over responsibility for the study subject’s protocol participation. In such cases, transfer of
the subject’s protocol registration to another participating investigator and institution
should be considered.
b. The prepared infusion bags are stored at the local outpatient infusion center. The
infusion center would perform each infusion bag change.
c. If the local outpatient infusion center will not administer prepared infusion bags
admixed by the registering/treating institution, the registering/treating institution
may provide intact vials of blinatumomab to the local outpatient infusion center,
with infusion bags prepared and administered by the local outpatient infusion center
staff.
d. In either case, the local outpatient infusion center would be managed as a satellite
pharmacy of the registering/treating institution (see evaluation criteria below).
• If an outpatient infusion center is not an option, use of a home health care service provider
can be considered.
Note: If a home health care agency is being considered to prepare and change the blinatumomab
infusion bag, the drug company that provides blinatumomab will cover the costs associated with
a home health care agency providing these services.
a. The first preference would be for all outpatient infusion bags to be prepared by the
registering/treating institution and shipped via overnight courier delivery service in
a 2o to 8oC pre-qualified shipping container to the servicing home health care
agency.
b. The prepared infusion bags are stored by the home health care agency and each
individual infusion bag transported to the subject’s home by the home health care
service nursing staff under refrigerated storage conditions for each infusion bag
change.
c. If home health care agency will not administer prepared infusion bags admixed by
the registering/treating institution, the registering/treating institution may provide
intact vials of blinatumomab to the home health care agency, with infusion bags
prepared and administered by the home health care agency staff.
d. In either case, the home health care agency would be managed as a satellite
pharmacy of the registering/treating institution (see evaluation criteria below).
5. If all options above are not feasible, shipping the prepared infusion bags directly to
patient’s home via overnight courier delivery service for administration by home
healthcare agency staff is acceptable.
a. The prepared infusion bags are to be shipped in a 2o to 8oC pre-qualified shipping
container containing one infusion bag per box. Example, if you are making 2 x 48
hour infusion bags, each infusion bag will be shipped in a separate 2o to 8oC pre-
qualified shipping container. The number of infusion bags that may be prepared
and shipped is dependent on the duration the shipping container used is qualified to
maintain 2o to 8oC temperature.
b. Patients should NOT open the shipping container upon arrival. Shipping containers
are to be stored in a secured area away from reach of children or pets.
c. Shipping containers must only be opened by the home health care service staff at
the time of the infusion bag change. Only one shipping container should be opened
at a time. If cold-chain management of the prepared infusion bag has been
The home health care service staff should immediately contact the registering/treating
institution site pharmacy as indicated on the shipment form. Within 1 business day, the
registering/treating institution site should send an email to the COG Industry Sponsored
Trials office at [email protected] with a copy to PMB/CTEP at
[email protected] to report all such occurrences of prepared, unusable
infusion bags shipped to a patient’s home.
d. Form documenting the time of packaging in the shipping container, duration of time
the container will maintain 2o to 8oC temperature and verification that cold-chain
management was maintained prior to administration must be included in each
shipping container and returned to registering/treating institution for documentation
purposes. (See Appendix VII-B)
When the registering/treating institution is considering use of a local infusion center or home
health care agency as a satellite pharmacy, the following must be assessed by the
registering/treating institution in relation to the suitability of the local infusion center or home
health care agency:
• Ability to appropriately store (temperature and security) the intact agent vials and/or
prepared infusion bags.
• Ability to provide documentation of controlled and monitored temperature storage
conditions while the IND agent is in the local infusion center or home health care agency
possession.
• Availability of appropriately trained staff to prepare doses in compliance with USP <797>
guidelines and the protocol, to label infusion bags according to the protocol instructions
and to store agent doses under appropriate controlled temperature conditions.
• For home health care agency services, the ability to transport each prepared dose
individually to the subject’s home under appropriate controlled storage conditions or the
ability to assess and confirm that cold-chain management of prepared infusion bags
shipped to the subject’s home is maintained prior to administration.
• Availability of appropriately trained staff to administer the prepared doses and perform the
infusion bag changes according to the protocol.
• Methods for proper disposal of the waste, empty vials, IV bags, etc. are in place.
• Plan for return of unused intact vials to the registering/treating institution is in place.
• Source documentation to confirm agent administration must be maintained by the local
infusion center or home health care agency and must be provided to the registering/treating
institution for incorporation into the patient’s medical/research records and for audit
purposes.
Version Date: 04/27/15 242
THIS PROTOCOL IS FOR RESEARCH PURPOSES ONLY, SEE PAGE 1 FOR USAGE POLICY AALL1331
The Lead Network Group for the trial must work with participating sites to:
b. Develop a plan for participating NCTN Network sites to assess and train local
outpatient infusion centers or home health care agency for patient treatment if required
and document training of such sites
c. Have a training manual available for local outpatient infusion centers or home health
care agencies on the clinical trial, appropriate agent preparation, handling and
administration requirements and appropriate record keeping requirements
To be completed by Site/Pharmacy:
From: (Investigator Name, Address) To Patient: (Patient Initials, Study ID No) Protocol No.:
_________________ ; ____________________
Patient Initials Study ID Number
Prepare shipment of IV bag at 2°C to 8°C in validated/pre-qualified insulated shipper as per manufacturer
instructions (see shipping container instructions). Please take care to use the applicable instructions for
summer or winter package preparation, respectively.
Please tick the boxes and fill in the information below when preparing the IV bag shipment!
□ Validated/pre-qualified shipping container duration of time 2°C to 8°C temperature is
maintained: ___________________________ hours
□ Cooling elements for provided box used according to manufacturer’s instruction
Confirmed by:
(print name, signature) (date)
NO
CONFIDENTIAL
a) Rationale
Minimal residual disease is known to be a powerful prognostic factor in childhood ALL.1-5,12,20-22 Although
the majority of work has been done in newly diagnosed ALL patients, several studies using both polymerase
chain reaction methods13,14,16-18 and flow cytometry19,23 have shown that MRD is highly predictive of second
marrow relapse.
Flow cytometric MRD results will be used for the following purposes in this study:
• For late B-ALL marrow and late B-ALL IEM patients, we will use end-Block 1 flow cytometric MRD
levels of < 0.1% vs. ≥ 0.1% to define low risk group vs. intermediate risk group, respectively. The low
risk group will be eligible for LR randomization. The intermediate risk group will be eligible for HR/IR
Randomization.
• For the LR patients participating in LR randomization, we will use end-Block 2 MRD levels of < 0.01%
vs. ≥ 0.01% as a randomization stratification criteria to ensure balanced randomization.
• For the IR and HR patients participating in HR/IR Randomization, we will use end-Block 1 MRD levels
of <0.1% vs. ≥ 0.1% as a randomization stratification criteria to ensure balanced randomization.
• For HR/IR Randomization patients proceeding to HSCT, we will use the end-Block 3/pre-HSCT MRD
level, the peri-engraftment MRD level and the day +100 MRD level to determine eligibility for rapid
tapering of immunosuppression.
• In HR/IR Randomization, we will use flow cytometric MRD+ rates at the end of Blocks 2 and 3 as a
secondary efficacy objective.
b) Technique
Aliquots of fresh bone marrow specimens at study entry and during therapy at various designated time
points (see study schema and TDMs) will be adjusted to suitable cell concentrations and stained with the
following combinations of monoclonal antibodies in 6-color immunofluorescence:
Tube 1: CD20-FITC/CD10-PE/CD38-PerCP-Cy5.5/CD58-APC/CD19-PE-Cy7/CD45-APC-Cy7
Tube 2: CD9-FITC/CD13-PE+CD33-PE/CD34-PerCP-Cy5.5/CD10-APC/CD19-PE-Cy7/CD45-APC-
Cy7
After incubation, samples will be lysed with ammonium chloride, fixed with 0.25% ultra pure
formaldehyde, and washed once before analysis. Samples will be run on a Becton Dickinson FACSCanto.
A minimum of 750,000 events will be collected, and data will be analyzed by software developed by Dr.
Brent Wood, University of Washington that facilitates the hierarchical gating strategy useful for identifying
phenotypically aberrant cells. In cases in which the above panels are not informative for detecting abnormal
cells, additional markers including but not limited to CD15 and TdT will be added to the panel to help
identify MRD populations. In addition, if the administration of blinatumomab creates difficulties in
identifying leukemic cells by virtue of CD19 expression, cytoplasmic CD79a and/or CD22 will be used to
aid gating.
a) Rationale
Recent genomic studies of ALL have identified genomic alterations of CRLF2, which encodes the thymic
lymphopoietin receptor (TSLPR) and which results in upregulation of CRLF2 as an important contributor
to leukemic pathogenesis.3-5,7 Upregulation may occur by gene alterations, most often fusing CRLF2 to
either P2RY8 or IGH@, as well as rarely, a CRLF2 point mutation (F232C),3-5,7 though there may also be
as yet unrecognized mechanisms. In addition, patients with whose blasts show upregulated CRLF2 typically
also have simultaneous activating mutations in kinase genes including IKZF1, JAK1 and JAK2, and as a
result have abnormalities in signal transduction networks.3,4,7 Our results suggested high risk children with
overexpressed CRLF2 as detected by PCR have very poor outcome irrespective of whether structural
rearrangements can be identified,8 while other studies have suggested that this is only true of patients with
P2RY8-CRLF2 translocations.2,9 Moreover, in our studies CRLF2 did not appear to be prognostically
significant in standard risk patients.8 However, virtually nothing is known about CRLF2 overexpression in
the relapse setting, and whether its expression has continued prognostic significance in this already poor-
risk group of patients. In addition, because of the underlying kinase abnormalities in this patient population,
these patients are potentially ideal candidates for treatment with novel signal transduction inhibitors.
Assessing CRLF2 expression may help to identify patients who might be candidates for specific therapy in
trials that may be developed as these agents mature.
b) Technique
In conjunction with the immunophenotyping performed at study entry as a baseline for flow cytometric
MRD testing, we will also quantify surface TSLPR expression and correlate with outcome.
c) Specific Aims
Aims:
1) To determine if the frequency of high CRLF2 expression (which correlates with CRLF2
genomic lesions) is higher in the first marrow relapse B-ALL patient population than in the
initial diagnosis B-ALL patient population.
2) To determine if first marrow relapse B-ALL patients with high CRLF2 expression have an
inferior outcome to those without high CRLF2 expression.
d) Power calculations
Aim 1: Comparison of the frequency of CRLF2 expression among B-ALL patients in first marrow
relapse to that in newly diagnosed B-ALL patients.
From past COG studies for newly diagnosed B-ALL, it is estimated that 10% of standard risk (SR) and
21% of high risk (HR) patients will relapse. Assuming there are 2/3 SR and 1/3 HR newly diagnosed
patients, it is estimated that around 49% and 51% of patients in first relapse will be NCI SR and HR,
respectively. Given that about 7% and 12% of the SR and HR patients have high level CRLF2 expression
via flow cytometery, the overall rate of high CRLF2 expression (which correlates with genomic lesions)
in newly diagnosed B-ALL patients is about 10%.
The following table provides the power to detect a difference in rates of high CRLF2 expression between
newly diagnosed and 1st relapse B-ALL patients when the rate of high CRLF2 expression of the 1st
relapse patients is 13%, 15%, 18% and 20% (i.e. RR=1.3, 1.5, 1.8 and 2.0), respectively. The power
calculations are based on a one-sample exact test at 5% significance levels (one-sided). A total of 426
B-ALL marrow relapse ALL patients [180 early (CR1<36 months) and 246 late (CR1 ≥36 months)]
were considered in this power calculation.
Aim 2: Comparison of EFS in first marrow relapse B-ALL patients with high level CRLF2 expression
vs. those without high level CRLF2 expression.
It is anticipated that a total of 180 early (CR1<36 months) and 246 late (CR1 ≥36 months) marrow B-
ALL patients will be enrolled to AALL1331. According to past studies, the 3-year EFS of early and late
marrow relapse patients are approximately 26% and 65%, respectively, giving an overall 3-year EFS of
about 48%. Assuming the above rates of high-level CRLF2 expression and minimum 2 years of follow-
up, the table gives powers for comparing EFS between patient groups with and without high level
CRLF2 expression. The power calculations are based on the one-sided log-rank test at the 5%
significance level.
CRLF2 high rate for Sample Size 3-year EFS rates
Significance
1st relapsed pre-B CRLF2 Power
Level CRLF2- CRLF2+ CRLF2-
pts +
0.05 0.13 55 371 0.18 0.52 100.0
0.13 55 371 0.20 0.52 99.9
0.13 55 371 0.23 0.52 99.8
0.13 55 371 0.26 0.51 99.0
0.13 55 371 0.30 0.51 96.3
0.13 55 371 0.33 0.50 88.8
0.13 55 371 0.36 0.50 77.7
0.13 55 371 0.39 0.49 55.1
0.15 64 362 0.18 0.53 100.0
0.15 64 362 0.20 0.53 100.0
0.15 64 362 0.23 0.52 99.9
0.15 64 362 0.26 0.52 99.6
0.15 64 362 0.30 0.51 97.5
0.15 64 362 0.33 0.51 93.5
0.15 64 362 0.36 0.50 81.2
0.15 64 362 0.39 0.50 65.0
0.18 77 349 0.18 0.55 100.0
0.18 77 349 0.20 0.54 100.0
0.18 77 349 0.23 0.53 100.0
0.18 77 349 0.26 0.53 99.9
0.18 77 349 0.30 0.52 99.1
0.18 77 349 0.33 0.51 95.7
0.18 77 349 0.36 0.51 88.8
0.18 77 349 0.39 0.50 69.6
a) Rationale
For patients with relapsed/refractory ALL, response patterns to blinatumomab are essentially binary. Some
patients have a striking response, with a systemic cytokine release syndrome (CRS, most notably IL-6)
accompanying clearance of leukemic blasts and culminating in an MRD-negative remission, and others do
not respond.10,11,97 For patients that have received blinatumomab in the setting of MRD positivity (i.e., with
very low tumor burden), response rates are generally higher and significantly less likely to be associated
with CRS.4,5 Attempts to correlate responses with various peripheral blood parameters (such as ALC and
T-cell subset quantitation) have been largely unsuccessful in either setting, although the patient numbers
have been limited.3
The bone marrow microenvironment contains various immunologic components that have been shown to
either enhance or suppress cytotoxic T-cell effector response to tumor antigens. The bone marrow serves
as a reservoir of memory T-cells with heightened antigen specificity compared to peripheral blood memory
T-cells, and the bone marrow microenvironment is capable of effectively priming naïve T-cell responses.98-
100
However, the bone marrow is also a reservoir for suppressive regulatory CD4/CD25/FOXP3+ T-cells
(T regs).101 Comparative studies of bone marrow vs. peripheral blood in patients with myeloma have shown
that marrow-infiltrating lymphocytes (MILs) are more effectively activated and expanded and are more
capable of tumor-specific cytotoxicity than peripheral blood lymphocytes (PBLs).102 In addition, bone
marrow contains myeloid-derived suppressor cells (MDSCs) that have been shown to preferentially capture
and present tumor associated antigens to T regs, resulting in tumor tolerance, and preventing the capture
and presentation of these antigens by dendritic cells to activate tumor-specific cytotoxic effector T-cells.103
IL-6 is among the key mediators that skews the marrow T-cell repertoire from T regs to cytotoxic effector
T-cells.104
As has been suggested by previous studies, lymphocyte populations and cytokine profiles in the peripheral
blood may also provide insight into responses3 and adverse effects11. Since our study will include more
patients than any blinatumomab trial to date, our study may be particularly well-suited to detect these
associations.
We hypothesize that the balance of enhancing vs. suppressive components of the cytotoxic T-cell effector
response in the bone marrow microenvironment may be playing a particularly important role in determining
the presence or absence of clinical response to blinatumomab. Specifically, we hypothesize that the bone
marrow of patients that respond to blinatumomab (i.e., achieve complete continuous remission after
treatment with blinatumomab) will be characterized by a relative predominance of cytotoxic
memory/effector T-cells and a paucity of T regs and MDSCs, while the opposite pattern will be
characteristic of the bone marrow of non-responders (i.e., those that relapse after treatment with
blinatumomab).
• Specific Aim 1: To determine if the balance of enhancing vs. suppressive components of the
cytotoxic T-cell effector response in the bone marrow microenvironment at baseline correlates with
disease free survival (DFS) in relapsed ALL patients treated with blinatumomab.
We further hypothesize that lymphocyte populations and cytokine profiles in the peripheral blood at
baseline and at post-treatment time points (hours 6 and 12 on day 1, and once on days 2, 7, 14 and 21) may
also provide insight into adverse effects and clinical response, and so we will collect peripheral blood for
flow cytometric characterization of lymphocyte subsets (including T-cell subsets as above, as well as
circulating normal B-cells) and isolate plasma for multiplex cytokine profiling (including IL-6, IL-2R, IL-
8, IL-10, MCP-1, MIP-1B, and INF-γ).
• Specific aim 2: To determine if peripheral blood patterns of lymphocyte subsets and cytokines,
both at baseline and at post-treatment time points, correlates with incidence of blinatumomab-
related reportable adverse events (see section 5.2.1), and DFS in relapsed ALL patients treated with
blinatumomab.
c) Methods
• Processing, cryopreservation and quality control
Refer to Section 13.5 for specimen collection and shipment. All blood and marrow samples will first
be centrifuged to allow isolation of plasma. Plasma will be stored in 250 uL aliquots and stored at -80
C for subsequent batched assays. Cell pellets will be diluted in HBSS and centrifuged over Lymphocyte
Separation Medium (LSM) to isolate mononuclear cells, then suspended in cryoprotectant solution and
viably cryopreserved in liquid nitrogen in aliquots of 10e6 cells for subsequent batched assays. Assays
will be performed as cohorts of five patients have provided a complete set of samples.
The Brown laboratory routinely processes blood and marrow specimens from cooperative group
clinical trials and performs a wide variety of assays using plasma and viably cryopreserved cells.
Samples are generally received in the laboratory within 48 hours of collection and processed
immediately. Plasma and cells are sufficiently stable in transport under these conditions for the
proposed assays. The flow cytometric assays are routinely performed in the Pardoll laboratory, which
is collaborating on this project and has provided the processing protocol described above.
Vials of viably cryopreserved marrow and blood cells will be thawed and washed and viable cells
counted by trypan blue exclusion. Cells will then be stained with isotype controls and the following
antibody panels, and analyzed in triplicate by flow cytometry
o For lymphocyte subsets: CD3, CD4, CD8, CD19, CD20, CD45RO, CD45RA, HLADR, CD27,
CD62L, CD25, CD28 and CD127.
o For MDSCs (marrow only): Lin, HLA-DR, CD33, CD11b, CD14, CD15
The endpoint for this assay will be the relative percentage and absolute number (in cells/uL) and of
each of the following:
o T regs
o Naïve T-cells
o Memory T-cells (central vs. effector)
o Activated T-cells
o B-cells
o Monocytic MDSC (marrow only)
o Granulocytic MDSC (marrow only)
Vials of frozen plasma will be thawed and a panel of cytokines will be quantitated in triplicate using a
custom cytometric bead array kit (custom BD CBA) designed to measure the following: IL-6, IL-2R,
IL-8, IL-10, MCP-1, MIP-1B, and INF-γ. Isotype controls will serve as negative controls, and standard
curves will be generated using control cytokine solutions.
The endpoint for this assay will be levels of each cytokine in pg/mL.
The methods described above are performed routinely in the Brown (processing and
cryopreservation) and Pardoll (flow cytometric measurement of lymphocytes, MDSCs and
cytokines) laboratories, and are likely to optimize the chances that the proposed studies will be
successful.
• Specific Aim 1:
We will calculate ratio (R) of enhancing to suppressive components in bone marrow microenvironment
for each patient as follows:
We will use logistic regression to model outcome (no relapse vs. relapse) as a function of R. The
Benjamin-Hochberg procedure will be used to control for multiple comparisons.
The anticipated result is that R is negatively correlated with risk of relapse (i.e., a higher ratio of
enhancing to suppressive components will reduce risk of relapse). If this anticipated result is seen,
multivariate analysis that includes known associations with relapse (duration of first remission, MRD,
etc.) will be performed to determine whether the marrow microenvironment ratio adds anything
independent to known predictors of relapse in blinatumomab-treated patients.
This analysis will be performed on the entire cohort of patients treated with blinatumomab, and also
separately on the HR/IR cohort and on the LR cohort.
We anticipate that a total of 156 evaluable patients will be treated with blinatumomab on this study
(Section 9.2), of which 72 will be HR/IR and 84 will be LR. We assume that we will have 90%
compliance with submission of the marrow sample. We assume that about 10% of submitted samples
will fail analysis for technical reasons. Thus, we expect that our sample size for R will be 127 (59
HR/IR and 68 LR).
Since we are proposing to use standard assays (flow cytometric quantification of marrow cell subsets)
in a novel way (calculation of enhancing vs. suppressive components), we do not have a priori
knowledge of the expected range of observed ratios. As such, these studies are exploratory and
definitive power calculations are not possible. However, if we assume the log(R) follows a Gaussian
distribution with a mean of 0 and standard deviation of 1 (i.e., a standard normal distribution), and
assuming an overall relapse rate of 50% for the trial, a sample size of 127 will have 80% power to
detect an effect size of 0.5, using a 2-sided significance level of 0.05. For example, if we assume patients
who eventually relapse have a mean R of 1 (log(R)=0), then we would be able to detect that patients
who do not eventually relapse have a mean R of >3.16 (log(R)=0.5) or a mean R of <0.316 (log(R)=-
0.5). This effect size is not unreasonable to expect. These calculations are based on methods from Hsieh,
et al. Statist. Med. 17, 1623-1634 (1998).
The relationships between the ratios and rates of relapse will be described, and subsequent studies will
be designed to validate these findings prospectively in the context of future clinical trials.
• Specific Aim 2:
There are 12 variables to be determined for each blood sample (7 samples per patient):
1) Total T-cell number
2) CD4 count
3) CD8 count
4) Memory T-cell count
5) Activated T-cell count
6-12) Level of each of 7 individual cytokines
For each of the 12 variables, we will determine the baseline value (BV; hour 0) and the maximal change
value (MCV; value at the post-treatment timepoint for which the variable has increased or decreased to
the greatest degree). For the MCV, a landmark approach will be used in which all cases are followed
for 21 days, and the maximal change observed up to that point will be used to predict outcome from
that point forward for all patients who are still observable up to that landmark time point.
As a first level analysis, we will use logistic regression to model each of the 2 outcomes as a function
of each of the 12 variable’s BV, and each of the 12 variable’s MCV. The Benjamin-Hochberg procedure
will be used to control for multiple comparisons.
1) BV of some subsets of T-cell counts (memory T-cells, e.g.) are positively correlated with the risk of
blinatumomab-related reportable adverse events, and negatively correlated with risk of relapse.
2) MCV of a subset of cytokines (IL-6 and INF-γ, e.g.) is positively correlated with the risk of
blinatumomab-related reportable adverse events, and negatively correlated with risk of relapse.
Subsequent levels of analysis will attempt to characterize combinations of T-cell subset counts and
cytokine levels that may perform better than individual variables in modeling the outcomes of interest.
If any of the anticipated results are seen with respect to the relapse outcomes, multivariate analysis that
includes known associations with relapse (duration of first remission, MRD, etc.) will be performed to
determine whether any of the T-cell subset/cytokine variables adds anything independent to known
predictors of relapse in blinatumomab-treated patients.
This analysis will be performed on the entire cohort of patients treated with blinatumomab, and also
separately on the HR/IR cohort and on the LR cohort.
We anticipate that a total of 156 evaluable patients will be treated with blinatumomab on this study
(section 9.2), of which 72 will be HR/IR and 84 will be LR. We assume that we will have at least 90%
compliance with submission of the blood samples, and that about 10% of submitted samples will fail
analysis for technical reasons. Thus, we expect that our sample size will be 127 (59 HR/IR and 68 LR).
Since we are proposing to use standard assays (flow cytometric quantification of blood T-cell subsets
and cytokines) in a novel way (combining variables to model risks of adverse events and relapse after
treatment with blinatumomab), we do not have a priori knowledge of the expected ranges of values for
most variables. As such, these studies are exploratory and definitive power calculations are not possible.
However, some estimates based on specific assumptions are possible. If we assume that the baseline
value for memory T-cell count for the entire study population follows a Gaussian distribution with a
mean of 500/uL and a standard deviation of 100/uL, and we assume an overall relapse rate of 50% for
the trial, a sample size of 127 will have 80% power to detect an effect size of 50/uL, using a 2-sided
significance level of 0.05. For example, if we assume patients who eventually relapse have a mean BV
memory T-cell count of 500, then we would be able to detect that patients who do not eventually relapse
have a mean BV T-cell count of >550/uL, or a mean BV T-cell count of <450/uL. This effect size is
not unreasonable to expect. These calculations are based on methods from Hsieh, et al. Statist. Med.
17, 1623-1634 (1998).
The relationships between the variable(s) and rates of adverse events/relapse will be described, and
subsequent studies will be designed to validate these finding prospectively in the context of future
clinical trials.
a) Rationale
Many excellent studies have focused on gene expression profiles in ALL.51,52 However, changes in protein
cell stress pathways and deregulated signal transduction pathways, have been much less well studied in
ALL. There is an unmet need to learn more about chemotherapy-induced proteins expression changes in
order to determine if specific proteins or protein clusters can help predict response to therapy. Specific
protein expression patterns are likely to correlate with disease-free survival (DFS) and will enable us to
identify specific populations at risk for relapse. Since it is very difficult to salvage patients after second
relapse, one of the goals of this study is to determine which patients classified as low risk based on timing
and site of relapse (Section 9.2) would benefit from being risk stratified instead into the high risk group.
This would improve DFS in all patients with relapsed ALL. Relevant protein expression profiles will be
validated in future COG trial for patients with relapsed ALL and (if the increase in correct risk assignment
increases to meets statistical specifications) will be used, in conjunction with molecular classification
systems, as an adjunct for risk assignment.
We hypothesize that activation of cell stress pathways and signal transduction pathways deregulated in
ALL will correlate with DFS, and that the expression of specific proteins or proteins clusters will identify
patients classified as low risk patients that would benefit from more intensive therapy. We have two
specific aims to address these hypotheses:
1. To determine if specific protein expression profiles, as determined by reverse-phase protein
lysate array (RPPA) and phosphoflow cytometry, correlate with clinical outcome (DFS)
2. To determine if alterations in specific groups of cell stress proteins can be used to generate a
“high-risk” protein expression signature that identifies patients stratified to the low-risk group
(based on time and site of relapse) that would benefit from stratification into the high-risk
group.
c) The contributions that the proposed study will make to the current knowledge base
Little is known about the changes in cell stress proteins during chemotherapy. This analysis will contribute
greatly to our current knowledge base by 1) determining if specific proteins or protein clusters correlate
with response therapy, 2) increasing our knowledge about lymphoblast response to standard relapse
chemotherapy, 3) allowing a better understanding of the biology-defined ALL subgroups, aiding the
development of targeted therapies, and 4) allowing for the development of protein expression risk-
classifiers that could be validated and utilized, along with gene expression profiling, to aid in risk
stratification in subsequent relapsed clinical trials.
f) The comparability of the methods proposed to those previously used, and the likelihood that
the resulting data will be able to be compared with existing data. Analysis of RPPA is currently
being performed using the same methods in AAML1031, and so these results should be directly
comparable. Analysis of signal transduction networks will use identical methods to those piloted
in ADVL1011 and ADVL1114, and so these should be comparable as well.
b. Statistical analysis: Supercurve algorithms were used to generate a single value from the 5 serial
dilutions.1 Loading controls2 and topographical normalization3 procedures will account for protein
concentration and background staining variations. Analysis using unbiased clustering, perturbation
bootstrap clustering and principle component analysis was then performed as previously described.4 We
(SM Kornblau, KR Coombes (MDACC) and A. Qutub (Rice University) have developed a
novel modification of the Gap statistic, named “stability Gap” for selecting the optimal number of patient
clusters from the range of possible clusters118 This will be utilized to select the optimal number of
clusters for comparison in outcomes analysis. Analysis typically places samples into 3-7 clusters for further
analysis.
Comparison of the protein levels between paired samples will be done by performing paired t-
tests. Association between protein expression levels and categorical clinical variables will be assessed in
R using standard t tests, linear regression or mixed-effects linear models. Association between continuous
variable and protein levels will be assessed by using Pearson and Spearman correlation and linear
regression. Bonferroni corrections were performed to account for multiple statistical parameters for
calculating statistical significance. The Kaplan-Meier method was used to generate the survival
curves. Univariate and multivariate Cox proportional hazard modeling will be performed to investigate
association with survival with protein levels as categorized variables using Statistica version 10 software
(StatSoft, Tulsa, OK).
Other methods of analysis may include: Hierarchical Clustering, Principal Component Analysis
(PCA), Self-Organizing Maps (SOM) and other class discovery methods.119 Cluster stability will be
accessed using reproducibility measures, including GAP,120 and Stability Gap (manuscript in preparation),
as well as robustness and discrepancy indices.121 Differentially expressed proteins will be found using
paired t-test as well as repeated measures, mixed effect ANOVA and ANOVA with contrasts. To determine
if dynamic changes in specific protein pathways predict chemoresistance, we will use different regression
and classification methods: Self Organizing Maps(SOM) (when no outcome is used), class prediction
methods such as logistic regression, Support Vector Machine,122 Random Forest,123 Binary Tree Prediction,
Bayesian Compound Covariate Predictor, and Discriminant analysis
(http://linus.nci.nih.gov/techreport/Manual32.pdf).
For Specific Aim 2, we will determine which patients classified as low-risk (based on time and site of
relapse) that would benefit from the more intensive chemotherapy given to high-risk patients (i.e.,
reassignment to the high risk group). Following the generation of a risk-based protein expression classifier
from all low-risk patients, further testing of the putative classifier will be based on performance
characteristics using predefined cutpoints using ROC curves. Comparisons will be made using the AUC of
ROC curves between standard risk stratification (time and site of relapse), MRD response, and the putative
protein expression classifiers. Model overfitting and biased assessment of model performance will be
minimized using bootstrap clustering as previously described 105ROC curves will be generated for each
stratification group, as well as for combinations of groups. Since attainment of CR following subsequent
relapse is suboptimal,124,125 and DFS is guarded following subsequent relapse, protein expression classifiers
that maximize specificity with adequate sensitivity will be prioritized for further characterization.
o) Marker prevalence:
Since the methods will analyze multiple proteins (RPPA) and signal transduction pathways (phosphoflow),
the prevalence of each marker will vary.
p) Estimate what proportion of patients on a therapeutic trial will have available sample for
correlative study analysis; discuss possible biases.
Based on prior samples obtained for RPPA analysis from patients enrolled on the Phase 3 COG AML trial
AAML1031, we estimate 40% of enrollments will provide an evaluable sample. As the most common
reasons for sample dropout include technical issues (20% sample dropout due to sample quality, 40% due
to samples not being drawn at site, 10% due to delays in shipping), lack of consent (10-15%), and
insufficient lymphoblasts in peripheral blood to qualify for the study (20%). The latter 20% will have a
bias toward high-risk disease and early marrow relapse.
Specific aim 2: To determine if alterations in specific cell stress proteins can be used to generate a “high-
risk” protein expression signature that identifies patients stratified to the low-risk (based on time and site
of relapse) group that would have benefited from stratification into the high-risk group. For analysis of
clinical utility, assumptions are usually made based on prior clinical trials for a similar pediatric relapsed
ALL cohort. However, previous relapsed ALL COG studies have used different risk assessments, making
a priori cutpoint determination challenging. Therefore, we will present basic principles for determining
cutpoints for protein expression classifiers.
To aid in determining specific cutpoints, data from the recently completed AALL0433 clinical trial will be
used to determine the prevalence of relapse and DFS in a similar LR population.
Since our aim is to improve the detection of true high risk patients misclassified into the low-risk
group, we are interested in both the sensitivity and specificity of a putative protein classifier. The clinical
usefulness of a protein expression classifier is dependent the proportion of low-risk patients that relapse
(which can be estimated from the AALL0433 trial), as well as the proportion of low-risk patients with the
“high-risk” putative classifier (determined by this study). Depending on the change in DFS for those
correctly identified as having a “high-risk” protein classifier, the relative risks that can be used to identify
the clinical usefulness can be estimated. Once these estimates are known, we can estimate both absolute
risk reduction and decrease in relative risk of a low-risk patient with a “high-risk” classifier. Our goal would
be to identify a classification signature that outperforms the current method of risk stratification (time and
site of relapse) and/or allowed for an absolute risk reduction of at least 5% in the low-risk group.
In addition to determining the effect sizes (defined as the difference in the corresponding mean outcomes
over a common standard deviation) of assays between treatment groups, we will also compare protein
classifiers with clinical outcome (MRD status and EFS). For the analysis of data across time (0h, 6h and
24h) we do not know a priori the most relevant time points to compare. Short-term differences could have
returned to baseline by 24h, and may be most accurately estimated by changes between 0h and 6h. Long-
term differences would be best measured by 0h-24h comparison. Complex changes, such as increase at 6h
and decreases by 24h (seen with NF-κB after chemotherapy) are best measured using all three time points
as descriptors. However, for simplicity we have provided the affect sizes for changes from 0h to 24h in
Table XI-1.
Table XI-1: Effect-size differences based on sample size for protein cell stress pathways
1. RPPA and phospho-flow Samples size (max) Effect size *
studies
Baseline and change at 24h by 135 (67/tx group) 0.488
treatment arm (blinatumomab
therapy vs non-blinatumomab 110 (55/tx group) 0.539
therapy; randomized patients only)
Baseline and change at 24h by All patients:
MRD response (rate of MRD >= 215 (84 MRD >=0.1%/ 131 MRD 0.393
0.1%: 62% for high-risk, 28% for < 0.1%) for baseline comparisons.
low-risk; 39% overall)
176 (69 MRD >=0.1%/107 MRD < 0.435
0.1%) for changes over time.
Low-risk:
145 (41 MRD >=0.1%/ 104 MRD
< 0.1%) for baseline comparisons. 0.520
Baseline and change at 24h by 3- 135 (86 no event/ 49 with event) for 0.505
year DFS for randomized patients baseline changes.
(estimated 3-year DFS 64%)
110 (70 no event/ 40 with event) 0.560
For changes over time.
Assumptions: alpha = 0.05, beta = 0.2.Abbreviations: grp= group, tx = treatment
* With this effect size, the study will have at least 80% power at 2-sided significance level of 0.05 to
detect such difference given the corresponding sample size.
Table 1 outlines standard criteria for GVHD organ staging. However, confounding clinical syndromes (such
as non-GVHD causes of hyperbilirubinemia) may make staging GVHD in a given organ difficult. In
addition, timing of organ specific symptoms affects whether that symptom is more or less likely to be true
GVHD. Please refer to Tables 2 and 3 to assist you in deciding whether to attribute these clinical findings
to GVHD, especially in situations where a biopsy is not possible. For additional help, please see the text
which follows the tables. Table 4 reviews the approach to assessing GVHD as acute, chronic, or the overlap
between the two.
Finally, engraftment syndrome will be reported separately from the GVHD scoring presented below.
Engraftment Syndrome
A clinical syndrome of fever, rash, respiratory distress, and diarrhea has been described, just prior
to engraftment in patients undergoing unrelated cord blood and mismatched transplantation. If, in
the judgment of the local investigator, a patient experiences this syndrome, details of the event
should be reported when requested in the study CRFs.
Modified Glucksberg Staging Criteria for Acute Graft versus Host Disease
Table 1 Organ Staging (See tables and text below for details)
Stage Skin Liver (bilirubin) Gut (stool output/day)
0 No GVHD rash < 2 mg/dL Adult: < 500 mL/day
Child: < 10 mL/kg/day
For GI staging: The “adult” stool output values should be used for patients > 50 kg in weight. Use
3 day averages for GI staging based on stool output. If stool and urine are mixed, stool output is
presumed to be 50% of total stool/urine mix (see Section 3.2).
For stage 4 GI: the term “severe abdominal pain” will be defined as:
(a) Pain control requiring institution of opioid use, or an increase in on-going opioid use, PLUS
(b) Pain that significantly impacts performance status, as determined by the treating MD.
If colon or rectal biopsy is +, but stool output is < 500 mL/day (< 10 mL/kg/day), then consider as
GI stage 0.
Table 2 Evaluating Liver GVHD in the Absence of Biopsy Confirmation (See Table 3.0 below)
Establishing liver GVHD with skin or GI GVHD and other cause of hyperbilirubinemia
Skin and/or GI GVHD Worsening bilirubin level (includes Stable or improving bilirubin after
present worsening just prior to onset of skin diagnosis of skin or GI GVHD,
or GI tract GVHD) OR stable irrespective of treatment:
elevated bilirubin despite resolution Do not stage as liver GVHD
of non-GVHD cause of increased
bilirubin:
Stage as liver GVHD
Skin and/or GI GVHD Liver GVHD is staged according to the Glucksberg criteria. The
worsening elevated bili is attributed to GVHD alone.
There is one exception to this: the diagnosis of TTP, with high LDH and
unconjugated bilirubin precludes the diagnosis and staging of new liver
GVHD in the absence of a confirmatory liver biopsy.
Table 3 Evaluating GI GVHD in the Absence of Biopsy Confirmation (See Table 4.0 below)
Establishing GI GVHD with new onset diarrhea and no skin or liver GVHD
No Skin/liver GVHD Assume no GI GVHD, unless proven by biopsy
Day 0 through
engraftment
No Skin/liver GVHD NO other etiology of diarrhea Any other etiology of diarrhea
Engraftment through identified: identified:
day 100 Stage as GI GVHD Do not stage as GI GVHD
Chronic GVHD
Classic chronic GVHD No time limit No Yes
Overlap syndrome No time limit Yes Yes
• Scoring of acute GVHD may need to occur past day 100. In particular, patients should continue to
be scored for acute GVHD when classic acute GVHD symptoms (maculopapular rash, nausea,
vomiting, anorexia, profuse diarrhea - particularly if bloody and ileus) persist past day 100 or if
identical symptoms previously scored as acute GVHD resolve and then recur within 30 days during
immunosuppression taper but past day 100.
• Those patients being scored as having acute GVHD should NOT have diagnostic or distinctive
signs of chronic GVHD.
• Patients with both acute and chronic symptoms should be diagnosed as having Overlap
Syndrome and scored according to their chronic GVHD score.
1.1 Presence or Absence of Skin GVHD: Skin GVHD will be considered present if a rash characteristic
of acute GVHD develops after allogeneic marrow transplantation involving more than 25% of the
body surface not clearly attributable to causes such as drug administration or infection. The extent of
the body surface area involved can be estimated by the “Rule of Nines”. In estimating the extent of
skin GVHD, the area involved is calculated for individual anatomic areas, such as the arm or leg, and
then the total is derived from a simple summation. Areas that are non-blanching should not be
considered involved regardless of the overlying color of the rash (red, brown, etc.). Limited
distribution erythema (with the exception of palms and soles) in the absence of associated rash
elsewhere on the body will not be considered GVHD.
A. Hyperbilirubinemia (total bilirubin > 2.0 mg/dL) in the absence of other signs of acute GVHD in
the skin or GI tract:
i) Day 0-35: If hyperbilirubinemia alone is present with no other signs of acute GVHD in other
organ systems, acute GVHD will not be diagnosed based solely on laboratory abnormalities.
Acute GVHD will be diagnosed if findings on histopathology studies of liver from a biopsy or
autopsy are confirmatory.
B. Pre-existing hyperbilirubinemia clearly attributed to an etiology other than acute GVHD in the
presence of signs of acute GVHD in other organ systems.
ii) If hyperbilirubinemia worsens several days before or at the time of onset of signs of acute GVHD
in other organ systems, GVHD will be considered to be present unless histopathology studies
of liver are available and negative on a biopsy during that time interval or autopsy results
exclude GVHD.
iii) If hyperbilirubinemia persists and is not improving after resolution of a pre-existing non-GVHD
liver disease process (e.g. localized infection of liver, systemic sepsis, biliary tract obstruction)
when signs of acute GVHD are present in other organ systems or no other intervening cause
has been diagnosed, then acute GVHD will be considered to be present in the absence of a new,
clearly identifiable cause of non-GVHD liver disease or unless a liver biopsy or autopsy
specimen is negative.
C. Prior acute GVHD in liver with new onset of a disease process that exacerbates pre-existing or
recently resolved hyperbilirubinemia:
be carried forward until the new disease process resolves). Example: Acute GVHD of the liver and
gut is diagnosed on day 20. Treatment of acute GVHD results in falling bilirubin levels to liver
stage 1. Sepsis or TTP develops with transient worsening of the hyperbilirubinemia. The liver stage
is not increased, despite a higher bilirubin level, because the cause of worsening hyperbilirubinemia
is attributed to sepsis or TTP.
ii) If an etiology other than acute GVHD is clearly identified as causing or exacerbating
hyperbilirubinemia in the presence of already worsening acute liver GVHD or GVHD of the skin
or GI tract is simultaneously worsening, then the liver GVHD will be staged according to the actual
bilirubin level, even though another cause of hyperbilirubinemia is present.
3.1 Assessing for the Presence or Absence of GVHD of the Gastrointestinal Tract
A. Diarrhea (> 500 mL/day in adults or > 10 mL/kg in pediatric patients) in the absence of other signs
of acute GVHD in other organ systems
i) Day 0-engraftment: If diarrhea alone is present without other signs of acute GVHD in other
organ systems, acute GVHD will not be considered present. Diarrhea will be attributed to acute
GVHD if histopathology studies of gastrointestinal tract from a biopsy or autopsy are
diagnostic.
ii) Engraftment-day 100: If diarrhea persists and is not improving, is exacerbated, or develops de
novo in the absence of acute GVHD in other organ systems, histopathology studies of gut
biopsies or from autopsy specimens are not available, and no other etiologies are clearly
identified, acute GVHD will be considered to be the cause. A stool specimen should be
examined to rule out infectious causes (e.g. rotavirus, adenovirus, and C. difficile toxin). It is
recommended, if at all possible, that biopsies be obtained for diagnostic purposes.
B. Pre-existing diarrhea clearly attributed to an etiology other than acute GVHD in the presence of
signs of acute GVHD in other organ systems:
i) If pre-existing diarrhea caused by a process other than GVHD has been documented clinically
or by lab assessment and is stable or improving at the onset of signs of acute GVHD in the skin
or liver, then acute GVHD of the intestine will not be considered to be present in the absence
of biopsy confirmation or autopsy report.
ii) If diarrhea or gastrointestinal symptoms are already present, but worsen significantly at the
time of onset of signs of acute GVHD in the skin or liver, GVHD will be considered present,
unless biopsy or autopsy are negative.
iii) If diarrhea persists after resolution of a pre-existing disease process with signs of acute
GVHD present in other organ systems, GVHD will be considered present, unless biopsy or
autopsy are negative.
C. Prior or present acute GVHD in other organ systems with new onset of diarrhea:
If diarrhea is clearly attributable to an etiology other than acute GVHD (e.g., infection) and a
history of acute GVHD exists or acute GVHD is present in other organ systems and is stable, then
the gastrointestinal tract will not be evaluable for acute GVHD until the resolution or stabilizing of
the other disease process (e.g., infection) in the absence of biopsy or autopsy confirmation.
D. Persistent anorexia, nausea or vomiting in the absence of signs of acute GVHD in other organ
systems:
Persistent anorexia, nausea or vomiting in the absence of other known causes of these
symptoms will be considered stage 1 acute GVHD if confirmed by endoscopic biopsy.
If a biopsy is not possible (e.g. secondary to thrombocytopenia) but the clinical findings
are compatible with acute GVHD, then the patient will be treated and recorded as having
acute GVHD.
3.2 Staging of the Gastrointestinal Tract for the Severity of Acute GVHD
The severity of gastrointestinal tract GVHD will be staged according to modified Glucksberg criteria.
To minimize errors caused by large day-to-day variation, diarrhea volume is measured as an average
over 3 days and reported as the volume in milliliters per day. When urinary mixing is noted the stool
volume will be considered half of the total volume unless nursing staff is able to give a better estimate
from direct observation. Abdominal cramps are considered significant for staging if the severity results
in a clinical intervention (e.g. analgesia, fasting, etc.). Blood in the stools is considered significant if
the blood is visible or hematochezia/ melena is present and not clearly attributed to a cause other than
GVHD (e.g. epistaxis/ hemorrhoids).
• a completed Statement of Investigator Form (FDA Form 1572) with an original signature
• a current Curriculum Vitae (CV)
• a completed and signed Supplemental Investigator Data Form (IDF)
• a completed Financial Disclosure Form (FDF) with an original signature
Fillable PDF forms and additional information can be found on the CTEP website at
<http://ctep.cancer.gov/investigatorResources/investigator_registration.htm>. For questions, please
contact the CTEP Investigator Registration Help Desk by email at
<[email protected]>.
The Cancer Therapy Evaluation Program (CTEP) Identity and Access Management (IAM)
application is a web-based application intended for use by both Investigators (i.e., all physicians
involved in the conduct of NCI-sponsored clinical trials) and Associates (i.e., all staff involved in
the conduct of NCI-sponsored clinical trials).
Associates will use the CTEP-IAM application to register (both initial registration and annual re-
registration) with CTEP and to obtain a user account.
Investigators will use the CTEP-IAM application to obtain a user account only. (See CTEP
Investigator Registration Procedures above for information on registering with CTEP as an
Investigator, which must be completed before a CTEP-IAM account can be requested.)
An active CTEP-IAM user account will be needed to access all CTEP and CTSU (Cancer Trials
Support Unit) websites and applications, including the CTSU members’ website.
This study is supported by the NCI Cancer Trials Support Unit (CTSU).
• CTSU IRB Certification (for sites not participating via the CIRB)
• CTSU IRB/Regulatory Approval Transmittal Sheet (for sites not participating via the NCI
CIRB)
Submit completed forms along with a copy of your IRB Approval to the CTSU Regulatory Office,
where they will be entered and tracked in the CTSU RSS.
CTSU Regulatory Office
1818 Market Street, Suite 1100
Philadelphia, PA 19103
Phone: 1-866-651-2878
Fax: 215-569-0206
E-mail: [email protected] (for regulatory document submission only)
Check the status of your site’s registration packets by querying the RSS site registration status page
of the members’ section of the CTSU website. (Note: Sites will not receive formal notification of
regulatory approval from the CTSU Regulatory Office.)
Go to https://www.ctsu.org and log in to the members’ area using your CTEP-IAM username
and password
Click on the Regulatory tab at the top of your screen
Click on the Site Registration tab
Enter your 5-character CTEP Institution Code and click on Go
The lists below do not include everything that may interact with chemotherapy. Study Subjects and/or
their Parents should be encouraged to talk to their doctors before starting any new medications, using
over-the-counter medicines, or herbal supplements and before making a significant change in diet.
Cyclophosphamide
Cyclosporine
Dexamethasone
Etoposide
• Arthritis medications
• Leflunomide, tofacitinib
• Anti-rejection medications
• Cyclosporine, tacrolimus
• Antiretrovirals and antivirals
• Atazanavir, boceprevir, darunavir, delaviridine, efavirenz, etravirine, fosamprenavir,
indinavir, lopinavir, nelfinavir, nevirapine, ritonavir, saquinavir, Stribild, telaprevir,
tipranavir
• Anti-seizure medications
• Carbamazepine, oxcarbazepine, phenobarbital, phenytoin, primidone
• Heart medications
• Amiodarone, dronedenarone, verapamil
• Some chemotherapy (be sure to talk to your doctor about this)
• Many other drugs, including the following:
• Aprepitant, atovaquone, bosentan, deferasirox, dexamethasone, ivacaftor, lomitapide,
mifepristone, natalizumab, pimozide, sitaxentan
Fludarabine
Leucovorin
• Folic acid
*Supplements may come in many forms, such as teas, drinks, juices, liquids, drops, capsules, pills, or
dried herbs. All forms should be avoided.
• Alcohol
• Echinacea
• Some vitamins, including those that contain folic acid or high doses of vitamin C
*Supplements may come in many forms, such as teas, drinks, juices, liquids, drops, capsules, pills, or
dried herbs. All forms should be avoided.
Mercaptopurine
Mitoxantrone
Mycophenolate mofetil
Pegaspargase
Tacrolimus
Thioguanine.
Drugs that may interact with thioguanine*
• Arthritis medications: leflunomide, tofacitinib
• Other medications, such as allopurinol, clozapine, natalizumab, olsalazine, sulfasalazine
**Supplements may come in many forms, such as teas, drinks, juices, liquids, drops, capsules, pills, or
dried herbs. All forms should be avoided.
Thiotepa
Drugs that may interact with thiotepa*
• Echinacea
*Supplements may come in many forms, such as teas, drinks, juices, liquids, drops, capsules, pills, or
dried herbs. All forms should be avoided.
Vincristine
Drugs that may interact with vincristine
• Antibiotics
• Clarithromycin, erythromycin, nafcillin, rifabutin, rifampin, telithromycin
• Antidepressants and antipsychotics
• Aripiprazole, nefazodone, trazodone
• Antifungals
• Fluconazole, itraconazole, ketoconazole, posaconazole, voriconazole
• Arthritis medications
• Leflunomide, tocilizumab, tofacitinib
• Anti-rejection medications
• Cyclosporine, tacrolimus
• Antiretrovirals and antivirals
• Atazanavir, boceprevir, darunavir, delaviridine, efavirenz, etravirine, fosamprenavir,
indinavir, lopinavir, nelfinavir, nevirapine, ritonavir, saquinavir, Stribild, telaprevir,
tenofovir, tipranavir
• Anti-seizure medications
• Carbamazepine, oxcarbazepine, phenobarbital, phenytoin, primidone
• Heart medications
• Amiodarone, digoxin, dronedenarone, propranolol, verapamil
• Some chemotherapy (be sure to talk to your doctor about this)
• Many other drugs, including the following:
• Aprepitant, deferasirox, ivacaftor, lomitapide, mifepristone, natalizumab, pimozide,
warfarin
REFERENCES:
1. Nguyen K, Devidas M, Cheng SC, et al: Factors influencing survival after relapse from acute
lymphoblastic leukemia: a Children's Oncology Group study. Leukemia 22:2142-50, 2008
2. Handgretinger R, Zugmaier G, Henze G, et al: Complete remission after blinatumomab-induced
donor T-cell activation in three pediatric patients with post-transplant relapsed acute
lymphoblastic leukemia. Leukemia 25:181-4, 2012
3. Klinger M, Brandl C, Zugmaier G, et al: Immunopharmacologic response of patients with B-
lineage acute lymphoblastic leukemia to continuous infusion of T cell-engaging CD19/CD3-
bispecific BiTE antibody blinatumomab. Blood 119:6226-33, 2012
4. Topp MS, Gokbuget N, Zugmaier G, et al: Long-term follow-up of hematologic relapse-free
survival in a phase 2 study of blinatumomab in patients with MRD in B-lineage ALL. Blood
120:5185-7, 2012
5. Topp MS, Kufer P, Gokbuget N, et al: Targeted therapy with the T-cell-engaging antibody
blinatumomab of chemotherapy-refractory minimal residual disease in B-lineage acute
lymphoblastic leukemia patients results in high response rate and prolonged leukemia-free
survival. J Clin Oncol 29:2493-8, 2011
6. Lia Gore, Gerhard Zugmaier, Rupert Handgretinger, et al: Cytological and molecular remissions
with blinatumomab treatment in second or later bone marrow relapse in pediatric acute
lymphoblastic leukemia (ALL). J Clin Oncol 31, 2013 (suppl; abstr 10007). Published on
Meeting Library (http://meetinglibrary.asco.org).
7. Raetz EA, Borowitz MJ, Devidas M, et al: Reinduction platform for children with first marrow
relapse of acute lymphoblastic Leukemia: A Children's Oncology Group Study[corrected]. J Clin
Oncol 26:3971-8, 2008
8. Tallen G, Ratei R, Mann G, et al: Long-term outcome in children with relapsed acute
lymphoblastic leukemia after time-point and site-of-relapse stratification and intensified short-
course multidrug chemotherapy: results of trial ALL-REZ BFM 90. J Clin Oncol 28:2339-47,
2010
9. Parker C, Waters R, Leighton C, et al: Effect of mitoxantrone on outcome of children with first
relapse of acute lymphoblastic leukaemia (ALL R3): an open-label randomised trial. Lancet
376:2009-17, 2010
10. Fan C, Cool JC, Scherer MA, et al: Damaging effects of chronic low-dose methotrexate usage on
primary bone formation in young rats and potential protective effects of folinic acid
supplementary treatment. Bone 44:61-70, 2009
11. Teachey DT, Rheingold SR, Maude SL, et al: Cytokine release syndrome after blinatumomab
treatment related to abnormal macrophage activation and ameliorated with cytokine-directed
therapy. Blood 121:5154-7, 2013
12. Paganin M, Zecca M, Fabbri G, et al: Minimal residual disease is an important predictive factor
of outcome in children with relapsed 'high-risk' acute lymphoblastic leukemia. Leukemia
22:2193-200, 2008
13. Attarbaschi A, Mann G, Panzer-Grumayer R, et al: Minimal residual disease values discriminate
between low and high relapse risk in children with B-cell precursor acute lymphoblastic leukemia
and an intrachromosomal amplification of chromosome 21: the Austrian and German acute
lymphoblastic leukemia Berlin-Frankfurt-Munster (ALL-BFM) trials. J Clin Oncol 26:3046-50,
2008
14. Eckert C, Biondi A, Seeger K, et al: Prognostic value of minimal residual disease in relapsed
childhood acute lymphoblastic leukaemia. Lancet 358:1239-41, 2001
15. Lew G, Lu X, Yanofsky R, et al: The significance of minimal residual disease (MRD) in relapsed
childhood B-lymphoblastic leukemia (B-ALL): A report from Children's Oncology Group (COG)
protocol AALL0433.
16. Barredo JC, Devidas M, Lauer SJ, et al: Isolated CNS relapse of acute lymphoblastic leukemia
treated with intensive systemic chemotherapy and delayed CNS radiation: a pediatric oncology
group study. J Clin Oncol 24:3142-9, 2006
17. Ritchey AK, Pollock BH, Lauer SJ, et al: Improved survival of children with isolated CNS
relapse of acute lymphoblastic leukemia: a pediatric oncology group study. J Clin Oncol 17:3745-
52, 1999
18. Goulden N, Langlands K, Steward C, et al: PCR assessment of bone marrow status in 'isolated'
extramedullary relapse of childhood B-precursor acute lymphoblastic leukaemia. Br J Haematol
87:282-5, 1994
19. Neale GA, Pui CH, Mahmoud HH, et al: Molecular evidence for minimal residual bone marrow
disease in children with 'isolated' extra-medullary relapse of T-cell acute lymphoblastic leukemia.
Leukemia 8:768-75, 1994
20. Schrappe M, Zimmermann M, Moricke A, et al: Dexamethasone in Induction Can Eliminate One
Third of All Relapses in Childhood Acute Lymphoblastic Leukemia (ALL): Results of An
International Randomized Trial in 3655 Patients (Trial AIEOP-BFM ALL 2000). ASH Annual
Meeting Abstracts 112:7-, 2008
21. Silverman LB, Gelber RD, Dalton VK, et al: Improved outcome for children with acute
lymphoblastic leukemia: results of Dana-Farber Consortium Protocol 91-01. Blood 97:1211-8,
2001
22. Gaynon PS, Trigg ME, Heerema NA, et al: Children's Cancer Group trials in childhood acute
lymphoblastic leukemia: 1983-1995. Leukemia 14:2223-33, 2000
23. Hijiya N, Liu W, Sandlund JT, et al: Overt testicular disease at diagnosis of childhood acute
lymphoblastic leukemia: lack of therapeutic role of local irradiation. Leukemia 19:1399-403,
2005
24. Sirvent N, Suciu S, Bertrand Y, et al: Overt testicular disease (OTD) at diagnosis is not associated
with a poor prognosis in childhood acute lymphoblastic leukemia: results of the EORTC CLG
Study 58881. Pediatr Blood Cancer 49:344-8, 2007
25. van den Berg H, de Groot-Kruseman HA, Damen-Korbijn CM, et al: Outcome after first relapse
in children with acute lymphoblastic leukemia: a report based on the Dutch Childhood Oncology
Group (DCOG) relapse all 98 protocol. Pediatr Blood Cancer 57:210-6, 2011
26. van den Berg H, Langeveld NE, Veenhof CH, et al: Treatment of isolated testicular recurrence of
acute lymphoblastic leukemia without radiotherapy. Report from the Dutch Late Effects Study
Group. Cancer 79:2257-62, 1997
27. Dordelmann M, Schrappe M, Reiter A, et al: Down's syndrome in childhood acute lymphoblastic
leukemia: clinical characteristics and treatment outcome in four consecutive BFM trials. Berlin-
Frankfurt-Munster Group. Leukemia 12:645-51, 1998
28. Freeman AI, Weinberg V, Brecher ML, et al: Comparison of intermediate-dose methotrexate with
cranial irradiation for the post-induction treatment of acute lymphocytic leukemia in children. N
Engl J Med 308:477-84, 1983
29. Tsuchida M, Ohara A, Manabe A, et al: Long-term results of Tokyo Children's Cancer Study
Group trials for childhood acute lymphoblastic leukemia, 1984-1999. Leukemia 24:383-96, 2010
30. Bader P, Kreyenberg H, Henze GH, et al: Prognostic value of minimal residual disease
quantification before allogeneic stem-cell transplantation in relapsed childhood acute
lymphoblastic leukemia: the ALL-REZ BFM Study Group. J Clin Oncol 27:377-84, 2009
31. Sramkova L, Muzikova K, Fronkova E, et al: Detectable minimal residual disease before
allogeneic hematopoietic stem cell transplantation predicts extremely poor prognosis in children
with acute lymphoblastic leukemia. Pediatr Blood Cancer 48:93-100, 2007
32. Knechtli CJ, Goulden NJ, Hancock JP, et al: Minimal residual disease status before allogeneic
bone marrow transplantation is an important determinant of successful outcome for children and
adolescents with acute lymphoblastic leukemia. Blood 92:4072-9, 1998
33. Pulsipher MA, Langholz B, Wall DA, et al: The Relationship of Acute Gvhd and Pre- and Post-
Transplant Flow-MRD to the Incidence and Timing of Relapse in Children Undergoing
Allogeneic Transplantation for High Risk ALL: Defining a Target Population and Window for
Immunological Intervention to Prevent Relapse. ASH Annual Meeting Abstracts 120:470-, 2012
34. Bader P, Kreyenberg H, Hoelle W, et al: Increasing mixed chimerism is an important prognostic
factor for unfavorable outcome in children with acute lymphoblastic leukemia after allogeneic
stem-cell transplantation: possible role for pre-emptive immunotherapy? J Clin Oncol 22:1696-
705, 2004
35. Rettinger E, Willasch AM, Kreyenberg H, et al: Preemptive immunotherapy in childhood acute
myeloid leukemia for patients showing evidence of mixed chimerism after allogeneic stem cell
transplantation. Blood 118:5681-8, 2011
36. Horn B, Soni S, Khan S, et al: Feasibility study of preemptive withdrawal of immunosuppression
based on chimerism testing in children undergoing myeloablative allogeneic transplantation for
hematologic malignancies. Bone Marrow Transplant 43:469-76, 2009
37. Schwartz GJ, Gauthier B: A simple estimate of glomerular filtration rate in adolescent boys. J
Pediatr 106:522-6, 1985
38. Relling MV, Pui CH, Sandlund JT, et al: Adverse effect of anticonvulsants on efficacy of
chemotherapy for acute lymphoblastic leukaemia. Lancet 356:285-90, 2000
39. Bermudez M, Fuster JL, Llinares E, et al: Itraconazole-related increased vincristine neurotoxicity:
case report and review of literature. J Pediatr Hematol Oncol 27:389-92, 2005
40. Ariffin H, Omar KZ, Ang EL, et al: Severe vincristine neurotoxicity with concomitant use of
itraconazole. J Paediatr Child Health 39:638-9, 2003
41. Przepiorka D, Blamble D, Hilsenbeck S, et al: Tacrolimus clearance is age-dependent within the
pediatric population. Bone Marrow Transplant 26:601-5, 2000
42. Brenner DE, Wiernick PH, Wesley M, et al: Acute doxorubicin toxicity. Relationship to
pretreatment liver function, response, and pharmacokinetics in patients with acute
nonmlumphocytic leukemia. Cancer 53:1042-1048, 1984
43. Langer T, Martus P, Ottensmeier H, et al: CNS late-effects after ALL therapy in childhood. Part
III: neuropsychological performance in long-term survivors of childhood ALL: impairments of
concentration, attention, and memory. Med Pediatr Oncol 38(5):320-8, 2002
44. Mulhern RK, Palmer SL: Neurocognitive late effects in pediatric cancer. Curr Probl Cancer
27(4):177-97, 2003
45. Harila-Saari AH, Paakko EL, Vainionpaa LK, et al: A longitudinal magnetic resonance imaging
study of the brain in survivors in childhood acute lymphoblastic leukemia. Cancer 83(12):2608-
17, 1998
46. Asato R, Akiyama Y, Ito M, et al: Nuclear magnetic resonance abnormalities of the cerebral
white matter in children with acute lymphoblastic leukemia and malignant lymphoma during and
after central nervous system prophylactic treatment with intrathecal methotrexate. Cancer
70(7):1997-2004, 1992
47. Iuvone L, Mariotti P, Colosimo C, et al: Long-term cognitive outcome, brain computed
tomography scan, and magnetic resonance imaging in children cured for acute lymphoblastic
leukemia. . Cancer 95(12):2562-70, 2002
48. Paakko E, Vainionpaa L, Pyhtinen J, et al: Minor changes on cranial MRI during treatment in
children with acute lymphoblastic leukaemia. Neuroradiology 38(3):264-8, 1996
49. Biti GP, Magrini SM, Villari N, et al: Brain damage after treatment for acute lymphoblastic
leukemia. A report on 34 patients with special regard to MRI findings. Acta Oncol 28(2):253-6,
1989
50. Wilson DA, Nitschke R, Bowman ME, et al: Transient white matter changes on MR images in
children undergoing chemotherapy for acute lymphocytic leukemia: correlation with
neuropsychologic deficiencies. Radiology 180(1):205-9, 1991
51. Schrappe M, Reiter A, Ludwig WD, et al: Improved outcome in childhood acute lymphoblastic
leukemia despite reduced use of anthracyclines and cranial radiotherapy: results of trial ALL-
BFM 90. German-Austrian-Swiss ALL-BFM Study Group. Blood 95(11):3310-22, 2000
52. Brouwers P, Riccardi R, Fedio P, et al: Long-term neuropsychologic sequelae of childhood
leukemia: correlation with CT brain scan abnormalities. J Pediatr Hematol/Oncol 106(5):723-8,
1985
53. Rollins N, Winick N, Bash R, et al: Acute methotrexate neurotoxicity: findings on diffusion-
weighted imaging and correlation with clinical outcome. AJNR Am J Neuroradiol 25(10):1688-
95, 2004
54. Laitt RD, Chambers EJ, Goddard PR, et al: Magnetic resonance imaging and magnetic resonance
angiography in long term survivors of acute lymphoblastic leukemia treated with cranial
irradiation. Cancer 76(10):1846-52, 1995
55. Kramer JH, Norman D, Brant-Zawadzki M, et al: Absence of white matter changes on magnetic
resonance imaging in children treated with CNS prophylaxis therapy for leukemia. Cancer
61(5):928-30, 1988
56. Winick NJ, Bowman WP, Kamen BA, et al: Unexpected acute neurologic toxicity in the
treatment of children with acute lymphoblastic leukemia. J Natl Cancer Inst. 84(4):252-6, 1992
57. Bleyer WA: Neurologic sequelae of methotrexate and ionizing radiation: a new classification.
Cancer Treat Rep 65 89-98, 1981
58. Meadows AT, Gordon J, Massari DJ, et al: Declines in IQ scores and cognitive dysfunctions in
children with acute lymphocytic leukaemia treated with cranial irradiation. Lancet 2(8254):1015-
8, 1981
59. Williams JM, Ochs J, Davis KS, et al: The subacute effects of CNS prophylaxis for acute
lymphoblastic leukemia on neuropsychological performance: a comparison of four protocols.
Arch Clin Neuropsychol 1(2):183-92, 1986
60. Mulhern RK, Fairclough D, Ochs J: A prospective comparison of neuropsychologic performance
of children surviving leukemia who received 18-Gy, 24-Gy, or no cranial irradiation. J Clin
Oncol. 9(8):1348-56, 1991
61. Steinberg GK, Kunis D, DeLaPaz R, et al: Neuroprotection following focal cerebral ischaemia
with the NMDA antagonist dextromethorphan, has a favourable dose response profile. Neurol
Res. 15(3):174-80, 1993
62. Steinberg GK, Bell TE, Yenari MA: Dose escalation safety and tolerance study of the N-methyl-
D-aspartate antagonist dextromethorphan in neurosurgery patients. J Neurosurg 84(5):860-6,
1996
63. Blin O, Azulay JP, Desnuelle C, et al: A controlled one-year trial of dextromethorphan in
amyotrophic lateral sclerosis. Clin Neuropharmacol 19(2):189-92, 1996
64. Gredal O, Werdelin L, Bak S, et al: A clinical trial of dextromethorphan in amyotrophic lateral
sclerosis. Acta Neurol Scand 96(1):8-13, 1997
65. Hollander D, Pradas J, Kaplan R, et al: High-dose dextromethorphan in amyotrophic lateral
sclerosis: phase I safety and pharmacokinetic studies. Ann Neurol. 36(6):920-4, 1994
66. Wiedemann BC, Balis FM, Murphy RF, et al: Carboxypeptidase-G2, thymidine, and leucovorin
rescue in cancer patients with methotrexate-induced renal dysfunction. J Clin Oncol 15(5):2125-
34, 1997
67. DeAngelis LM, Tong WP, Lin S, et al: Carboxypeptidase G2 rescue after high-dose
methotrexate. J Clin Oncol 14(7):2145-9, 1996
68. Weinshilboum R. M, Sladek S. L: Mercaptopurine pharmacogenetics: monogenic inheritance of
erythrocyte thiopurine methyltransferase activity. Am.J.Hum.Genet 32:651-662, 1980
69. Bostrom B, Erdmann G: Cellular pharmacology of 6-mercaptopurine in acute lymphoblastic
leukemia. Am.J.Pediatr.Hematol.Oncol. 15:80-86, 1993
70. Weinshilboum R: Thiopurine pharmacogenetics: clinical and molecular studies of thiopurine
methyltransferase. Drug Metab Dispos 29:601-605, 2001
112. Neeley ES, Kornblau SM, Coombes KR, et al: Variable slope normalization of reverse phase
protein arrays. Bioinformatics 25:1384-9, 2009
113. Kornblau SM, Qutub A, Yao H, et al: Proteomic profiling identifies distinct protein patterns in
acute myelogenous leukemia CD34+CD38- stem-like cells. PLoS One 8:e78453, 2013
114. Maude SL, Tasian SK, Vincent T, et al: Targeting JAK1/2 and mTOR in murine xenograft
models of Ph-like acute lymphoblastic leukemia. Blood 120:3510-8, 2012
115. Tasian SK, Doral MY, Borowitz MJ, et al: Aberrant STAT5 and PI3K/mTOR pathway signaling
occurs in human CRLF2-rearranged B-precursor acute lymphoblastic leukemia. Blood 120:833-
42, 2012
116. Tasian SK, Li Y, Ryan T, et al: In Vivo Efficacy of PI3K Pathway Signaling Inhibition for
Philadelphia Chromosome-Like Acute Lymphoblastic Leukemia. Blood 122(21): ASH Annual
Meeting 2013 abstract #672, 2013
117. Gill S and Tasian SK, Ruella M, Shestova O, et al: Efficacy against Human Acute Myeloid
Leukemia and Myeloablation of Normal Hematopoiesis in a Mouse Model Using Chimeric
Antigen Receptor-Modified T-Cells. Blood 123: accepted for publication, 2014
118. SM Kornblau CH, YH Qiu, SY Yoo, N Xhang, TM Kadia, A Ferrajoli, KR Coombes, AA Qutub:
Hippo Pathway (HP) activity in AML: different prognostic implications of TAZ vs. YAP
inactivation by phosphorylation. Blood (2013), 2013
119. Dupuy A, Simon RM: Critical review of published microarray studies for cancer outcome and
guidelines on statistical analysis and reporting. J Natl Cancer Inst 99:147-57, 2007
120. Tibshirani R, Walther G, T. H: Estimating the number of data clusters via the gap statistic.
J.R.Statist.Doc.B. 63:10-20, 2001
121. McShane LM, Radmacher MD, Freidlin B, et al: Methods for assessing reproducibility of
clustering patterns observed in analyses of microarray data. Bioinformatics 18:1462-9, 2002
122. Hsu CW, Lin CJ: A comparison of methods for multiclass support vector machines. IEEE Trans
Neural Netw 13:415-25, 2002
123. Statnikov A, Wang L, Aliferis CF: A comprehensive comparison of random forests and support
vector machines for microarray-based cancer classification. BMC Bioinformatics 9:319, 2008
124. Belgaumi AF, Al-Seraihy A, Siddiqui KS, et al: Outcome of risk adapted therapy for
relapsed/refractory acute lymphoblastic leukemia in children. Leuk Lymphoma 54:547-54, 2013
125. Reismuller B, Peters C, Dworzak MN, et al: Outcome of children and adolescents with a second
or third relapse of acute lymphoblastic leukemia (ALL): a population-based analysis of the
Austrian ALL-BFM (Berlin-Frankfurt-Munster) study group. J Pediatr Hematol Oncol 35:e200-4,
2013
126. Benjamini Y, Y. H: Methods of assessing reporducibiolity of clustering patterns observed in
analysis of microarray data. Journal of the Royal Statistical Society B. 57:289-300, 1995