137
Mastocytosis Olivier Hermine MD, PhD Hematology Department, Necker Hospital Centre National de référence des mastocytoses, Necker Hospital CNRS UMR 81 47 Institut Imagine, Necker Hospital Paris, France

Aucun titre de diapositive - OVHaihemato.cluster013.ovh.net/AIH/documents/Cours DES/DES 2017-0… · Marshall, Nature Rev Immunol 420: 787- (2004) Cancer Allergy Autoimmunité Alloreactivity

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

  • Mastocytosis

    Olivier Hermine MD, PhD

    Hematology Department, Necker Hospital

    Centre National de référence des mastocytoses, Necker Hospital

    CNRS UMR 81 47

    Institut Imagine, Necker Hospital

    Paris, France

  • Mast cells in hypersensitivity type 1

  • •Mast cells are tissue cells

    •They are prevalent in areas

    • which interface directly with

    • the external environment

    •close to blood vessels and

    •nerve endings

    •They can respond very

    •rapidy to a stimulus

    •with the production of a

    •whole array of mediators

    •Mast cells are sentinentals with strategic location

    •from JS. Marshall, Nature Rev Immunol 420: 787- (2004)

  • Cancer Allergy

    Autoimmunité

    Alloreactivity

    Acute

    chronic

    Mastocytosis

    Inflammation

    fibrosis Neurological

    And

    Psychiatric

    Diseases

  • •Mastocytosis

    •Mast cells and Diseases

  • Definition - Mast cell accumulation in various organs (Skin, GI tract, Liver, Bone and Bone Marrow, etc) -Myeloproliferative disorder; Aggressive vs indolent disease

    - Association with hematological disorders - Clinical heterogeneity (Infiltration vs Mediators release)

    Photos Pr Bodemer et Dr Barete

    MASTOCYTOSIS

    Children

  • •MCAS

  • Updated WHO Classification of Mastocytosis 2016

    Cutaneous mastocytosis (CM)

    - Maculopapular CM (MPCM) = urticaria pigmentosa (UP)

    - Diffuse CM (DCM)

    - Mastocytoma of skin

    Systemic mastocytosis (SM)

    - Indolent SM (ISM)

    - Smoldering SM (SSM)

    - SM with associated hematologic neoplasm (AHN)*

    - Aggressive SM (ASM)

    - Mast cell leukemia (MCL)

    Mast cell sarcoma

    -------------------------------------------------------------------------------------

    *The previous term SM-AHNMD (SM with clonal hematologic

    non-mast cell-lineage disease) and the new term AHN can be

    used synonymously.

  • Physiopathology

  • MC TC MC T

    Stem Cell CD34+ Bone Marrow

    Circulation SCF

    IL-10

    IL-6

    SCF

    CD34+

    c-kit high

    FceRI neg

    CD13+

    MC C

    IL-4 SCF

    ?

    Simplified pathways of human MC differentiation

    Tissues

    IL-4 Survival : SCF

    NGF

    IL-4

    IFN-g

    …...

  • 0

    100

    200

    Day0 SCF- SCF+

    0

    500

    1000

    Day0 SCF- SCF+

    0

    200

    400

    Day0 SCF- SCF+

    •Day 30

    •Day 30

    •Day 30

    •CD34-Kit+ •CD34+Kit+

    •Healthy

    •Donor

    •(SCF+)

    •Mastocytosis

    •c-Kit D816V

    •Mastocytosis

    •c-Kit WT

    •Histamine level

    •ng/

    ml

    •ng/

    ml

    •ng/

    ml

    •CD34-Kit+

    •CD34+Kit+

    Sophie Georgin et al, Blood 2010

    C-kit+CD34-Mast cell precursors in peripheral blood

  • •C-kit activation pathway

    •Lyn

  • •Ig1 •Ig2 •Ig3 •Ig4 •Ig5 •TM •JM •TK1 •TK2

    •1 •2 •3 •4 •5 •6 •7 •8 •9 •10 •11 •12 •13 •14 •15 •16 •17 •18 •19 •20 •21

    •Dog mast cell tumor D826-828- InsDT •Q430R

    •ins421 aa45

    •S479I4

    •N508I

    •Del/Ins555-559(5)

    •ITD571-590(15) •n=88

    •Patrice Dubreuil, Katia Hanssen, Sébastien Leutard, Kevun Hahn, Christine Bodemer, Olivier Hermine CEREMAST

    •D479N

    •V560G D418

    D564-576

    •E860G D419 (12) •InsFF419

    •C443Y •S476I

    •ITD501-502(2)

    •ITD505-508

    •D572A •D816Y(4)

    •K509I(6)

    •D816I

    •ITD502-503(4)

    •D816V(353)

    •Ig1 •Ig2 •Ig3 •Ig4 •Ig5 •TM •JM •TK1 •TK2

    •1 •2 •3 •4 •5 •6 •7 •8 •9 •10 •11 •12 •13 •14 •15 •16 •17 •18 •19 •20 •21

    D417-418 D419Y

    •n=603 •Human mastocytosis

    •MCL leukemia

    •And Sarcoma

    •No PTD

    •Exons 8 to 11 •Exon 17 •WT

    •30% of the dogs are mutated

    •No PTD

    Adult (Indolent and agressive)

    •PTD

    Pediatric

    •No PTD

  • Single Gene (C-kit) several diseases

  • •Differential colony-forming ability of ECD and D816X mutants expressing Rat2 cells

    •Without

    •SCF

    •With

    •SCF

  • •Outcome and mutations of c-kit

    Patients mastocytosis

    Statut of c-Kit

    WT Mutation autre que

    D816V D816V

    Children (N= 59) 15% 46% 39%

    Adults (N= 485) 28% 3% 69%

    •age

    •Mutation %

    •persistent

    •Mutations

    •ex8 & ex9

    •puberty

    •Mutations

    •D816V

    •regressive

    •50

    •75

    •25

  • Children Mastocytosis

    Adult Mastocytosis

    Kit Mutations

    No senescence Chronic disease

    AGRESSIVES Fatal Outcome

    Non PTD PTD

    Senescence Régression

    Conclusions and Perspectives

    P-p38 P-p38

    p53 ATRX ?

    APBs

    Telomeres

    protection

    By TRF2

    No protection

    of Telomere

    By TRF2 Germline mutations

    (p53; p16) ?

  • •Role of Telomere Maintenance

    •Mutation

    •D816V

    •(Exon 17)

    •C-Kit

    • Mastocyte Survival

    •=> immortalisation

    •Indolent Mastocytosis •Télomere maintenance

    •Mutation

    •Exons 8 à 11

    •C-Kit et WT •Proliferation

    •Senescence

    •Short telomeres

    •SCF

    •Spontaneous Regression

    •Tumor progression - Genetic instability - ALT

  • •Role of Telomere Maintenance

    •Mutation

    •D816V

    •(Exon 17)

    •C-Kit

    • Mastocyte Survival

    •=> immortalisation

    •Indolent Mastocytosis •Télomere maintenance

    •Mutation

    •Exons 8 à 11

    •C-Kit et WT •Proliferation

    •Senescence

    •Short telomeres

    •SCF

    •Spontaneous Regression

    •Tumor progression - Genetic instability - ALT

    •Agressive Mastocytosis •Télomere maintenance

  • •Cytosine

    •5-me-Cytosine •5-hme-Cytosine

    •Dnmt

    •Tet2

    •Inhibitors

    •(e.g. RG108)

    Tet2 is involved in the conversion of 5 meC to 5hmeC at specific genomic loci

    •Mutations in SMP and AML

    •CMML++

    •CMML is associated with MS

  • Tet2 mutations are frequent and are correlated to aggressive disease

    •Mastocytosis Patient Cohort:

  • •Bone marrow derived Tet2-deficient mast cells infected with c-KitD816V have a competitive growth advantage

    Confirmed in two independent Tet2-KO models (lacz and flox)

    Consistent whether infections are done using « young » or « old » MC cultures (old > 80 days culture with IL3)

    •CD

    11

    7

    •Tet2(+/+)

    •GFP

    •Tet2(+/-) •Tet2(-/-)

    •cKitD816V+ •cKitD816V+ •cKitD816V+

  • •SRSF2-P95 Hotspot Mutation is Highly Associated with Aggressive Forms of Mastocytosis and Mutations in Epigenetic Regulator Genes •Katia Hanssens, Fabienne Brenet, Julie Agopian, Sophie Georgin-Lavialle, Gandhi Damaj, Laure Cabaret, Maria Olivia Chandesris, Paulo de Sepulveda, Olivier Hermine, Patrice Dubreuil *§ and Erinn Soucie* (Haematologica; in press)

  • •ASSOCIATED HEMATOLOGICAL MALIGNANCIES

    Travis 1988

    Horny 2004

    Sotlar 2010

    Pardanani 2009

    Damaj et al 2013

    AHNMD; n (%) 20 (33) 22 (33) 48 134 (40%) 62

    Myeloid % 82 90 83 89 82

    MDS % 32 9 8 3 28

    CMML % 39 27 23 16

    MPN % 9 21 45 16

    AL % 10 21 3 5

    Lymphoid % 11 18

    Lymphoma% 6 5 8

    MM% N=2 10 4 5 (MGUS)

    CLL% 2

    •*Damaj et al, CEREMAST, Unpublished data

  • Sotlar J Pathol

    2010

    Horny JCP,

    2004

    Pardanani Blood 2009

    Tefferi Leuk,emia

    2009

    Traina Plosone

    2012

    *Damaj 2013

    N 48 20 134 23 8 62

    D816; n(%) 45 (94) 16 (80) 50 (63) 44 (86)

    JAK-2; n(%) na na 6 (8) 2 (7.5)

    TET-2; n(%) na na na 8 (35) 5 (62) 12 (32)

    ASXL1; n (%) na na na 2(25) 6 (17)

    FGFR4; n(%) na na na 7 (18)

    CBL; n(%) (12.5)

    GENE MUTATIONS IN SM-AHNMD (1)

    TET2, ASXL1 are positive, only in the myeloid AHNMD

    •*Damaj et al, CEREMAST, Unpublished data

  • •Figure 1

    •P = 0.003 •ASXL1 negative. median OS: 92.33 (95% CI, not reached)

    •AXSL1 positive: median OS: 12.86 (95% CI, 5.56 – 20.17)

    Gandhi Damaj1,2,3, Magalie Joris1, Olivia Chandesris2,4, Katia Hanssens5, Erinn Soucie5, Danielle Canioni6, Brigitte Kolb7, Isabelle Durieu8, Emanuel Gyan9, Cristina Livideanu10, Stephane Chèze11, Momar Diouf12, Reda Garidi13, Sophie Georgin-Lavialle14, Vahid Asnafi15, Ludovic Lhermitte15, Christian Lavigne16, David Launay17, Michel Arock18,19, Olivier Lortholary20, Patrice Dubreuil5* and Olivier Hermine2,3,4*

  • Mastocytosis Heterogeneity and c-kit

    •MDS

    •CMML

    •AML

    •Sarcoma

    •MCL

    •Indolent

    Masotcytosis

    •Agressive

    mastocytosis

    •C-kit 816 V

    •Tet2

    •ASXL1

    •JAK2,

    • FIP1L1-PDGFR,

    •CMML

    •Regression

    •C-kit 816 V

    •negative

    •C-kit 816 V

    •Other ?

    •C-kit 816 V

    •Pediatric forms

    •Other ckit mutations

    •WT c-kit

    •Adult Forms

    •Sarcoma

    •MCL

    •Other ckit mutations

    •WT c-kit

    •Others SMP

    •other ?

    •NHL

    •Agressive

    •Masotcytosis

    (rare)

    •Cytokines (CD40L)

    •Tet2

    •ASXL1

    •C-kit 816 V

    •Tet2

    •ASXL1

    •others

    •Tet2

  • Diagnosis

  • Urticaria pigmentosa

  • Bone involvement

  • Osteosclerosis

  • Mastocytosis Diagnosis

    1. DARIER ’S SIGN FOR CUTANEOUS MASTOCYTOSIS (Skin involvement is not required)

    2. HISTOLOGY FOR CUTANEOUS AND/OR SYSTEMIC

    MASTOCYTOSIS (required)

    - TOLUIDINE BLUE

    - ANTI -TRYPTASE STAINING

    - CD117+, CD2+ and/or CD25+, CD15-

    3. MAST CELL MEDIATORS

    - Total tryptase >20ng/ml

    - Soluble C-kit level

  • Major and Minor WHO Criteria of Systemic Mastocytosis (SM criteria)*

    Major criterion:

    Multifocal dense infiltrates of mast cells

    (>15 mast cells in aggregates) in bone marrow biopsies and/or in sections of other extracutaneous organ(s)

    Minor criteria:

    a. >25% of all mast cells are atypical cells

    (type I or type II) on bone marrow smears or are spindle-shaped in mast cell infiltrates detected on sections of visceral organs

    b. KIT point mutation at codon 816 in the bone marrow or another extracutaneous organ

    c. mast cells in bone marrow or blood or another extracutaneous organ expresses CD2 or/and CD25

    d. Baseline serum tryptase concentration >20 ng/ml (in case of an unrelated myeloid neoplasm is not valid as an SM criterion)

    If at least one major and one minor or three minor criteria

    are fulfilled

    the diagnosis is systemic mastocytosis = SM

    *SM criteria have been defined by the WHO in 2001 and confirmed in 2008 and 2016.

  • Treatment

  • Prognosis

    •Blood 2009

  • Prognosis

    •Blood 2009

    •Indolent Mastocytosis >80%

    •Agressive Mastocytosis

  • Therapeutic decision

    • Agressive disease : Reduction of life expectancy

    and organ failure

    • Indolent disease : No life expectancy reduction,

    no organ failure , Handicap associated with

    symptoms (patient vs physician)

  • Protocols for treatment of Indolent

    Mastocytosis

    Should we treat these patients ?

    On what basis ?

    What should be the end points of treatment ?

  • Identification of all systemic manifestations in patients suffering from

    mastocytosis

    • From 2004, 363 mastocytosis patients and 90 controls in France were asked to rate their overall disability (OPA score) and the severity of 38 individual symptoms.

    • A specific questionnaire (AFIRMM V1), encompassing these 38 symptoms, has been created and validated.

    PLoS ONE. 2008 May 28;3(5):e2266

  • Identification of all systemic manifestations in patients suffering from

    mastocytosis

  • Course of Tumoral mass (3 months)

  • •Patient ISM

    •53 years old

    •Cognitive impairment

    •IRM/ASL: artérial spin labelling, Flow blood in the brain

    •Perfusion MRI

    •Control

  • •Hyperperfusion of central grey nuclei : 11 patients with cognitive imapirement vs 33 controls

    •p

  • •Hypoperfusion of the anterior Cingulum antérieur in 10 patients with depression and mastocytosis Vs. 18 patients with Mastocytosis but not depressed

    •p

  • •IDO

    •Hypothesis

    •TNFa

    •Tryptophane

    •Kynurenine

    •Acid

    •xanthurenic

    •Acid

    •quinolenic

    •Neurotoxicity

    •Oxidative stress

    •apoptosis

    •-

    •+

    •Cognition (?)

    •serotonin

    •Low serotonin

    •Depression ?

    •Proteases

    •Histamine

    Others

    •ASL

    •abnormalities

  • Fig 2

  • Eviction of mast cells stimulants: depend on the patient history

    Aim at inhibiting mediator release by mast cell or mediators effects.

    - Anti-H1 : pruritus, flush and sometimes GI pains.

    - Anti-H2 : essentially GI pains.

    - Aspirin : for flushing, tachycardia, but may cause vascular collapse!!!

    - Corticoids : for local treatment of cutaneous lesions, ascite, malabsorption, GI cramps

    (budesonide: corticoïde à délitement entéral)

    - Cromoglycate disodium : non specific mediator release symptoms

    - Anti-leucotriènes (montelukast-singulair): for respiratory manifestations

    - Epinephrin : Hypotension

    - Biphosphonates : bone pain and bone loss

    SYMPTOMATIC THERAPIES OF MASTOCYTOSIS

  • Cytoreductive treatment

    • Interferon alpha

    • Cladribine

    • Kinase inhibitors

    • Thalidomide

    • Rapamycine

  • Cytoreductive Treatments

    • High dose steroids

    • Alpha Interferon (?)

    • Cladribine

    • Kinases Inhibitors

    • Thalidomide

    • mTOR inhibitors (Rapamycine, Temsirolimus)

    • Bone marrow transplantation

    • Treatment of AHNMD

  • Cladribine Treatment • 0.14 mg/kg 5 days a week in 2 hour infusion or

    subcutaneously

    • 1 to 6 cycles repeated every 4 to 12 weeks

    • Usually no premedication with corticosteroids or

    antihistaminic

    • Antibiotic prophylaxis with Co-trimoxazole and Valaciclovir

  • •Clinical response

    •Clinical improvement for

    • 10/11 mediator release

    symptoms including

    anaphylaxis

    • 5/6 mast cell infiltration-related

    symptoms including urticaria

    pigmentosa, and organomegaly

    (P

  • •38 patients (80%) comprising 11 CR, 17 MR and 10 PR.

  • •Progression free survival

    •Median PFS = 3.7 (0.1-8.6)

  • •Overall survival

    •Whole group

    •Median OS = 8.2 (0.1-9.1) years

    •According to WHO sub-types

  • Discussion on Cladribine

    2-CdA is active in mastocytosis

    Its activity seems to be more pronounced on fatigue, flush, GI

    symptoms and pruritus, skin infiltration

    Median duration of response remains to be precisely determined but

    some patients have long term responses

    Haematological toxicities and infections were manageable and were

    more frequent in the aggressive forms of the disease

    Further work is needed to determine optimal therapy schedule and

    usefulness of maintenance therapy to prevent relapse

  • 15e Colloque de la Recherche de la Ligue contre le cancer, Nice, 24-25 janvier 2013

    Oncogenic Mutations of c-kit in Mastocytosis

    1

    2

    Oncogenic Signal

    Adult Mutations 85%

    Clonal disease +++

    No regression

    Pediatric Mutations 75%

    Clonal disease +++

    Regression Sarcoma

    Human

    Dogs

    Exons 8 to 11 Exon 17 WT

  • Effect of STI 571on proliferation of cell lines

    with juxtamembrane or V814 c-kit mutations

    0

    20

    40

    60

    80

    100

    120

    0 0.1 1 10

    µM

    % C

    PM

    Ba/F3 Kit+IL3

    Ba/F3 Kit+SCF

    ∂27

    FMA 3

    KIT G559

    IC2 V814

    P815 (V814)

  • c-kit Mutations in Mastocytosis

    NH2

    COOH

    c c

    c c

    c c

    c c

    F522C

    Akin et al. Blood. 103:3222, 2004

    •Location: Exon 10 (Transmembrane)

    •Type: Germline

    •Clinical features:

    Childhood onset CM

    SSM in early 20s

    Well-differentiated phenotype

    •Functional studies: Gain-of-function

    •Inhibition by imatinib: Yes (in vitro and clinically)

  • Therapy with imatinib in a patient with transmembrane c-kit

    mutation: Proof of concept

    0 25 50 75 100 1250

    50

    100

    150

    200

    Days

    Try

    pta

    se (

    ng

    /ml) 100 mg/d

    200 mg/d

    300 mg/d

    400 mg/d

    Akin, Fumo, Yavuz, Lipsky, Neckers, Metcalfe. Blood, 103:3222, 2004

  • Signaling of PI-3k/Akt/mTOR •C-KIT

    •- •PI •3 kinase

    •AKT

    •- •m •TOR

    •1 •Cyclin D •CDK’s

    •G •1 •S

    •Cell division

    •G •0 •(resting cell)

    •+ •P-p70S6k

    •Translation/apoptosis

    •4EBP/eIF4E

    •RAPAMYCINE

  • •BMMC

    •c-kit wt •BMMC

    •c-kit 816

    •La mutation D816V du c-kit confère à la cellule BMMC la sensibilité à la Rapamycine

    •Lignée BMMC •cellules mastocytaires dérivées de moelle osseuse de souris

    •transgéniques pour le c-kit humain

  • Rapamycin and MS

    •Stop IFN

    •CDA 1

    •CDA 2

    •Start IFN

    •Start Rapamycin

  • PKC412 (N-benzyl staurosporine)

    • Inhibits PKC, VEGFR,

    Kit, PDGFR, flt3

    • Phase I/II trials in

    hematologic and solid

    tumors

    – Nausea, vomiting,

    diarrhea, fatigue

    • Inhibits D816V c-kit

    •Gotlib et al. Blood, in press, 2005

    •BAF3 D816V cells

  • Baseline Patient and Disease Characteristics.

    Gotlib J et al. N Engl J Med 2016;374:2530-2541

  • Best Overall Response to Midostaurin in the Primary Efficacy Population.

    Gotlib J et al. N Engl J Med 2016;374:2530-2541

  • Overall Survival

    •Median duration of follow-upa: 27 months (range: 11-38)

    Kaplan-Meier Estimate for Overall Survival Median

    ASM Not reached

    MCL 22.6 months

    •a Time from treatment start to data cut-off.

    Deaths, n

    12

    9

    3

  • •Prospective survey of PKC412 compassionate use for adult patients suffering from AdSM in France. Survival

    curves in PKC412 treated and control groups at the last follow-up in April 2015.

  • Baseline TMSAS

    Best TMSAS value

    on treatment

    Lack of energy Feeling drowsy

    Diarrhea

    Feeling bloated

    Difficulty concentrating

    Difficulty sleeping

    Cough

    Dry mouth

    Pain

    Weight loss

    Worrying

    Shortness of breath

    Feeling nervous

    Sweats Dizziness

    Feeling irritable

    Feeling sad

    Itching Lack of

    appetite Nausea

    Change in way food tastes

    Swelling of

    arms/legs

    Skin changes

    Problems with sex

    Vomiting

    Numbness in hands/feet

    Urinary problems

    Don’t look like myself

    Difficulty swallowing

    Mouth sores

    Constipation

    Hair loss

    Decreased Frequency of MSAS Symptoms

    (n = 79)

  • TITLE: HEMATOPOIETIC STEM CELL TRANSPLANTATION FOR

    ADVANCED SYSTEMIC MASTOCYTOSIS

    Authors: Celalettin Ustun1, Andreas Reiter2, Bart L Scott3, Ryotaro Nakamura4,

    Gandhi Damaj5, Sebestian Kreil2, Ryan Shanley6, William J. Hogan7, Miguel-

    Angel Perales8, Tsiporah Shore9, Herrad Baurmann10, Robert Stuart11, Bernd

    Gruhn12, Michael Doubek13, Jack W. Hsu14, Eleni Tholouli15, Tanja Gromke16,

    Lucy A. Godley17, Livio Pagano18, Andrew Gilman19, Eva Maria Wagner20, Tor

    Shwayder21, Martin Bornhäuser22, Esperanza B. Papadopoulos8, Alexandra

    Böhm23, Gregory Vercelotti1, Maria Teresa Van Lint24, Christoph Schmid25,

    Werner Rabitsch26, Vinod Pullarkat27, Faezeh Legrand28, Ibrahim Yakoub-

    agha29, Wael Saber30, John Barrett31, Olivier Hermine32, Hans Hagglund33,

    Wolfgang R. Sperr34, Uday Popat35, Edwin P.Alyea36, Steven Devine37, H.

    Joachim Deeg3, Daniel Weisdorf1, Cem Akin38, Peter Valent34

  • ITK for indolent diseases

    • New ITK

    • Inhibiton of Mast cell activation

    • Cytoreductive on Mast cells

    • Not cytoreductive on other cells

    • Not toxic (short term and long term)

    – Genotoxic, carcinogenic

    – Cardiotoxic (Abl++, Src, VEGF, Herg chanel, etc)

  • 0,0

    0,2

    0,4

    0,6

    0,8

    1,0

    1,2

    1,4

    1010,10,050,01

    DO

    49

    0 n

    m

    AB 1010 (µM)

    Inhibition of c-Kit phosphorylation

    •Masitinib formula: C28H30N6OS

    •Molecular weight:

    •AB1010 (mesylate salt): 594.76

    •AB1003 (free base): 498.66

    •Masitinib® belongs to the ATP binding pocket molecules kinase inhibitor 2-amino-4-aryl-thiazole family

    •c-Kit •c-Kit

    •Proliferation

    •AB1010

    •Proliferation

    •AB1010

    •Anti-c-Kit

    •Anti-Phospho Kit Tyr 729

    •Anti-Phospho Tyr 4G10

    M

    e

    N

    S

    N

    H

    N

    H

    O

    N

    N

    N

    M

    e

  • 0,0

    0,2

    0,4

    0,6

    0,8

    1,0

    1,2

    1,4

    1010,10,050,01

    DO

    49

    0 n

    m

    AB 1010 (µM)

    Inhibition of c-Kit phosphorylation

    •Masitinib formula: C28H30N6OS

    •Molecular weight:

    •AB1010 (mesylate salt): 594.76

    •AB1003 (free base): 498.66

    •Masivet® belongs to the ATP binding pocket molecules kinase inhibitor 2-amino-4-aryl-thiazole family

    •c-Kit •c-Kit

    •Proliferation

    •AB1010

    •Proliferation

    •AB1010

    •Anti-c-Kit

    •Anti-Phospho Kit Tyr 729

    •Anti-Phospho Tyr 4G10

    M

    e

    N

    S

    N

    H

    N

    H

    O

    N

    N

    N

    M

    e

  • •Masitinib

    •Masitinib selectivity

    •Data from Ambit analysis

    •(http://www.kinomescan.com/TREEspot/TREEspot.aspx)

    •Data from AB Science)

    •122

  • Masitinib demonstrated efficacy in improving handicaps associated with

    mastocytosis

    •Trend for a better response in patients started with the

    •highest dose of 6 mg/kg/day

    0

    5

    10

    Wee

    k 0

    Wee

    k 12

    Wee

    k 24

    Week 0

    Week 12

    Week 24

    0

    1,5

    3

    Wee

    k 0

    Wee

    k 12

    Wee

    k 24

    0

    4

    8

    12

    16

    Wee

    k 0

    Wee

    k 12

    Wee

    k 24

    0

    7

    14

    Wee

    k 0

    Wee

    k 12

    Wee

    k 24

    patients with pruritus score ≥6 (n=14)

    patients with at least 1 flush/day (n=16)

    patients with Hamilton score ≥10 (n=13)

    patients with pollakiuria: miction ≥8/Jour (n=7)

    •-49,9%

    •-63,7%

    •-50%

    •-36,9%

    •Mictions/day

    •Depressive

    •status

    •Pruritus Score

    •Flushes/day

    •PP population

  • Effects of Masitinib on psychiatric

    symptoms

  • ASL MRI outcome

    •Before Masitinib therapy •Six months after Masitinib therapy

  • •Le masitinib a démontré une activité dans deux études de phase 2 dans les deux variantes génétiques de la maladie, avec ou sans mutation D816.

    Prurit Flush Dépression Asthénie

    AB04010 sans

    D816V

    AB06013

    avec D816V

    AB04010

    sans D816V

    AB06013

    avec D816V

    AB04010

    sans D816V

    AB06013

    avec D816V

    AB06013

    avec D816V

    % de variation entre le niveau de référence et la semaine 12 (LOCF)

    N 19 21 17 6 3 9 13

    Moyenne ± SD

    -33.5 ± 43 -45.0 ± 52 -70.1 ± 50 -60.3 ±50 -40.6 ± 48 -44.2 ± 33 -38.1 ±28

    P-value 0.016 0.002 0.002 0.047 NS 0.002

  • Caractéristiques des patients

  • Masitinib Placebo p-value

    Odd ratio (CI95%)

    Critère principal

    4H75% Le taux de réponse cumulée à 75% sur les handicaps de prurit, ou de bouffée de chaleur, ou de dépression ou d’asthénie

    mITT/MDF

    mITT/OC

    18.7%

    24.1%

    7.4%

    7.9%

    0.0076*

    0.0014*

    3.63

    4.90

    •* Avec 10,000 re-randomisation en population mITT ; Résultats calculés selon la méthode MDF (Missing Data Equal Failure). Ainsi, si une donnée est manquante en raison de l’absence du patient lors d’une visite protocolaire, la réponse est considérée négative. ; OC: Cas observés

    Critères secondaires

    3H75% Le taux de réponse cumulée à 75% sur les handicaps de prurit, ou de bouffée de chaleur, ou de dépression

    mITT/MDF 24.7% 9.8% 0.0071 3.06

    2H75% Le taux de réponse cumulée à 75% sur les handicaps de prurit, ou de bouffée de chaleur

    mITT/MDF 27.2% 10.7% 0.038 2.63

    Prurit 75% Le taux de réponse cumulée à 75% sur les handicaps de prurit

    mITT/MDF 22.0% 7.3% 0.0322 3.13

    Analyses supportives

    Flush75% Le taux de réponse cumulée à 75% sur les handicaps de bouffée de chaleur

    mITT/MDF 39.9% 19.0% NS 3.03

    Dépression (Hamilton 75%) Le taux de réponse cumulée à 75% sur les handicaps de dépression

    mITT/MDF 18.6% 7.6% NS 2.71

    Asthénie (FIS 75%) Le taux de réponse cumulée à 75% sur les handicaps d’asthénie

    mITT/MDF 7.7% 3.2%

  • •Les analyses sur la réponse par patient ont confirmé le bénéfice pour les patients.

    •AB06006 –Analyses supportives – Période W8-W24

    Analyses supportives

    Patient ayant 2 handicaps lors de leur entrée dans l’étude Nombre de patients ayant une réponse (75 %) pour au moins 2 handicap(s) au cours de la période globale

    mITT/MDF 21.0% 0% NA

    Patient ayant 3 handicaps lors de leur entrée dans l’étude Nombre de patients ayant une réponse (75 %) pour au moins 3 handicap(s) au cours de la période globale

    mITT/MDF 12.5% 0% NA

    Patient ayant 4 handicaps lors de leur entrée dans l’étude Nombre de patients ayant une réponse (75 %) pour au moins 4 handicap(s) au cours de la période globale

    mITT/MDF 16.7% 0% NA

    Masitinib Placebo p-value Odd ratio

    (CI95%)

    Analyses supportives

    RÉPONSE PAR PATIENT à 75% Nombre de patients ayant une réponse (75%) pour au moins 1 handicap au cours de la période globale

    mITT/MDF 26.7% 12.8% 0.0193 2.48

    mITT / OC 34.1% 13% 0.005 4.90

    •mITT population : population en intention de traiter modifiée •Ces résultats sont calculés selon la méthode Missing Data Equal Failure (MDF). Ainsi, si une donnée est manquante en raison de l’absence du patient lors d’une visite protocolaire, la réponse est considérée négative.

    •OC: Cas observés – NA : Non applicable

  • •Le pourcentage de patients répondeurs à 50% est supérieur à 60% sur prurit, flush, et dépression.

    •AB06006 - Analyse supportive – Taux de réponse par patient et par handicap

    Alnalyse Handicape Niveau de réponse

    Taux de réponse sous Masitinib

    Au moins une réponse sur le

    handicap

    Prurit 75% 46%

    50% 60%

    Flush 75% 69%

    50% 75%

    Dépression (Hamilton) 75% 30%

    50% 65%

    Asthénie (FIS)

    75% 14%

    50% 27%

    •Non GEE model. Analysis per patient

  • Masitinib Placebo p-value Odds

    ratio(CI95%)

    Analyses à long terme

    4H75% Le taux de réponse cumulée à 75% sur les handicaps de prurit, ou de bouffée de chaleur, ou de dépression ou d’asthénie

    mITT/MDF 17.2% 7.1% 0.0102 3.37

    mITT/OC 27.1% 10.2% 0.0031 3.66

    3H75% Le taux de réponse cumulée à 75% sur les handicaps de prurit, ou de bouffée de chaleur, ou de dépression

    mITT/MDF 22.1% 8.6% 0.0030 3.10

    mITT/OC 35.1% 12.1% 0.0003 3.87

    •Le bénéfice observé au cours des six premiers mois de traitement était maintenu à 2 ans.

    •AB06006 – Analyses à long terme sur le critère principal et les critères secondaires

    •Période W8-W96

    •mITT population : population en intention de traiter modifiée •Ces résultats sont calculés selon la méthode Missing Data Equal Failure (MDF). Ainsi, si une donnée est manquante en raison de l’absence du patient lors d’une visite protocolaire, la réponse est considérée négative.

    •OC: Cas observés

  • Masitinib Placebo p-value

    Tryptase - Patients ayant un niveau de tryptase ≥20 µg/L au moment de

    leur entrée dans l’étude 46 44

    0.0001 Variation relative moyenne par rapport à la valeur de référence

    Moyenne±SD -18.0 ± 21.4 2.2 ± 26.9

    Urticaire Pigmentaire (UP) - Patients ayant de l’urticaire pigmentaire

    au moment de leur entrée dans l’étude 33 36

    0.0210 Variation relative de surface corporelle couverte d’urticaire pigmentaire

    (Correction de Wallace par rapport à la valeur de référence) -12.34 ± 26.41 15.91 ± 59.79

    Signe de Darier – Nombre de patients (au moment de leur entrée dans

    l’étude) 37 37

    0.0187 Taux de disparition du Signe de Darier (oui/non) chez les patients ayant

    le Signe de Darier lors de leur entrée dans l’étude 18.92% 2.70%

    •Le masitinib a démontré une activité sur les marqueurs objectifs de l'activation et de l’excès des mastocytes

    •AB06006 – Analyses d'efficacité basées sur des critères d’évaluation objectifs

    •Période W8-W96

  • •Chez les patients ayant la mutation D816V, le masitinib a été capable de réduire la surface couverte d’urticaire pigmentaire après un traitement à long terme.

    •AB06013 – Effets sur la peau chez un patient de sexe masculin avant (2007) et après (2008, 2014) traitement avec le masitinib

    •.

    •09-2007 VELA •03-2008 VELA

    •(+ 6 mois)

    •04-2014 VELA

    •(+ 6.5 ans)

    •Dr Stéphane BARETE – Service de dermatologie Hôpital Tenon

  • Masitinib

    (pm=1321)

    Placebo

    (pm=1031) Delta

    Au moins un événement indésirable

    (indépendamment de la causalité) 5.3 6.1 -0.8

    Décès 0.0 0.1 -0.1

    Effet indésirable sérieux non mortel (SAEs) 2.1 1.8 0.3

    Effet indésirable sévère (CTCAE grade 3/4) 3.2 2.9 0.3

    Effet indésirable entraînant le retrait du patient (non mortel) 1.5 0.4 1.1

    Effet indésirable conduisant à une réduction de dose 1.4 0.1 1.3

    •. Pm = patient-mois. Incidence: Le numérateur est le nombre de patients avec au moins un événement indésirable, le dénominateur est la somme de la durée d'exposition (en mois). Le taux est calculé pour 100 patients-mois. Delta = masitinib moins placebo

    •L'incidence des événements indésirables sur la période globale de l’étude était comparable entre les bras de traitement masitinib et placebo, sans effets toxiques mortels

    •AB06006 – Incidence (pour 100 patients-mois) des événements indésirables

    •Période globale de l’étude

    L'incidence des effets indésirables était une mesure plus pertinente que la fréquence des effets indésirables, dans la mesure où certains patients ont reçu du masitinib pendant plus de 2 ans

    La durée moyenne du traitement était de 18,9 mois pour le masitinib et de 16,4 mois pour le placebo

  • •La mastocytose systémique indolente est une maladie sévère et invalidante qui représente un fort besoin médical non satisfait.

    •Mastocytose

    •Mastocytose indolente

    •(cutanée ou systémique)

    •Mastocytose Agressive

    •Pas de symptôme ou

    •symptômes sans handicap •Symptômes avec handicap

    •Médicaments cytoréducteurs

    •Interferon

    •Cladribine

    •Temsirolimus+ Arac

    •Anticorps

    •PKC412

    •Allogreffe

    •Traitement symptomatique ou non

    •Traitement symptomatique

    •Anti H1, 2, Cromolyn, etc

    •Pas de handicap •Handicap

    •Pas d’autre traitement

    •IFN, 2CDA,

    •Masitinib ?

  • Masitinib does not inhibit c-kit D816 V ??

  • •Bien qu'il ne bloque pas la mutation D816V, le masitinib semble capable d'inhiber la dégranulation lors d’une hyperactivation des mastocytes. •

    •Le masitinib inhibe l'hyperactivation des mastocytes FcƐR-dépendent

    •The RBL-2H3 rat mast cell leukemia cell line is heterozygous for KIT, carrying both wild-type (WT) and exon 17 mutation alleles D817V . These cells have constitutive activation of KIT and are capable of degranulation upon FcɛRI-stimulation by ovalbumin (OVA). OVA administration induces β-hexosaminidase release (an established biomarker of mast cell degranulation) from these constitutive activated mast cells, whereas no degranulation is induced by stem cell factor (SCF, the ligand for c-Kit) activation.

  • HYPOTHESIS

    •MCAS

    •MSI

    •Families of MCAS and Mastocytosis

    C-Kit D816V mutation is not the only mutation that explains different phenotypes of the disease

    One or many mutations, probably germ line mutations, are responsible of mast cell activation symptoms and MCAS

    Could this mutation play a role in other inflammatory diseases?

    - Absence of correlation between : pathologic mast cell burden and intensity of mast cell activation symtoms in the same patient

    - Use of Masitinib (tyrosine kinase inhibitor): relieves mast cell activation symptoms (non efficient when D816V is mutated)

    •Crit Rev Oncol Hematol. 2015 Feb;93(2):75-89. doi: 10.1016/j.critrevonc.2014.09.001. Epub 2014 Sep 28.

    •The genetic basis of mast cell activation disease - looking through a glass darkly. Molderings GJ.

    http://www.ncbi.nlm.nih.gov/pubmed/?term=Molderings GJ[Author]&cauthor=true&cauthor_uid=25305106http://www.ncbi.nlm.nih.gov/pubmed/?term=Molderings GJ[Author]&cauthor=true&cauthor_uid=25305106

  • • Mastocytose et maladies inflammatoire mastocytoses familiales

  • • Mastocytose et maladies inflammatoires mastocytoses familiales

  • •159

    Comparaison

    des 2 populations

    nb % forme familiale nb % forme familiale prévalence

    Polyarthrite rhumatoïde 600 000 1,00% 6% 100 1,38% 19% 3,00E-04

    Spondylarthrites Ankylosantes 200 000 0,32% 10% 34 0,47% 17% 2,00E-02

    Sclérose en plaques 60 000 0,10% 6% 44 0,61% 20% 2,00E-44

    Maladie de Crohn 70 000 0,11% 25% 35 0,48% 17% 6,00E-21

    Atteints de Mastoçytose (7247 inds)population Française

  • Spectrum of MCAS

    •Mastocytosis

    •Other diseases

    •RA, psoriasis,

    •MS,IBD,PS,ICys

    •Depression

    •Idiopathic

    •Mast cell activation

  • Spectrum of MCAS

    •Mastocytosis

    •Other diseases

    •RA, psoriasis,

    •MS,IBD,PS,ICys

    •Depression

    •Idiopathic

    •Mast cell activation

    •C-kit+ X • X

    •X =kinase inhibited by Masitinib

  • Spectrum of MCAS

    •Mastocytosis

    •C-kit*

    •PLCgamma2*?

    •Other?

    •PLCgamma2*?

    •other

    •Other diseases

    •RA, psoriasis,

    •MS,IBD,PS,ICys

    •Depression

    •Idiopathic

    •Mast cell activation

    •PLCgamma2*?

    •other

    Other =Inhibited by Masitinib

  • Cancer Allergy

    Autoimmunité

    Alloreactivity

    Acute

    chronic

    Mastocytosis

    Inflammation

    fibrosis Neurological

    And

    Psychiatric

    Diseases

  • Preliminary evidence suggests that masitinib may be able to reverse hypoperfusion in mastocytosis patients, with a concomitant improvement in cognitive functions.

    •Functional disorders

    associated with mastocytosis do

    not appear in conventional MRI

    but appear on MRI measuring cerebral blood

    flow.

    •Could this also be the case in Alzheimer's

    disease?

    •Representative images from mastocytosis patient comparing ASL-MRI •before masitinib treatment (A) and after 6 month treatment (B)

    •A •B

    •PRECLINICAL DATA – ASL-MRI OBSERVATIONS

  • •BRAIN BLOOD PERFUSION: SIMILARITY MASTOCYTOSIS AND ALZHEIMER’S DISEASE

    •Representative images from a control patient (Fig. A&B), an AD patient (Fig. C), and a mastocytosis patient (Fig. D), illustrating

    the impaired cognitive functions.

    •[Figs. A&C] Alzheimers Dement. 2012 Jan; 8(1): 51–59. [Figs. B&D] Pr Bodaert Pr O. Hermine. Hôpital Necker-Paris.

    •C

    •Patients

    •Alz

    heim

    er

    •D

    •Mas

    tocy

    tosi

    s

    •B

    •Heal

    thy

    volu

    nteer

    •A

    •Control

    •Heal

    thy

    volu

    nteer

    • Measurement of cerebral blood flow using arterial spin

    labeling MRI reveals similar

    hypoperfusion patterns between

    mastocytosis patients with

    impaired cognitive functions (memory and/or attention) and AD patients

  • 167

    PUTATIVE MECHANISM OF ACTION OF MASITINIB IN ALZHEIMER’S DISEASE

    has been hypothesized that phosphorylated tau could trigger a

    neurodegenerative cascade that may require the expression of

    cell cycle markers in neurons prior to the apoptotic process [72].

    Moreover, Fyn overexpression was found to accelerate synapse

    loss and the onset of cognitive impairment in the J9 (APPswe/

    Ind) transgenic AD mouse model, while blocking the Fyn

    expression rescued synapse loss in the J20 (APPswe/Ind)

    mice [73]. While tau is generally considered an axonal protein,

    it does have a role in dendrites as well. It has been demon-

    strated that tau participates in postsynaptic targeting of Fyn to

    dendrites [74,75]. In dendrites, Fyn is capable of interacting with

    NMDARs, thus regulating receptor activity [75,76]. Fyn kinase

    could potentiate the neurotoxic process through the phosphory-

    lation of the subunit 2 (NR2B) of the NMDAR in dendritic

    spines, which results in stabilization of the receptor’s interaction

    with the postsynaptic density protein PSD-95. The exact mech-

    anism of b-amyloid–Fyn–NMDAR interaction likely involves

    the formation of the complex between b-amyloid, the prion

    protein (PrP) and the metabotropic glutamate receptor

    5 (mGluR5) which can promote Fyn activation, ultimately

    leading to the phosphorylation of the NR2B [74,77–79]. As men-

    tioned above, tau plays a key role by delivering Fyn

    postsynaptically.

    Interestingly, saracatinib (AZD0530) is another Src inhibitor

    which targets Fyn. Although at the beginning this drug was

    developed for cancer treatment, it was abandoned for this

    application. Currently, two clinical trials are testing the efficacy

    of saracatinib in early-stage AD ([80] and [81]).

    In conclusion, treatment with masitinib may provide dual

    benefits in AD pathology. On the one hand, inhibition of the

    MCs may reduce neuroinflammation via MCs–glia axis

    modulation. On the other hand, cognitive

    improvementsasaresult of Fyn inhibition

    are likely independent of b-amyloid pro-

    duction and amyloid plaqueload (FIGURE 2).

    Safety & tolerability

    Based on a number of clinical trials

    which considered masitinib for a range of

    conditions including AD, the drug

    appears to be well tolerated in humans.

    The majority of the reported AEs are

    consistent with other TKIs already on the

    market. Edema, rash, nausea, vomiting

    and diarrhea are commonly reported.

    However, these AEs are generally tran-

    sient and are well within the acceptable

    thresholds for this type of symptomatic

    treatment. For an in-depth overview of

    the safety data reported in clinical trials,

    refer to Table 4 of [9] and Table 3 of [47].

    Conclusion

    The failure of AD treatments developed

    over the last few decades demonstrates the

    complexity of the underlying pathology. Masitinib may not be

    an all-encompassing cure-all for the disease, but depending on

    the results of ongoing and future clinical trials, it may have a

    chance to becomean additional disease-modifying compound in

    the arsenal of physicians aiming to treat this debilitating

    condition.

    Expert commentary

    Over the years, a number of hypotheses have been proposed to

    explain the etiology of AD. These include: the amyloid cascade

    hypothesis, mitochondrial hypothesis, dendritic hypothesis, cell

    cycle hypothesis and the inflammatory hypothesis [4–8,10–12,18].

    If one assumes that each of these hypotheses describes an aspect

    of the disease, then the complexity of AD pathology becomes

    apparent. With the involvement of multiple signaling cascades,

    it is quite possible that a single drug could target more than

    one pathway. Such is the case of masitinib, a TKI which is

    capable of both c-kit and Fyn inhibition.

    Increased production of b-amyloid may be a cause of early-

    onset familial AD. As a result of genetic mutations affecting

    APP processing, excessive accumulation of b-amyloid in the

    brain leads to senile plaque formation. Since amyloid plaques

    are clearly not a component of a healthy brain, immune system

    response is eventually activated. Indeed, previous studies have

    shown the existence of an inflammatory process around the

    plaques, which presumably favors the loss of neighboring neu-

    rons [82–87]. Activated microglia would then be responsible for

    the generalized cerebral inflammation. If that was the principal

    driving force behind AD-related neurodegeneration, then the

    NSAID treatment would be expected to be of great benefit to

    AD sufferers. This is not entirely the case, however, as only

    Inhibition of neuroinflammator y

    process Glial cells

    Modulation of NMDA

    receptor

    Inhibition of

    tau phosphorilation

    Inhibition of FYN kinase

    Memory process improvement

    Prevention of cell cycle re-entry

    Mast cellsMasitinib

    Neuron

    Figure 2. A proposed mechanism w hereby masit inib could exert neuroprotect ive

    effects in Alzheimer’s disease. Fyn signaling in the hippocampus is, at least partially,

    regulating cell cycle re-entry, neuronal memory process, tau phosphorylation and also

    NMDA receptor. In addition, masitinib inhibits c-kit receptor in mast cells and, thus, the

    neuroinflammatory process.

    NMDA: N-methyl-D-aspartate.

    Drug Prof ile Folch, Petrov, Ettecho et a l.

    doi: 10.1586/14737175.2015.1045419 Expert Rev. Neurother.

    Exp

    ert

    Rev

    iew

    of

    Neu

    roth

    erap

    euti

    cs D

    ownl

    oade

    d fr

    om i

    nfor

    mah

    ealt

    hcar

    e.co

    m b

    y 16

    1.11

    6.33

    .27

    on 0

    5/11

    /15

    For

    per

    sona

    l us

    e on

    ly.

    B-Amyloid Substance

    CD47

    Masit inib for the treatment

    of mild to moderate

    Alzheimer’s diseaseExpert Rev. Neurother. Early online, 1–10 (2015)

    Jaume Folch1,

    Dmitry Petrov2,

    Miren Ettecho2,

    Ignacio Pedros1,

    Sonia Abad2,

    Carlos Beas-Zarate3,4,

    Alberto Lazarowski5,

    Miguel Marin6,

    Jordi Olloquequi7,

    Carme Auladell8 and

    Antoni Camins* 2,6

    1Unitat de Bioquimica i Biotecnologı́a,

    Facultat de Medicina i Ciències de la

    Salut, Universitat Rovira i Virgili, Reus,

    Tarragona, Spain2Unitat de Farmacologia I Farmacognòsia,

    Facultat de Farmàcia, Institut de

    Biomedicina (IBUB), Centrosde

    Investigacion Biomedica en Red de

    EnfermedadesNeurodegenerativas

    (CIBERNED), Universitat de Barcelona,

    Barcelona, Spain3Departamento de Biologı́a Celular

    y Molecular, C.U.C.B.A., Universidad

    de Guadalajara and Division de

    Neurociencias, Sierra Mojada 800, Col.

    Independencia, Guadalajara, Jalisco

    44340, Mexico4Centro de Investigacion Biomedica de

    Occidente (CIBO), Instituto Mexicano del

    Seguro Social (IMSS), Jalisco 44340,

    Mexico5Instituto de Investigacionesen

    Fisiopatologı́a y Bioquı́micaClı́nica

    (INFIBIOC), Facultad de Farmacia y

    Bioquı́mica, Universidad de Buenos

    Aires (UBA), BuenosAires, Argentina6Centro de Biotecnologı́a, Universidad

    Nacional de Loja, Av. Pı́o Jaramillo

    Alvarado y Reinaldo Espinosa, La

    Argelia, Loja, Ecuador7Facultad de Cienciasde la Salud,

    Universidad Autonoma de Chile, Talca,

    Chile8Departament de Biologia Cellular,

    Facultat de Biologia, Universitat de

    Barcelona, Barcelona, Spain

    * Author for correspondence:

    [email protected]

    Alzheimer’s disease (AD) is a degenerative neurological disorder that is the most common

    cause of dementia and disability in older patients. Available treatments are symptomatic in

    nature and are only sufficient to improve the quality of life of AD patients temporarily.

    A potential strategy, currently under investigation, is to target cell-signaling pathways

    associated with neurodegeneration, in order to decrease neuroinflammation, excitotoxicity,

    and to improve cognitive functions. Current review centers on the role of neuroinflammation

    and the specific contribution of mast cells to AD pathophysiology. The authors look at

    masitinib therapy and the evidence presented through preclinical and clinical trials. Dual

    actions of masitinib as an inhibitor of mast cell–glia axis and a Fyn kinase blocker are

    discussed in the context of AD pathology. Masitinib is in Phase III clinical trials for the

    treatment of malignant melanoma, mastocytosis, multiple myeloma, gastrointestinal cancer

    and pancreatic cancer. It is also in Phase II/III clinical trials for the treatment of multiple

    sclerosis, rheumatoid arthritis and AD. Additional research is warranted to better investigate

    the potential effects of masitinib in combination with other drugs employed in AD treatment.

    KEYWORDS: Alzheimer . inflammation . masitinib . neurodegeneration . tau

    Alzheimer’s disease (AD) was first described

    over a century ago by the physician Alois

    Alzheimer, but still, more than 100 years later,

    the root cause of the disorder is not

    completely understood [1]. Currently available

    pharmaceutical interventions are inadequate

    and provide only slight improvement in dis-

    ease symptoms [2,3]. Recently, researchers have

    made a number of breakthroughs in uncover-

    ing the molecular mechanisms involved in dis-

    ease pathogenesis [4,5]. This work has led to

    the identification of novel molecular targets

    and drug candidates, some of which are

    already on the market, with others in various

    stages of preclinical and clinical development.

    As the loss of cholinergic neurons in the

    frontal cortex and the hippocampus is a prom-

    inent histopathological feature of AD and

    other dementias, initial drug development

    efforts have focused on restoring central cho-

    linergic transmission [3]. Acetylcholinesterase

    inhibitors (AchEIs), which include donepezil,

    rivastigmine and galantamine, are indicated for

    the treatment of patients with mild to moder-

    ately severe Alzheimer’s dementia [2,3]. These

    compounds increase cholinergic transmission

    by inhibiting acetylcholinesterase at the synap-

    tic cleft. Unfortunately, the therapeutic efficacy

    of AchEIs is limited and the available drugs in

    this class do not halt disease progression [2,3,6].

    Cognitive decline in AD patients has also

    been linked to neuronal damage as a result of

    excitotoxicity caused by the persistent overacti-

    vation of N-methyl-D-aspartate receptor

    (NMDAR) by the amino acid glutamate [7,8].

    Memantine is an NMDAR antagonist admin-

    istered for the treatment of moderate to severe

    AD, which reduces glutamatergic excitotoxic-

    ity [3,9]. However, just as with AchEIs, the

    beneficial effects of memantine are modest,

    and the treatment is largely palliative.

    Another key aspect of AD pathology which

    hasbeen thesubject of intensiveresearch interest

    (with vast financial resources invested) concerns

    elucidating the exact role b-amyloid peptide

    plays in disease progression. It is well known

    that AD is neuropathologically characterized by

    senile(amyloid) plaques, consistingof extracellu-

    lar deposits of b-amyloid protein, and by the

    intraneuronal neurofibrillary tangles, comprising

    informahealthcare.com 10.1586/14737175.2015.1045419 Ó 2015 Informa UK Ltd ISSN 1473-7175 1

    Drug Prof ile

    Exp

    ert

    Rev

    iew

    of

    Neu

    roth

    erap

    euti

    cs D

    ownl

    oade

    d fr

    om i

    nfor

    mah

    ealt

    hcar

    e.co

    m b

    y 16

    1.11

    6.33

    .27

    on 0

    5/11

    /15

    For

    per

    sona

    l us

    e on

    ly.

    Masit inib for the treatment

    of mild to moderate

    Alzheimer’s diseaseExpert Rev. Neurother. Early online, 1–10 (2015)

    Jaume Folch1,

    Dmitry Petrov2,

    Miren Ettecho2,

    Ignacio Pedros1,

    Sonia Abad2,

    Carlos Beas-Zarate3,4,

    Alberto Lazarowski5,

    Miguel Marin6,

    Jordi Olloquequi7,

    Carme Auladell8 and

    Antoni Camins* 2,6

    1Unitat de Bioquimica i Biotecnologı́a,

    Facultat de Medicina i Ciències de la

    Salut, Universitat Rovira i Virgili, Reus,

    Tarragona, Spain2Unitat de Farmacologia I Farmacognòsia,

    Facultat de Farmàcia, Institut de

    Biomedicina (IBUB), Centrosde

    Investigacion Biomedica en Red de

    EnfermedadesNeurodegenerativas

    (CIBERNED), Universitat de Barcelona,

    Barcelona, Spain3Departamento de Biologı́a Celular

    y Molecular, C.U.C.B.A., Universidad

    de Guadalajara and Division de

    Neurociencias, Sierra Mojada 800, Col.

    Independencia, Guadalajara, Jalisco

    44340, Mexico4Centro de Investigacion Biomedica de

    Occidente (CIBO), Instituto Mexicano del

    Seguro Social (IMSS), Jalisco 44340,

    Mexico5Instituto de Investigaciones en

    Fisiopatologı́a y Bioquı́micaClı́nica

    (INFIBIOC), Facultad de Farmacia y

    Bioquı́mica, Universidad de Buenos

    Aires (UBA), BuenosAires, Argentina6Centro de Biotecnologı́a, Universidad

    Nacional de Loja, Av. Pı́o Jaramillo

    Alvarado y Reinaldo Espinosa, La

    Argelia, Loja, Ecuador7Facultad de Cienciasde la Salud,

    Universidad Autonoma de Chile, Talca,

    Chile8Departament de Biologia Cellular,

    Facultat de Biologia, Universitat de

    Barcelona, Barcelona, Spain

    * Author for correspondence:

    [email protected]

    Alzheimer’s disease (AD) is a degenerative neurological disorder that is the most common

    cause of dementia and disability in older patients. Available treatments are symptomatic in

    nature and are only sufficient to improve the quality of life of AD patients temporarily.

    A potential strategy, currently under investigation, is to target cell-signaling pathways

    associated with neurodegeneration, in order to decrease neuroinflammation, excitotoxicity,

    and to improve cognitive functions. Current review centers on the role of neuroinflammation

    and the specific contribution of mast cells to AD pathophysiology. The authors look at

    masitinib therapy and the evidence presented through preclinical and clinical trials. Dual

    actions of masitinib as an inhibitor of mast cell–glia axis and a Fyn kinase blocker are

    discussed in the context of AD pathology. Masitinib is in Phase III clinical trials for the

    treatment of malignant melanoma, mastocytosis, multiple myeloma, gastrointestinal cancer

    and pancreatic cancer. It is also in Phase II/III clinical trials for the treatment of multiple

    sclerosis, rheumatoid arthritis and AD. Additional research is warranted to better investigate

    the potential effects of masitinib in combination with other drugs employed in AD treatment.

    KEYWORDS: Alzheimer . inflammation . masitinib . neurodegeneration . tau

    Alzheimer’s disease (AD) was first described

    over a century ago by the physician Alois

    Alzheimer, but still, more than 100 years later,

    the root cause of the disorder is not

    completely understood [1]. Currently available

    pharmaceutical interventions are inadequate

    and provide only slight improvement in dis-

    ease symptoms [2,3]. Recently, researchers have

    made a number of breakthroughs in uncover-

    ing the molecular mechanisms involved in dis-

    ease pathogenesis [4,5]. This work has led to

    the identification of novel molecular targets

    and drug candidates, some of which are

    already on the market, with others in various

    stages of preclinical and clinical development.

    As the loss of cholinergic neurons in the

    frontal cortex and the hippocampus is a prom-

    inent histopathological feature of AD and

    other dementias, initial drug development

    efforts have focused on restoring central cho-

    linergic transmission [3]. Acetylcholinesterase

    inhibitors (AchEIs), which include donepezil,

    rivastigmine and galantamine, are indicated for

    the treatment of patients with mild to moder-

    ately severe Alzheimer’s dementia [2,3]. These

    compounds increase cholinergic transmission

    by inhibiting acetylcholinesterase at the synap-

    tic cleft. Unfortunately, the therapeutic efficacy

    of AchEIs is limited and the available drugs in

    this class do not halt disease progression [2,3,6].

    Cognitive decline in AD patients has also

    been linked to neuronal damage as a result of

    excitotoxicity caused by the persistent overacti-

    vation of N-methyl-D-aspartate receptor

    (NMDAR) by the amino acid glutamate [7,8].

    Memantine is an NMDAR antagonist admin-

    istered for the treatment of moderate to severe

    AD, which reduces glutamatergic excitotoxic-

    ity [3,9]. However, just as with AchEIs, the

    beneficial effects of memantine are modest,

    and the treatment is largely palliative.

    Another key aspect of AD pathology which

    hasbeen thesubject of intensiveresearch interest

    (with vast financial resources invested) concerns

    elucidating the exact role b-amyloid peptide

    plays in disease progression. It is well known

    that AD is neuropathologically characterized by

    senile(amyloid) plaques, consistingof extracellu-

    lar deposits of b-amyloid protein, and by the

    intraneuronal neurofibrillary tangles, comprising

    informahealthcare.com 10.1586/14737175.2015.1045419 Ó 2015 Informa UK Ltd ISSN 1473-7175 1

    Drug Prof ile

    Exp

    ert

    Rev

    iew

    of

    Neu

    roth

    erap

    euti

    cs D

    ownl

    oade

    d fr

    om i

    nfor

    mah

    ealt

    hcar

    e.co

    m b

    y 16

    1.11

    6.33

    .27

    on 0

    5/11

    /15

    For

    per

    sona

    l us

    e on

    ly.

  • 168

    ANIMAL MODEL (APPXPS1DE9): COGNITIVE EVALUATION IN A CURATIVE SETTING

    Masitinib was evaluated for its effect on memory deficit in AD mice (Tg) versus using the Morris Water Maze (MWM) in a curative setting (mice aged 12-14 months)

    The MWM test evaluates hippocampal-dependent learning, including acquisition of spatial memory and long-term spatial memory, which is often affected in AD.

    Blinded study

    During learning End of learning period

    Hidden platform (target)

    B Delatour et al ICM Paris

  • Genotype effect observed between wild-type (WT) and APPxPS1dE9 (Tg) mice treated with vehicle (Veh)

    1 2 3 4 5

    0

    5

    1 0

    1 5

    T o ta l d is ta n c e tra v e lle d V e h (T g v s W T 1 -5 D ) (R O M A N E 1 -2 )

    d a y s

    m

    T g V e h (n = 8 )

    W T V e h (n = 1 0 )

    •Tot

    al d

    ista

    nce (

    m)

    trav

    elled

    •by

    cont

    rol (V

    eh)

    mic

    e

    1 2 3 4 5

    0

    5

    1 0

    1 5

    T o ta l d is ta n c e tra v e lle d M a s it in ib (T g v s W T 1 -5 D ) (R O M A N E 1 -2 )

    d a y s

    m

    T g M a s it in ib (n = 9 )

    W T M a s itin ib (n = 9 )

    Disappearance of genotype effect on mice treated with masitinib

    Masitinib treatment improves cognitive function, with spatial memory returning to normal levels

    •Tot

    al d

    ista

    nce (

    m)

    trav

    elled

    •by

    mas

    itin

    ib t

    reat

    ed m

    ice

    •MWM Acquisition phase

    •Preclinical Data – Animal Model • (Cognitive Evaluation in a Curative Setting)

  • •Percentage use of spatial strategy. ** p

  • •PRECLINICAL DATA – ANIMAL MODEL (HISTOPATHOLOGY - AMYLOID LOADS)

    Masitinib treatment decrease the amyloid charges (Congo red +) in the hippocampus of young APP/PS1dE9 mice

    Tg

    Veh

    (n

    =6)

    Tg

    Masit

    inib

    (n

    =9)

    0 .0 0

    0 .0 2

    0 .0 4

    0 .0 6

    0 .0 8

    A m y lo id lo a d (H ip p o c a m p u s )

    ***

  • Alzheimer’s Disease – Previous clinical data •Phase 2 Effect of Masitinib

    •Alzheimers Res Ther. 2011 Apr 19;3(2):16. doi: 10.1186/alzrt75.

    •Efficacy results in mild-to-moderate Alzheimer's disease

    •(Phase 2 ; n=34 patients)

    -8

    -6

    -4

    -2

    0

    2

    4

    6

    8

    10

    12

    Week 0 Week 12

    p=0.035

    Week 24

    p=0.128

    Me

    an

    ch

    an

    ge

    fro

    m b

    as

    eli

    ne

    ±s

    tan

    da

    rd e

    rro

    r ADAS-ADL Masitinib Placebo

    wo

    rse

    nin

    g -

    imp

    rove

    me

    nt

    •The ADCS-ADL (Alzheimer's Disease Cooperative Study-Activities of Daily Living Inventory) is a subscale of daily living assessing items such as toileting, feeding, dressing, and physical ambulation

  • Mastocytoses

    Syndrome d’activation mastocytaire

    Mastocytoses : Prolifération clonale de mastocytes Accumulation (médullaire,

    cutanée++) Mutation somatique gène CKIT

    Syndrome d’activation mastocytaire : Activation exagérée mastocytes Pas critères de mastocytose Plus fréquent Clinique :

    • Asthénie • Flush, prurit • Douleurs ostéo-articulaires • Troubles digestifs • Troubles neuropsychiatriques • Symptômes d’allergie

    Traitement : Antihistaminiques Inhibiteurs de Tyrosine Kinase

    MALADIES LIEES AU MASTOCYTE

  • •Laboratoire d’hématologie - NEM

    •Vahid Asnafi Ludovic Lhermitte

    •Patrick Villarese

    •Julie Bruneau

    •Sophie Georgin-Lavialle

    •Zakia Belaid-Choucair

    •Geneviève Courtois

    •Marie Bouillié

    •Francine Côté

    •Pascal Amireault

    •Flavia Guillem

    •David Sibon

    •Yves Lepelletier

    •Inserm U891 - Marseille

    •Patrice Dubreuil

    •Sébastien Letard

    •Service de pathologie - NEM

    •Nicole Brousse Danielle Canioni

    •Sylvie Fraitag Stéphanie Leclerc-Mercier

    •Gisèle, Martine, Annie, Stéphanie, Marie-Aimée…

    •Service de pathologie - HMN

    •Philippe Gaulard Nicolas Ortonne

    •Nadine Martin

    •Laboratoire de cytogénétique - NEM

    •Serge Romana Isabelle Radford-Weiss

    •Gwendoline Soler

    •Centre de référence des mastocytoses - NEM

    •Olivier Hermine Christine Bodemer

    •Olivier Lortholary

    •Laboratoire Télomères - Institut Curie

    •Arturo Londono-Vallejo

    •Irena Draskovic

    Bonne année 2013!

    Sophie, Patrice, Alexandre et Thibault

    •Inserm U768 - NEM

    •Patrick Revy

    •Tangui Le Guen

    •Merci

    •U8147

    •Plateforme d’imagerie cellulaire - Imagine

    •Raphaëlle Devaux Nicolas Goudin

    •Meriem Garfa-Traore

  • Mechanism of action of Masitinib ?

  • Kinase inhibitor in Mastocytosis with C-kit D816V

    Melanocytes

    Normal Mastocytes

    Mutated Mastocytes

    SCF

    SCF

    SCF

    Urticaria pigmentosa

    Symptoms

    +

    Tumoral Syndrome

    C Findings

    D816V C-Kit* WT C-Kit

    WT C-Kit

    ITK

  • •Famillial MCAS and Mastocytosis

  • Rapamycin and MS

    •Stop IFN

    •CDA 1

    •CDA 2

    •Start IFN

    •Start Rapamycin

  • •Histamine, cytokines, protéases (tryptase), prostaglandines, ROS

    •Variations pression

    température

    •Peptides, hormones

    •IgE

    •SCF

    •c-Kit

    •HOMEOSTASIE

    •Mastocyte

    •FcεRI

    •REPONSE

    •IMMUNITAIRE

    •PROLIFERATION

    •Système cardiovasculaire

    •Vasodilatation hypotension

    •Perméabilité Œdème

    •Voies aériennes

    •Bronchoconstriction

    •Toux

    •Dyspnée

    •Peau

    •Prurit

    •Flush

    •Urticaire

    •DEGRANULATION

    •LT régulateur

    •TLRs

    •Pathogènes (Bactéries, virus,

    parasites)

    •LB

    •Cellule dendritique

    •Cytokines pro-inflammatoires (TNF-α, IL-6,

    IL-8 …)

    •OX40L

    •OX40

    •4-1BBL •HLA/Ag

    •FcεRI

    •LT effecteur

    •OX40 •4-1BB

    •TCR

    •TCR

    •CCL2, 4, 5, 10 CXCL10

    •TGFß TNF-α IL-6

    •CD40L

    •CD40

    •Histamine, PGD, PGE TNF-α, IL-

    1, IL16, IL-18

    •IL-4, IL-6, IL-13

    •FcεRI

    •IgE/Ag

    •HLA/Ag

    •HLA/Ag

    •BCR

    •Migration

    •Prolifération

    •Activation

    •Phénotype TH17

    •Migration

    •Maturation

    •Activation

    •Prolifération

    •Activation

    •Recombinaison isotypique (IgE)

    •Frenzel et al, JBS 2013

  • Anti IgE therapy

  • Psychiatric symptoms and anti IgE

    Treatment

    • 18 years old pts, Famillial history of MCAS and Mastocytosis.

    MCAS (dermographism, GI tract symptoms, Pain). No skin

    lesions, no bone marrow involvement, No C-kit D816V mutation.

    Nl tryptase level. History of chronic and acute hallucination

    episodes. Refractory to Anti- psychotic drugs. Xolair Treatment :

    total disappearance of Hallucinations.

    • 45years old pts, ISM, tryptase 45ng/ml. Episodes of flushes

    followed by acute aggressivness (Twice in trial because of acute

    violence). No efficacy of anti-psychotic drugs. MRI shows

    hypersignal in the limbic region of the white matter. Resolution of

    episodes on Xollair therapy.

  • Role of Ustekinumab (Stelara°) anti IL12/IL23

    • Young women 21 years. Past history of ISM. Flushes, GI Tract

    Pain, Fatigue, depression, memory loss. Joint pain, Lumbar pain.

    No clinical and Xray signs of psoriasis or spondylarthritis

    • Failure to Mast cell symptomatic treatment

    • Disappearance of all symptoms on Stelara treatment

    (90mg/3months)

    • 35 years old women. Past history of Pain+++, depression, fatigue,

    GI tract pain, diarrhea, flushes, dermographism, No UP, no mast

    cell infiltration. Ankylositing spondylarthritis (B27 negaive).

    Failure of Mast cell mediatior inhibitor, opoid treatmemt,anti-TNF

    therapy. Success of Stelara° on joint pain but also on most of the symptoms of MCAS including Psyschiatric

  • Cytoreductive treatment Aggressive

    diseases

    • Interferon alpha

    • Cladribine

    • Kinase inhibitors

    • Thalidomide

    • Rapamycine

  • PKC412: Discussion and perspectives

    • Longer follow up and comparison with matched patients who did not receive Midostaurin is required to assess its value to improve overall survival

    • Is complete response a goal to achieve ?

    • Use of Midostaurin in combination (2-CDA, 5-AZA, decitabine, chemotherapy) to improve the response rate and survival particularly in AHNMD, MCL and MCS

    • Use of Midostaurin before allogenous bone marrow transplantation

    • Midostaurin monitoring to improve safety and response : • Compliance

    • Absorption

    • Drug interactions

    • Organ dysfunction

    Dr P Bourget, Head Chief of the Clinical Pharmacy Department of Necker Hospital

    • Facing a multi-target drug and the need to treat for life / until progression, what do we know about the potential long term side effects ?

    – Induction of secondary neoplasia

    – Role in progression of the AHNMD

  • Perspective mTOR

    • Temsirolimus in combination

    • High efficiency of temsirolimus + high dose

    ARAC in agressive and mast cell leukemia

  • •Idolent

    •Aggressive

  • Mastocytosis

    Indolent Mastocytosis (cutaneous or systemic)

    Agressive Mastocytosis

    No symptoms or symptoms without handicap

    Symptoms with handicap

    Symptomatic treatment

    No handicap

    No further treatment Further treatment: IFN, 2CDA, TKI (Masitinib)

    mTOR, other

    Handicap

    No or symptomatic treatment

    Cytoreductive drugs Demethylating

    PKC412 Cladribine

    Temsirolimus+ Arac Others

    BMT

  • Treatment Strategy

    • Define prognosis factors

    • Define symptoms

    • C-findings : ASM or AHNMD (MDS/MPN) ?

    • Which treat first ? Or treat in combination

    • PKC412 vs Cladribine vs mTOR inhibtion

    • Chemotherapy

    • Demethylating agent

  • MANIFESTATIONS CLINIQUES

    SAM

    a

    40,8

    %

    n=

    53

    AEG:

    42,3

    %

    n=55

    HSM

    G

    32,3%

    n=42 ostéoporo

    se 23,8%

    n=31 fracture

    38.7%, n=12

    HTP/asci

    te 13,1%

    n=17

    Trouble

    s

    Digestif

    s : 30%

    n=39

    Délai médian diagnostic : 12 mois à partir des premiers

    symptômes

    Cutan

    é

    TMEP

    ou UP

    :

    25,4%

    n=33

  • Le gène KIT avait été séquencé chez 53 patients avec 44 mutations retrouvées (83%)

    GENETIQU

    E

    D816V, n=40 75%

    OTHER, n=4 8%

    Wild Type, n=4 8%

    NOT FOUND,

    n=5 9%

  • Parmi les 47 patients de la cohorte française, 32 ont eu une recherche de mutations additionnelles :

    MUTATIONS ADDITIONNELLES

    0

    2

    4

    6

    8

    10

    12

    14

    mutationTET2

    mutationJAK 2

    mutationSRSF2

    mutationASLX1

    mutationIDH2

    mutationCBL

    mutationU2AF1

    mutatioNRAS

  • TABLEAU TYPIQUE

    Clinique OMS Biologie Génétique

    AEG KIT D816V

    SAMA AHNMD Anémie +/- TET2

    HSMG Thrombopéni

    e +/- SRSF2

    Troubles

    digestifs +/- JAK2

  • ENFANTS

    ADULTE

    JEUNE

    > 70 ANS

    Signes cliniques Cutané Cutané et neuropsy AEG et

    troubles dig Stade OMS CM ISM

    AHNMD

    Biologie N N

    Anomalies NFS

    Tryptase N 1,5 – 2 N 10

    N

    Génétique KIT 50% 89%

    75%

    D816V Evolution Bénigne Bénigne Morbi-

    mortalité élevée

  • •Les sponsors…

    •MERCI

    Laboratoire d’hématologie

    Vahid Asnafi

    Ludovic Lhermitte

    Patrick Villarese

    Unité CNRS 8147

    Olivier Hermine

    Julie Bruneau

    Service de pathologie NEM

    Nicole Brousse

    Danielle Canioni

    Sylvie Fraitag

    Laboratoire de cytogénétique

    Serge Romana

    Isabelle Radford-Weiss

    Centre de référence des mastocytoses

    Olivier Hermine

    Chritine Bodemer

    Olivier Lortholary

    Stéphane Barète

    Olivia Chandesris

    Daniella Moura

    Inserm UMR 891

    Patrice Dubreuil

    Sébastien Letard

    Pr Jean-Marie Launay

    Pr Raphaël Gaillard

    Dr Harry Sokkol

    Laboratoire Télomères Curie

    Arturo Londono-Vallejo

    •Aux patients

    •Aux médecins qui incluent des patients