leptomeningeal metastases: standards of care · 8/8/2016  · • leptomeningeal metastasis (lm)...

32
Leptomeningeal metastases: standards of care Emilie Le Rhun Lille, France

Upload: others

Post on 20-Oct-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

  • Leptomeningealmetastases:

    standards of careEmilie Le Rhun

    Lille, France

  • Disclosures

    ELR has received research grants from Mundipharma and Amgen and honoraria for lectures or advisory board participation from Abbvie, Daiichi Sankyo, Mundipharma and Novartis.

  • Definition of LM• LM is defined as the spread of tumor cells within the leptomeninges and the

    subarachnoid space• LM is synonymous with neoplastic meningitis and can be further denoted by

    primary tumor as leptomeningeal carcinomatosis, gliomatosis or lymphomatosis

    Invasive lobularcarcinomaMGG X40

    Axial braingadolinium enhanced MRI

    Sagittal spinal gadolinium enhanced MRI

    Lumbar puncture

  • Background• Leptomeningeal metastasis (LM) affects up to 10%

    of patients with solid tumors

    • The median survival is limited to 2-3 months, witha 1-year survival rate below 10%

    • Only a few prospective clinical trials are available

    • Treatment strategies include intra-CSF chemotherapy, systemic pharmacotherapy, and focal or large volume radiotherapy and vary widelyacross centers

  • Epidemiology• LM occurs:- in the context of progressive systemic disease in approximately 70%- in around 20% at the time of first progression after initial treatment - in up to 10% at the time of cancer diagnosis

    • In recent large cohorts of LM patients, brain metastases were associated with LM in 33%-54% of breast cancer, 56%-82% of lung cancer, and 87%-96% of melanoma patients

    • Risk factors :- in all patients: opening of the ventricular system during brain metastases

    surgery or resection of cerebellar metastases, especially when using a piece-meal resection

    - in breast cancer patients: lobular subtype and triple-negative tumours - in lung cancer patients: EGFR-mutant and ALK-positive tumors- in melanoma patients: no risk factors identified

  • Levels of evidence in LM

    No standards for:• Neurological examination• Neuro-imaging assessment• CSF diagnosis

    • No trial on systemic treatment• No trial on radiotherapy• Only 5 trials on intra-CSF therapy….

  • Clinical evaluation

    • Typical clinical features include: headache, nausea and vomiting, mental changes, gait difficulties , cranial nerve palsies with diplopia, visual disturbances, hearing loss, sensorimotor deficits of extremities, cauda equina syndrome, radicular, neck and back pain

    Standardized neurological evaluationthat needs validation

    LANO grid, Chamberlain et al., 2016

  • Imaging evaluationSlice thickness 1 mm or less Gadolinium injected 10 min before data acquisition (EANO ESMO) at a dose of 0.1 mmol/kg Standardized and validated gridFollow-up on the same deviceMRI prior to lumbar puncture

    EANO ESMOBrain axial, coronal and sagittal T1 without

    and with contrast enhancement, axial T2, axial and coronal FLAIR

    Spine sagittal T2 and T1 without and with contrast

  • Imaging subtypesType A: LM with typical linear MRI abnormalities

    Type B: LM with nodular disease only as type B

    Type D: LM without MRI abnormalities, except possibly hydrocephalus

    Type C: LM with both linear and nodular disease

  • CSF flow studies

    for patients who are candidates for intra-CSF pharmacotherapy and in whom CSF flow obstruction may be present:• hydrocephalus, • large nodules potentially reducing the CSF

    circulation on MRI, • unexpected toxicity of intra-CSF treatment.

  • CSF analysis

    Specificity >95%Sensitivity: first lumbar puncture: 55%

    second lumbar puncture: 80%In recent large cohorts of LM patients, CSF cytology was considered positive in 66%-90%.

    Invasive Lobular Carcinoma- MGG X40

    Melanoma - MGG X40

  • CSF analysis

    EANO ESMOTechnical aspects - fresh CSF samples processed within 30

    minutes after sampling; alternatively, freshCSF samples fixed with Ethanol-Carbowax

    - CSF volume > 10 ml (at least 5 ml)

    - routine stainings: Papanicolaou and Giemsa; additional immunocytochemical stainings (uponindication and availability of material) - a second CSF sample should be analyzed if the initial CSF sample is negative for diagnosis

    Interpretation of the results

    - positive (presence of tumor cells) - equivocal (suspicious or atypical cells) - negative (absence of tumor cells)

    In presence of tumor cells, the diagnosis is confirmed (type I)In other cases the diagnosis can only be probable or possible (type II)

  • Diagnosis• Typical clinical features include: headache, nausea and vomiting, mental changes,

    gait difficulties , cranial nerve palsies with diplopia, visual disturbances, hearing loss, sensorimotor deficits of extremities, cauda equina syndrome, radicular, neck and back pain

    • MRI features:linear leptomeningeal disease (type A) nodular leptomeningeal disease (type B) both (type C) no neuroimaging evidence of LM except possibly hydrocephalus (type D)

    • CSF cytology: positive, equivocal, negativethe diagnosis can be confirmed by cytology (or histology) (type I) or not (type II)

    Based on these considerations, the likelihood of LM can be assigned “confirmed”, “probable”, “possible” or “no evidence for”

  • Classification of LM:The EANO ESMO Proposal

    Cytology/

    biopsy

    MRI Confirmed Probable* Possible* Lack of

    evidenceType I:positive CSF cytology or biopsy

    IA + Linear + n.a. n.a. n.a.

    IB + Nodular + n.a. n.a. n.a.

    IC + Linear +

    nodular

    + n.a. n.a. n.a.

    ID + Normal + n.a. n.a. n.a.

    Type II:clinical findings and neuroimaging only

    IIA - or

    equivocal

    Linear n.a.** With typical

    clinical signs

    Without typical

    clinical signs

    n.a.

    IIB - or

    equivocal

    Nodular n.a. With typical

    clinical signs

    Without typical

    clinical signs

    n.a.

    IIC - or

    equivocal

    Linear +

    nodular

    n.a. With typical

    clinical signs

    Without typical

    clinical signs

    n.a.

    IID - or

    equivocal

    Normal n.a. n.a. With typical

    clinical signs

    Without typical

    clinical signs

    *requires a history of cancer; **not applicable

  • Prognosis

    Median OSwithout tumour-specific treatment 6-8 weeks Breast cancer with treatment 1.75-4.5 months Lung cancer with treatment 3-6 months Melanoma with treatment 1.7-2.5 months

    Chamberlain 2009, Groves 2010, Grossman 1982, De Angelis 1998 , Gauthier 2010, Rudnicka 2007, Clatot 2009, De Azevedo 2011, Le Rhun 2013, Harstad 2008, Papadopoulos 2002, Geukes Foppen 2016, Grossman 1999 , Hammerer 2005, Morris 2011, Park 2011, Gwak2013, Lee 2013, Kuiper 2015

  • Prognostic factors• performance status at diagnosis of LM • primary tumour type• cerebrospinal fluid (CSF) protein levels• administration of combined modality treatment, systemic

    treatment, or intra-CSF treatment• initial clinical or CSF responses to treatment

    (Rudnicka 2007, Gauthier 2010, Lee 2011, de Azevedo 2011, Niwinska 2013, Yust Katz 2013, Le Rhun 2013, Clarke 2010, Oechsle 2010, Herrlinger 2004, Clatot 2009, Hyun 2016)

    An association of WBRT with overall survival has not been consistently reported.

    (Morris 2012, Park 2012, Gwak 2013, Abouharb 2014)

  • 224 participants between April 7 and August 8, 2016

  • Randomized trials assessing the response to intra-CSF treatment in LM

    Trial Design Population Primary endpoint Efficacy Safety

    Grossman

    1993

    IT MTX versus IT

    thiotepa

    Solid tumors (n=40),

    CUPS (n=1) and

    lymphomas (n=10)

    Neurological response

    rate

    IT MTX vs. IT thiotepa

    Neurological response rate: none

    Neurological stabilization: 32% vs. 12.5%

    Survival: 15.9 weeks vs. 14.1 weeks

    IT MTX vs. IT thiotepa

    Serious toxicities similar in both group

    Mucositis (p=0.04) and neurological complications (p=0.008)

    more frequent in MTX armHitchins 1997 IT MTX versus IT MTX

    + cytarabine

    Solid tumors (n= 30),

    cancers of unknown

    primaries (n=7) and

    lymphomas (n=7)

    Response rate IT MTX vs. MTX + cytarabine

    Response rate : 61 vs. 45% (p

  • Neuroimaging was not used to evaluate the neurologicalresponse; trial stopped after 35 patients enrolled between1991 and 1998 (instead of 50 initially expected)

    Median survival: 18.3 (SE 6.7) weeks in the intra-CSF arm vs. 30.3 (SE 10.9) weeksin the control arm

    Breast cancer patients; diagnosis of LM based on CSF cytology or MRI, no progressive or untreated brainmetastases

    Intraventricular MTX, appropriate systemic therapy, and if necessary RT to clinically relevant sites

    Appropriate systemic therapy, and if necessary RT to clinically relevant sites – No intraCSF therapy

  • Deposein trial (NCT01645839)

    Main inclusion criteria• Adult breast cancer patients requiring a systemic treatment at inclusion, ECOG PS: 0-2• Diagnosed with LM (CSF positive cytology; combination of typical clinical and MRI findings)• Meningeal metastases 0.5 if treated by SRS/SRT)• Asymptomatic brain metastases permitted• WBRT not allowed• Untreated CSF blockade not allowed

    Main objective

    • To compare the neurological progression free survival (clinical and imaging criteria) between the 2 groups

    Liposomal cytarabine until unacceptable toxicity, neurological progression or a max of 12 monthsSystemic treatment at the discretion of the investigator

    Group ABC patients with LMand inclusion criteria

    Systemic treatment alone

    Group B Systemic treatment+ liposomal cytarabine

    R

  • Intra-CSF therapyLimits of intra-CSF therapy In favor of intra-CSF therapyNo randomized trial has demonstrated that intra-CSF therapy prolongs survival in LM patients

    Used by a large majority of physicians in addition to systemic treatments across Europe (only 11% ofphysicians never use intra-CSF therapy)

    Recent prospective safety data have shown a good tolerance of liposomal cytarabine

    The compounds routinely used for intra-CSF treatment do not have a key role as single agents for systemic treatment of common cancers causing LM

    Compounds with systemic efficacy are currentlyunder evaluation as intra-CSF agents in clinical trials

    Intra-CSF therapy has only a limited penetration (1-3 mm) into solid tumor lesions

    Intra-CSF therapy may be inefficient and toxic in case of CSF flow blocks

    Rationale for the treatment of floating tumor cells in the CSF in the setting of little or no blood CSF barrierdysfunction

    Rationale for the treatment of linear diffuse or ependymal spread not yet accompanied by bloodbrain barrier dysfunction

  • Is there a role for intra-CSF treatment?in selected patients:

    In the presence of: - floating tumor cells in the CSF in the setting of little or

    no blood CSF barrier dysfunction- linear diffuse or ependymal spread not yet

    accompanied by blood brain barrier dysfunction

    Not as first option in patients with symptomatichydrocephalus who require ventriculoperitoneal shunt placement or with a ventricular device without on/off option

  • Route of administration of intra-CSF therapy

  • • Advantages of the ventricular route:

    - certainty that the drug is not delivered into the epidural or subdural space

    - more uniform distribution of the agent

    - more patient comfort, faster procedure, which improves compliance and safety of drug administration

    • Safety of ventricular devices shown in several cohorts of patients using different technologies and several devices (

  • Systemic pharmacotherapy

    • a priori no reason to believe that systemic pharmacotherapy for contrast-enhancing manifestations of LM should be less efficient than for other systemic manifestations of cancer

    • increased CSF protein levels in most LM patients confirm that the blood-CSF barrier is commonly disrupted in LM

    • no specific prospective trials reported on systemic treatment of LM, but retrospective series suggest some activity of systemic chemotherapy

    • the best systemic treatment for LM is determined by:- the primary tumour, - its molecular characteristics or the molecular characteristics of tumour cells

    of the CSF when available - and prior treatment of the underlying malignancy.

  • Radiotherapy• No randomized clinical trial to assess the efficacy and tolerance of

    RT in LM has been conducted.

    • Focal RT (involved-field or stereotactic RT or radiosurgery) can be used to treat nodular disease and symptomatic cerebral or spinal sites.

    • WBRT may be considered for extensive nodular or symptomatic linear LM or co-existing brain metastases.

    • Cerebrospinal RT is rarely an option for adult patients with LM from solid cancers because of risk of bone marrow toxicity, enteritis and mucositis, and the usual co-existence of systemic disease.

  • Supportive care• When required clinically, the lowest dose of steroids

    should be used for the shortest time possible.

    • Seizures should be managed using drugs that do not interact with systemic treatments. Primary prophylaxis is not recommended.

    • Ventriculoperitoneal shunting may provide durable relief from symptomatic hydrocephalus

  • Life expectancy < 1 month

    Palliative approach

    CSF cytology positive

    Type I LMpositive CSF or biopsy

    Type II LMclinical findings and neuroimaging only

    Life expectancy ≥ 1 month

    Active BMNo active BM

    CSF cytology negative(LM confirmed by biopsy)

    Type IIA• IT therapy (+)• Modification of

    systemictherapy or WBRT +

    Type IIB• IT therapy -• Modification of

    systemictherapy (+)

    • Focal RT +

    Type IIC• IT therapy (+)• Modification of

    systemic therapy +

    • WBRT and/or Focal RT +

    Type IIA• IT therapy (+)• Modification of

    systemictherapy +

    • WBRT (+)

    Type IIB• IT therapy -• Modification of

    systemictherapy +

    • Focal RT +

    Type IIC• IT therapy (+)• Modification of

    systemictherapy +

    • WBRT and/or Focal RT +

    Type IIA • IT therapy (+)• Modification of

    systemictherapy or WBRT or both +

    Type IIB• IT therapy -• Modification of

    systemictherapy +

    • Focal RT +

    Type IIC• IT therapy (+)• Modification of

    systemictherapy +

    • WBRT and/or Focal RT +

    Type IIA • IT therapy (+)• Modification of

    systemictherapy +

    • WBRT (+)

    Type IIB• IT therapy -• Modification of

    systemictherapy +

    • Focal RT (+)

    Type IIC• IT therapy (+)• Modification of

    systemictherapy +

    • WBRT and/or Focal RT (+)

    No active BM

    Stable ECD

    Progressive ECD

    Type IA• IT therapy +• Modification of

    systemictherapy (+)

    • WBRT (+)

    Type IB• IT therapy +• Modification of

    systemictherapy (+)

    • Focal RT +

    Type IC• IT therapy +• Modification of

    systemictherapy (+)

    • Focal RT +, WBRT (+)

    Type ID• IT therapy +• Modification of

    systemictherapy (+)

    • RT-

    Type IA• IT therapy +• Modification of

    systemictherapy +

    • WBRT (+)

    Type IB• IT therapy +• Modification of

    systemictherapy +

    • Focal RT (+)

    Type IC• IT therapy +• Modification of

    systemictherapy +

    • WBRT or SRT (+)

    Type ID• IT therapy +• Modification of

    systemictherapy +

    • RT -

    Type IA• IT therapy +• Modification of

    systemictherapy or WBRT or both +

    Type IB• IT therapy +• Modification of

    systemictherapy +

    • Focal RT +

    Type IC• IT therapy +• Modification of

    systemictherapy +

    • WBRT and/or Focal RT +

    Type ID• IT therapy +• WBRT and/or

    modification of systemictherapy +

    Type IA• IT therapy +• Modification of

    systemictherapy +

    • WBRT (+)

    Type IB• IT therapy +• Modification of

    systemictherapy +

    • Focal RT (+)

    Type IC• IT therapy +• Modification of

    systemictherapy +

    • WBRT and/or Focal RT (+)

    Type ID• IT therapy +• Modification of

    systemictherapy +

    • WBRT (+)

    Active BM

    Stable ECD

    Progressive ECD

    Stable ECD

    Stable ECD

    Progressive ECD

    Progressive ECD

  • Follow-up of LM

    EANO ESMO Neurological assessment: standardized gridCerebrospinal MRI: LANO gridCSF cytology: positive, negative, equivocal

    Evaluation every 2-3 months

  • Response Assessment

    EANO ESMOResponse Clinically improved or stable

    Neuroimaging improved

    CSF cytology improved or stableStable Clinically stable

    Neuroimaging stable

    CSF cytology stableSuspicion of

    progression

    Clinically worse, neuroimaging stable, CSF cytology stable

    or

    Clinically stable or worse, neuroimaging stable, CSF cytology worse

    (increased tumour cell counts)Progression Neuroimaging worse

    or

    de novo positive CSF cytology

  • Conclusions

    • Different subtypes of LM should be considered according to the results of cytology and cerebrospinal MRI

    • Management strategies for individual patients should be derived by integrating:

    - general and neurological health status- CSF findings - neuroimaging findings - absence or presence of solid brain and systemic metastases - histology and molecular status of the primary tumor- previous treatments

    • A standardized follow-up should be considered

    Leptomeningeal metastases:�standards of careDisclosuresDefinition of LMBackgroundEpidemiologyLevels of evidence in LMSlide Number 7Clinical evaluationImaging evaluationImaging subtypesCSF flow studiesCSF analysisCSF analysisDiagnosisClassification of LM:�The EANO ESMO ProposalPrognosisPrognostic factorsSlide Number 18Randomized trials assessing the response to intra-CSF treatment in LM Slide Number 20Deposein trial (NCT01645839) Intra-CSF therapyIs there a role for intra-CSF treatment?Route of administration of intra-CSF therapySlide Number 25Systemic pharmacotherapyRadiotherapySupportive careSlide Number 29Follow-up of LMSlide Number 31Conclusions