All content on this site is intended for healthcare professionals only. By acknowledging this message and accessing the information on this website you are confirming that you are a Healthcare Professional. If you are a patient or carer, please visit Know ALL.

The ALL Hub uses cookies on this website. They help us give you the best online experience. By continuing to use our website without changing your cookie settings, you agree to our use of cookies in accordance with our updated Cookie Policy

Introducing

Now you can personalise
your ALL Hub experience!

Bookmark content to read later

Select your specific areas of interest

View content recommended for you

Find out more
  TRANSLATE

The ALL Hub website uses a third-party service provided by Google that dynamically translates web content. Translations are machine generated, so may not be an exact or complete translation, and the ALL Hub cannot guarantee the accuracy of translated content. The ALL Hub and its employees will not be liable for any direct, indirect, or consequential damages (even if foreseeable) resulting from use of the Google Translate feature. For further support with Google Translate, visit Google Translate Help.

Steering CommitteeAbout UsNewsletterContact
LOADING
You're logged in! Click here any time to manage your account or log out.
LOADING
You're logged in! Click here any time to manage your account or log out.
2021-08-12T15:49:24.000Z

Consensus definitions for response criteria in pediatric ALL: results from the Ponte-di-Legno consortium

Aug 12, 2021
Share:

Pediatric patients with newly diagnosed acute lymphoblastic leukemia (ALL) are often treated in clinical trials, and it is important to have uniform consensus on definitions of response criteria, as these criteria—including complete remission (CR), treatment failure (TF), minimal residual disease (MRD), and relapse—are germane to treatment decisions.

It is necessary to be able to compare results from protocol to protocol as there are many therapeutic options being investigated, and the goal of each of these investigational studies is to increase event-free survival (EFS) and overall survival (OS) rates and to improve the patient’s quality of life. But which of these parameters are most relevant in ALL clinical trials? EFS is the most widely accepted parameter and is advantageous over OS in that it considers, in addition to death, other clinically important outcomes such as TF, relapse, therapy-related mortality, and second malignancies. OS also may not provide information about the efficacy of first-line therapy, as it is influenced by the efficacy of salvage therapy.

Relapse, primary TF, and CR must be defined consistently for time-to-event from diagnosis to be accurately calculated. Buchmann and colleagues,1 referred to herein as the Ponte-di-Legno (PDL) group representatives, collected and summarized the current definitions of CR, TF, relapse, and other events, and created consensus definitions for use in future trials.

Methods

This was a collaborative project between the PDL group and the following study groups: AIEOP-BFM, ALL-IC-BFM, ALLTogether, CoALL, COG, DCOG, DFCI ALL Consortium, EsPhALL/COG, JCCG, NOPHO, SEHOP-PETHEMA, SFCE, SJCRH, TPOG, and UKALL. Recent protocols from these study groups were reviewed for the definitions of CR, TF, relapse, and events, and a summary was provided to participating study groups. An overview of all definitions was prepared, and the first classifications were established prior to a face-to-face meeting of the PDL group in May 2019.

Terminology

The morphologic classifications of bone marrow (BM) and central nervous system (CNS) involvement are long-standing and are defined here, as they will be referred to in the discussion of the results.

BM classification by cytomorphology is defined as follows:

  • M1 marrow: <5% blasts
  • M2 marrow: ≥5 to <25% blasts
  • M3 marrow: ≥25% blasts
    • COG has historically defined M3 marrow as >25% blasts.

CNS classification by cytomorphology and/or clinical findings (+/− imaging and biopsy) is defined as follows:

  • CNS1: Absence of blasts on cytospin preparation in cerebrospinal fluid (CSF) and no clinical or imaging findings of CNS leukemia
  • CNS2: ≤5/µl nucleated cells in CSF, cytospin preparation positive for blasts, and no clinical or imaging findings of CNS disease
  • CNS3: >5/µl nucleated cells in CSF, cytospin preparation positive for blasts, or clinical or imaging findings of CNS disease
    • COG, EsPhALL/COG, DFCI, NOPHO, SFCE, SJCRH, and TPOG defined CNS3 as ≥5/µl nucleated cells in CSF.

Treatment phases

Most protocols define treatment phases thus:

  • Induction, lasting 4–6 weeks (phase Ia in BFM-type regimens)
  • Consolidation (phase Ib in BFM-type regimens)
  • Intensification, targeting the extramedullary space
  • Re-intensification (or delayed intensification), with or without CNS irradiation

In a small number of cases, the later phases may be replaced by allogeneic hematopoietic stem cell transplantation.

Results

Complete remission

There were large differences between the groups’ definitions of CR regarding timing of CR assessment, threshold levels of leukemic cells to define CR, and detection method, as seen in Table 1.

Table 1. Remission assessment in current treatment protocols for pediatric ALL*

Anatomic sites

Methods

Study group

Timepoint

Exceptions

BM

<5% blasts in BM

Cytomorphology

AIEOP-BFM

Day 33, EOI

 

ALL-IC BFM

Day 33, EOI

CoALL

Day 29, EOI

DCOG

Day 33, EOI

DFCI

Day 32, EOI

JCCG

Day 33, EOI

TPOG

Day 35‒42, EOI

<5% blasts in BM

Cytomorphology + confirmation by FCM-/PCR-MRD/genetics

NOPHO

Day 29, EOI

SJCRH mainly uses FCM-MRD

SEHOP-PETHEMA

EOI

SFCE

Day 35‒42, EOI

SJCRH

Day 38‒42, EOI

<5% blasts in BM

PCR-MRD +/− cytomorphology

UKALL

Day 29, EOI

 

 

<1% blasts in BM

FCM-/PCR-MRD +/− cytomorphology

ALLTogether

EOI as the earliest

 

COG

EOI for early CR, EOC for late CR

EsPhALL/COG

EOC block 3

CNS

CNS1

Cytomorphology, imaging, clinical examination

Every participating study group

See above for respective study group

 

CNS1

Cytomorphology + confirmation by FCM-MRD

ALLTogether

See above for respective study group

If suspect cells are seen, evaluation at EOC (Day 71)

EM

Resolution of leukemic infiltrates

Clinical examination, imaging, histology

ALL-IC

See above for respective study group

DCOG: Retesting of testicular involvement after Prot. M; NOPHO: Retesting
EM involvement at Day 85

COG

DCOG

JCCG

EsPhALL/COG

Reduction of initial leukemic mass to 1/3

Clinical examination, imaging, histology

AIEOP-BFM

See above for respective study group

UKALL: Retesting of testicular involvement at Week 8

ALLTogether

CoALL

DFCI

SEHOP-PETHEMA

SFCE

UKALL

ALL, acute lymphoblastic leukemia; BM, bone marrow; CNS, central nervous system; EM, extramedullary; EOC, end of consolidation; EOI, end of induction; FCM-MRD, flow cytometric minimal residual disease; MRD, minimal residual disease; PCR-MRD, polymerase chain reaction minimal residual disease.
*Adapted from Buchmann, et al.1

Treatment failure

Significant differences between groups were also identified regarding definitions of TF as events, as shown in Table 2.

Table 2. Definitions of treatment failure events*

Event

Methods

Study group

Induction failure (EOI)
≥5% blasts in BM

Cytomorphology and/or FCM-/PCR-MRD or genetics

JCCG, SEHOP-PETHEMA, SFCE, SJCRH, TPOG

Induction failure (EOI)
≥25% blasts in BM

Cytomorphology and/or FCM-/PCR-MRD or genetics

DFCI, UKALL

No induction failure events; TF is an event if CR has not been achieved at later timepoints

Different combinations of methods

AIEOP-BFM, ALLTogether, ALL-IC-BFM, CoALL, COG, DCOG, EsPhALL-COG, NOPHO

BM, bone marrow; CR, complete remission; EOI, end of induction; FCM-MRD, flow cytometric minimal residual disease; PCR-MRD, polymerase chain reaction minimal residual disease.
*Adapted from Buchmann, et al.1

There was a broad consensus among the groups that TF is defined as failure to achieve CR by a pre-specified time point, though the groups do not agree on that time point. Some of the study groups consider failure to achieve CR at end of induction (EOI) as a TF event, while others use a later time point after stratifying the patient to more intensive post-induction therapy, and COG trials define TF as failure to achieve CR at end of consolidation (EOC); these differences are important when trying to compare EFS results from different trials.

Consensus for complete remission and treatment failure

Each study group should define EOI and EOC for their protocols. The consensus was that MRD measurement is the gold standard to assess CR; all available methods for MRD assessment are acceptable if quality standards have been established. CR is defined in Table 3.

Table 3. Definition of CR

Timing

CR is to be assessed no earlier than EOI

Anatomic site requirements

BM: MRD <1% and/or M1 cytomorphology

CNS: CNS1

Testes: Normalization of clinical examination (negative biopsy if clinical examination is not considered normal)

EM: No evidence of leukemic infiltrates as evaluated clinically and by imaging; a pre-existing leukemic mass (mediastinal mass included) must have decreased to at least 1/3 of the initial tumor volume

Methods

BM: MRD measurement; if MRD is not available, response is assessed by cytomorphology

CNS: CSF cytomorphology (FCM or genetic analysis may be used in unclear cases), clinical neurological exam, and imaging (in case of clinical findings)

Non-CNS EM disease: Physical examination, imaging, or histologic examination of tissue biopsy

BM, bone marrow; CNS, central nervous system; CR, complete remission; CSF, cerebrospinal fluid; EM, extramedullary; FCM, flow cytometric; MRD, minimal residual disease.
*Adapted from Buchmann, et al.1

Regarding TF:

  • Failure to achieve CR at a clearly pre-defined time point (such as EOI or EOC) should be considered as a TF event.
  • There was progress toward a consensus that a TF event should be defined no earlier than EOC, allowing patients who do not achieve CR by EOI to potentially achieve CR with consolidation therapy.

Relapse

A patient can only have a relapse event after CR has been previously achieved, and relapse should be subclassified by anatomic sites of involvement at the time of relapse: isolated BM relapse, isolated CNS relapse, isolated testicular relapse, isolated other EM relapse, and combinations of these sites.

Consensus for relapse

The consensus for relapse can be seen in Table 4.

Table 4. Consensus for diagnosis of relapse*

BM relapse (MRD available)

BM #1

BM #2

MRD

Others

 

≥25%

5 to <25%

1 other test§ with 1% blasts

5 to <25%

None

2 tests§ with 1% blasts

1 to <5%

2 other tests§ with 1% blasts

1 to <5%

0 or 1 other test§ with 1% blasts

2 tests§ with 1% blasts

BM relapse (MRD unavailable)

BM #1

BM #2

Cytomorphology

Others

 

M3

M2

1 other test with 1% blasts

M2

None

M2

M1

2 other tests with 1% blasts

CNS relapse

CSF #1

CSF #2

Cytomorphology

Cytomorphology

 

CNS3

CNS2

CNS2

1 other positive test

BM, bone marrow; CNS, central nervous system; CSF, cerebrospinal fluid; FCM, flow cytometric; FISH, fluorescence in situ hybridization; MRD, minimal residual disease; NGS, next-generation sequencing; PCR, polymerase chain reaction.
*Adapted from Buchmann et al.1
Not necessary to define relapse.
Second BM/CSF evaluation at least 1 week later.
§FCM/PCR/NGS-based MRD or FISH/karyotype/PCR demonstrating leukemia-specific marker, or M2/M3 morphology.
FISH/karyotype/PCR demonstrating leukemia-specific marker.
May be defined by cytomorphology, imaging, or biopsy.

Conclusion

While the implementation of these new consensus criteria will require serious effort from most of the study groups, it is of utmost importance that common response and relapse assessment criteria are established in pediatric ALL as current endpoints are poorly defined, creating significant challenges when comparing efficacy between trials and hindering consistency in defining EFS and eligibility criteria for enrollment in relapsed/refractory ALL trials.

  1. Buchmann S, Schrappe M, Baruchel A, et al. Remission, treatment failure, and relapse in pediatric ALL: an international consensus of the Ponte-di-Legno Consortium. 2021. Online ahead of print. DOI: 10.1182/blood.2021012328

Related articles

Newsletter

Subscribe to get the best content related to ALL delivered to your inbox