Download DDD107498: A novel clinical candidate for malaria

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Discovery and development of integrase inhibitors wikipedia , lookup

Neuropsychopharmacology wikipedia , lookup

Discovery and development of ACE inhibitors wikipedia , lookup

Psychopharmacology wikipedia , lookup

Drug design wikipedia , lookup

Neuropharmacology wikipedia , lookup

Discovery and development of non-nucleoside reverse-transcriptase inhibitors wikipedia , lookup

Pharmacogenomics wikipedia , lookup

Drug interaction wikipedia , lookup

Biosimilar wikipedia , lookup

Medication wikipedia , lookup

Prescription costs wikipedia , lookup

Pharmacognosy wikipedia , lookup

Pharmaceutical industry wikipedia , lookup

Bad Pharma wikipedia , lookup

Pharmacokinetics wikipedia , lookup

Drug discovery wikipedia , lookup

Theralizumab wikipedia , lookup

Bilastine wikipedia , lookup

Transcript
DDD107498: A Novel
Clinical Candidate for
Malaria
Dr. Kevin Read
NHS Research Scotland, 2016
Defeating Malaria Together
The Drug Discovery Unit
• A “Biotech company” within a university
– State of art equipment and facilities
– Current funding streams - ~£33 million
– 85 scientists, attracted from across the biopharma industry and
academia
• Translational Research Engine
– Combines excellence in discovery science with drug discovery
industry expertise
– Delivers preclinical candidates and advances novel targets via
collaboration and partnership
• Complements the pharmaceutical industry
– Neglected Tropical Diseases (NTDs)
– Innovative Targets Portfolio (ITP)
UNMET
NEED
Current Areas of Focus - NTDs
Disease
HIV / AIDS
New Cases
(per annum)
Health burden
(million
DALYsa)
Deaths
(per annum)
2 million
92
1.2 million
Tuberculosis
9.6 million
44
1.5 million
Malaria
214 million
55
0.6 million
8,000
7 million (total)
400,000
1.2
0.5
3.3
18,000
43,000
64 million
2.1
27,000
42 million (Rx)
4.0
22,000
Sleeping sickness
Chagas' disease
Leishmaniasis (VL)
Cryptosporidiosis
Schistosomiasis
a Disability-adjusted
48,000
life years lost to the community, c.f. War = 20 million DALYs
Three Approaches to Drug Discovery:
Target-based, Phenotypic and Drug Repurposing
10+ years and as much as £800 Million
Target
Selection
Assay
Development
Hit
Discovery
Hit
Validation
Hits to
Leads
Lead
Optimization
Pre-clinical
Development
Clinical
Development
Product
registration
Fully
characterised &
genetically
validated target
Target
screen
Fragment or
lead-like
compounds
Cell
screen
Drug-like
compounds
Target hits
Cell hits
Animal
screen
Existing
drugs
Leads
Preclinical
candidate
4
Track Record of Delivery – NTDs
• Phase II clinical candidate for visceral leishmaniasis
• Preclinical development for Malaria 2015
• Preclinical candidates
• Visceral leishmaniasis: Two selected in 2016; differing MoAs
• Animal trypanosomiasis: candidate in field trials
• In vivo proof of concept
• Chagas’ disease: 3 series with animal efficacy, 1 lead optimisation
• Tuberculosis: 2 series in late lead optimisation in collaboration
with TBDA
Malaria
About Malaria
Half of the world’s population, 3.2
billion people, is at risk of malaria
198 millions cases in 2013
584 000 deaths, mostly among
children under five (437 000)
90% of all malaria deaths occur in
Africa
$12 billion lost GDP
Consumes 40% of public health
spending
There are four types of human Plasmodium:
Plasmodium falciparum
Plasmodium vivax
Plasmodium malariae
Plasmodium ovale
Malaria is caused by parasites of the
species Plasmodium that are
transmitted by the bites of the
female Anopheles mosquitoes
Plasmodium falciparum and Plasmodium vivax are the most common
Plasmodium falciparum is the most deadly
Malaria Report 2014: www.who.int
Why we need new drugs for malaria
Quinolines, etc.
Chloroquine
Amodiaquine
Quinine
Mefloquine
Halofantrine
Quinoline and amino-alcohols
Resistance (20¢)
Safety / resistance
Compliance / safety / resistance
Resistance / safety / cost
Safety / resistance / cost
HN
N
HN
ACTs
Lumefantrine /
artemether (Coartem)
AS/AQ (Coarsucam)
Antifolates
Sulphadoxine /
pyrimethamine
Naphthoquinone
Atovaquone /
proguanil
N
HO
H
H3CO
N
Cl
Cl
Chloroquine
N
Amodiaquine
Quinine
Cl
F3 C
HO
N
OH
N
H
N
CH3
Cl
CH3
Artemesinins
Artemether
Arteether
Artesunate
H
H 2C
OH
N
N
CF3
CF3
Cl
Cl
Cl
H3C
Emerging resistance /
Compliance / safety / GMP
standard / availability / cost of raw
material
Mefloquine
Lumefantrine
H 3C
Artemesinin analogues
CH3
H3C
Dihydroartemsinin
Artemether
Arteether
Artesunate
O O
O
O
Resistance potential /
compliance / ($2.40) / availability
CH3
RO
R=H
R=Methyl
R=Ethyl
R=Succinyl
Other antimalarials
O
O
S
Resistance (25¢)
OH
N
Halofantrine
H 2N
N
H
N
N
N
H3C
N
OCH3
OCH3
NH2
Cl
Sulphadoxine
NH2
Pyrimethamine
Cl
O
H
N
Resistance potential / cost ($73)
OH
O
Atovaquone
Cl
H
N
NH
H
N
NH
Proguanil
CH3
CH3
Drug resistance is a major challenge…
• Signs of resistance to artemisinin have emerged in SE Asia
• Why? People often do not take complete doses, or take single agent
therapies e.g. chloroquine, or substandard drugs
Dondorp et al., N. Engl. J. Med., 2009; 361(5): 455–467
Menard et al., Nature, 2014; 505, 50–55
Malaria R&D challenges
Malaria R&D challenges
1x
CHILDREN
& PREGNANT
WOMEN
PREVENTION
OF RELAPSE
TRANSMISSION
BLOCKING
SINGLE DOSE
CURES
• Better medicines for uncomplicated malaria
•
•
Tackling resistance to first-line anti-malarials
Aiming for a single-dose cure
• Medicines for vulnerable populations
•
•
Formulations for children
Protecting pregnant women
• Medicines for malaria elimination/eradication
•
•
Stopping relapse (P. vivax)
Blocking transmission and chemoprevention
CHEMO
PREVENTION
DDU Malaria Project Starting Point:
Hit Discovery
•
In late 2009 the DDU’s focused kinase compound library was screened
phenotypically against P. falciparum
•
3 chemical series taken forward with
MMV funding starting April 2010
•
•
POTENCY
SELECTIVITY
KINETICS
SAFETY
Candidate selected October 2013
Partnered with Merck Serono for Clinical
Development in late 2014
Hit Discovery Workflow
Kinase Library - PI Frequency Distribution
Assay
development
• SYBRgreen 384-well assay validated versus
[3H]-hypoxanthine method
4731
compounds
• Screen at 3 μM against P. falciparum
• >70% inhibition in single point assay (Z’ =
0.84)
400
300
250
200
150
100
50
97
90
>110
83
104
76
69
62
55
48
41
34
PI
120 hits
• Purity and identity of hits confirmed
• Potency confirmed in 10 point assay
11 series
• Chemical clustering
• Human cell counter screen
Mefloquine
100
80
Percent Inhibition
6
27
-1
20
-8
13
-15
-22
-29
-36
-43
0
-50
Number of compounds
350
60
40
20
4 series
• Overlap with MMV portfolio
3 series
• Hit and analogues confirmed
• Mammalian kinase panel screen
0
0.0001
Hits to lead
MMV Progression Criteria used to drive project
0.01
Concentration (uM)
1
Hit Series
EARLY LEADS
Series ID
MMV02
MMV03
MMV04
EC50 vs. P f (M)
0.2
0.3
0.1
<0.1
EC50 vs. MRC5 (M)
39
> 50
20
> 10 fold
logD pH 7.4
4.0
3.2
4.3
<5
MW
342
346
418
<500
3
4
5
No. of examples <1 mol
•
MMV03 series dropped due to flat SAR
•
MMV02 series stopped for developability issues
MMV04 Series: Hits to Lead - focus on
properties
MMV04
Series
Pf EC50 (nM)
120
350
700
50
MW
418
357
377
430
LogP
4.3
3.7
3.7
2.1
Solubility
(μM)
39
36
180
>230
MLM Cli
(ml/min/g)
5.3
8.6
3.4
0.8
PPB (%)
>99
97
96
59
Meets MMV progression criteria
Lead Optimization – Getting the right balance
POTENCY
SELECTIVITY
PK
SAFETY
Potency: Pf EC50 50 nM
Bioavailability: low
Efficacy (rodent malaria):
15mg/kg bid for 4 days
Intraperitoneal injection
Potency: Pf EC50 1 nM
Bioavailability: high (74%)
Efficacy (rodent malaria):
ED90: 0.6mg/kg, Oral route
DDD107498: Active against drug-resistant
parasites
1000
EC50 (nM)
100
10
DDD107498
Artesunate
1
Chloroquine
0.1
NF54
D6 (MFQ)
(sensitive)
K1
(CQ,SUL,
PYR, CYC)
W2 (CQ,
SUL, PYR,
CYC))
7G8 (CQ,
PYR, CYC))
TM90C2A
(CQ, PYR,
MFQ, CYC)
No cross-resistance to current antimalarial drugs
V1/S (CQ,
SUL, PYR,
CYC)
CQ: Chloroquine
MFQ: Mefloquine
SUL:Sulphadoxine
PYR: Pyrimethamine
CYC:cycloguanil
Cellular Selectivity: > 20,000 fold (MRC-5) and > 59,000 (HEPG-2)
DDD107498: Active against recent clinical
isolates
 Active against Both P. falciparum and P. vivax clinical isolates
 More potent than all current drugs
P. falciparum
P. vivax
Drug
n
Median EC 50 [range] nM
n
Median EC 50 [range] nM
Chloroquine
28
106.0 [28.5-218.8]
22
165.9 [40.2-383.4]
Amodiaquine
27
16.9 [7.8-28.3]
21
26.4 [5.3-49.5]
Piperaquine
28
26.4 [7.4-58.1]
22
24.3 [3.4-81.4]
Mefloquine
28
10.6 [2.8-31.8]
22
21.3 [2.4-81.4]
Artesunate
27
6.6 [1.7-25.9]
22
3.1[0.3-19.7]
DDD107498
28
0.8 [0.5-3.3]
21
0.5 [0.3-1.4]
Fresh clinical isolates from southern Papua, Indonesia
Studies carried out by Jutta Marfurt and Ric Price, Menzies School of Health Research
DDD107498: Highly potent against multiple life
cycle stages
DDD107498: Good in vitro Developability
DDD107498
clogP
3.3
logD (experimental)
2.5
MWt
468
in vitro Cli (mouse, rat, dog, cyno.,human)
(ml/min/mg microsomal protein)
PPB % (mouse, rat, dog, human)
Solubility (SGF, FeSSIF, FaSSIF) (M)
Permeability PAMPA (nm/s)
pKa (experimental)
≤0.017, ≤0.013, 0.036, 0.038,
0.021
63, 68, 67, 77
>6410, >6410, 3970
76
8.71 and 6.82
SGF Stability
>4h
Plasma and blood stability ( mouse/human)
>8h
CYP450 Inhibition (1A2, 2C9, 2C19, 2D6,
3A4) (M)
Bioactivation risk (GSH trapping, MDI)
≥ 77
No adducts, No MDI
19
DDD107498: Pharmacokinetics in Mouse
Good oral bioavailability and long half life. Vd high
The mean concentration-time profile of DDD00107498 following a single Oral
IV base/kg
1 mg/kg (n=3)
Mouse
POmouse
3 mg/kg
(n=3)
administration at 3Mouse
mg free
to the female
NMRI
(n=3)
Clb
Vdss
Whole blood concentration (ng/mL)
T½
12 ml/min/kg
Cmax
90 ng/ml
15.0 L/kg
Tmax
1h
16.0 h
T½
19.3 h
F%
74
120.0
100.0
80.0
60.0
40.0
20.0
0.0
0
500
1000
1500
2000
Time-point (min)
P.Berghei mouse model ED90 = 0.6mg/kg PO
2500
Blood Stage Efficacy in P. falciparum
SCID mouse model
Treatment
10
0
% parasitemia
0.05
1
0.1
ED90
0.3
0.6
0.1
1
1
0
-1
-2
3
Limit of detection
0.01
ED90= 0.62 mg/Kg
log10 (% parasitemia)
DDD00107498
(mg/Kg)
Vehicle
0
1
2
3
4
5
6
7
Day after infection
ED90 0.6 mg/kg
MPC = 10-12 ng/mL
0.01
0.1
1
Dose (mg/Kg)
10
100
Targets Multiple Life-Cycle Stages:
Transmission
P. berghei
ookinetes.
EC50 ~ 5nM
Transmission
blocking stages
EC50 ~ 1-10nM
P. yoelli & P.
berghei liver
schizonts
EC50 ~ 1nM
P. falciparum &
P. vivax blood
stage form
EC50 ~ 1nM
Transmission blocking potential: In Vitro
Koen Dechering, TropIQ
• Standard Membrane Feeding Assay (SMFA)
Gamete Formation:
• Male EC50 1.8 nM
• Female EC50 1.2 nM
DDD107498 demonstrates very potent in vitro
transmission blocking potential
Transmission Blocking Potential: In Vivo
Bob Sinden/Michael Delves, Imperial College, London
• Mice infected with P. berghei and then used to feed mosquitos.
DDD107498 dosed at 3 mg/kg orally
• Significant reduction in live oocysts found in mosquitos that had
fed on treated mice
• Intensity (oocysts per midgut): reduction by 98%
• Prevalence (infected mosquitos): reduction by 90%
• Subsequent transmission to naïve mice inhibited by 90%.
• Could be due to killing gametocytes, preventing exflagellation
or oocyst development
DDD107498 demonstrates very strong in vivo
transmission blocking potential
Targets Multiple Life-Cycle Stages:
Chemoprotection
P. berghei
ookinetes.
EC50 ~ 5nM
Transmission
blocking stages.
EC50 ~ 1-10nM
P. yoelli & P.
berghei liver
schizonts.
EC50 ~ 1nM
P. falciparum &
P. vivax blood
stage form.
EC50 ~ 1nM
DDD107498: Potent Chemoprotection In Vitro
Elizabeth Winzeler, Stephen Meister, UCSD
Liver cells infected with sporozoites
Invasion
Drug
Treatment
Protocol
1
2
3
4
5
6
Liver Schizont development
DDD107498 Atovaquone
EC50 (nM)
EC50 (nM)
0.66
106
0.52
1.5
0.55
1.3
0.44
0.59
0.45
1.1
0.11
0.30
• DDD107498 incubated with P. berghei infected liver cells
• Potent across liver stage life cycle
• Potent when given for brief period during invasion (protocol 1)
• Potent when given 24 hours after infection (protocol 5)
Suggests DDD107498 may not require constant drug
pressure to have chemoprotective effect
DDD107498: Potent Chemoprotection in Vivo
Dennis Kyle, USF
• Mice dosed with
DDD107498 two hours
prior to infection with
sporozoites
• No sign of infection in
mice treated at 3 mg/kg
after day 30 and limited
sign in mice treated at 1
mg/kg (4/5 mice cured)
Percentage Mice Parasite Free, Day 30
120
100
100
100
100
80
Percent
• Exposure consistent with
other PK
80
60
40
25
20
Minimum fully protective
dose set at 3mg/kg
0
0
DDD, 10
mg/kg
DDD, 3
mg/kg
DDD, 1
mg/kg
DDD, 0.3
mg/kg
ATQ, 3
mg/kg
No
treatement
DDD107498 Mode of Action: Inhibition of
Translation Elongation Factor 2
tRNA
5’
3’
mRNA
pep de bond
forma on
5’
3’
GTP: PfeEF2
ribosome
Blocked by
DDD107498
GDP: PfeEF2
5’
•
•
DDD107498
3’
Responsible for GTP-dependent translocation of the ribosome along mRNA
Essential for protein synthesis
Asexual blood-stage P. falciparum cultured in the presence of DDD107498 at 5 x
EC50 until parasites became resistant
• Determined by sequence analysis of DNA from ten resistant mutant lines
• Confirmed by molecular and cellular biology experiments
DDD107498: Good Pharmacokinetic
profile in preclinical species
Whole Blood Concentration (ng/mL)
Beagle Dog - Oral dose (3 mg/kg)
Sprague Dawley Rat – Oral Dose (5mg/kg)
30
25
20
15
10
5
0
0
6
Long predicted human T½
12
18
24
30
36
Time after dose (hours)
42
48
Safety/Toxicology Profile of DDD107498
Phototox: No phototoxicity flag: UV scan 290-700nm
Potencies in Off-Target Assays (IC50 values)
• Ion Channels
•
•
•
•
hERG (IKr)
hNaV1.5
hKV1.5
CaV1.2
25 µM*
16 µM
> 100 µM
> 100 µM
• Dundee Kinase Panel (125 human kinases) > 10 µM
• Broad Screening (receptors, enzymes, ion channels)
•
•
Ricerca (68 targets)
CEREP (100 targets)
> 10 µM
> 10 µM
Rat 7 day safety study – no major issues, satisfactory safety margins
Genotox: Ames, in vitro and in vivo micronucleus studies negative
_________________________________________________________________________________________________________________________________________________
*hERG IC50/free Cmax at predicted human dose: 184 fold
30
DDD107498: Summary
 Possible single dose blood-stage treatment with
transmission blocking (TCP2; TCP3b) and chemoprotection
(TCP4) potential
 Excellent agreement in potency between blood and transmission assays
 Novel mode of action - active against resistant strains
 Good pharmacokinetic properties
 Low clinical drug-drug Interaction risk
 Acceptable safety margins
 No safety (toxicological) concerns have been identified to
date
 Cost of goods estimated to deliver at <$1/dose
 Under clinical development by MMV and Merck
DDD107498: Phase 0/Phase I project plan
J
2016
F M A M J J A S O N D J
Animal bio-analytical
method Validation
2017
F M A M J J A S O N D J
2018
F M A M J J A S O N D
Human bio-analytical
method Validation
Safety pharmacology
Embryo-foetal studies (DRF and main: GLP)
Pivotal toxicology studies (GLP)
Juvenile Tox (DRF: GLP)
Non-clinical Evaluation
Clinical trial supplies for Phase I
Drug Substance & Drug Product
for Phase I already available
Drug Substance development for Phase II
Drug Product development for Phase II
Clinical trial supplies for Phase IIa
CMC
Study planning
Single Ascending Dose Phase I
Food effect Phase I
Human Malaria Challenge Phase I
Clinical Development
Final report
Multiple Ascending Dose Phase I
Nature, 2015, 522,
315-320
The Dundee Team:
Beatriz Baragana
Irene Hallyburton
Neil Norcross
Caroline Wilson
Raffaella Grimaldi
Laste Stojanovski
Frederick Simeons
Maria Osuna-Cabello
Suzanne Norval
Kevin Read
Ian Gilbert
Alan Fairlamb
Acknowledgements
“Science is the most communal
of human endeavours”
Dundee
Achim Porzelle
Andrew Woodland
Jennifer Riley
Fabio Zuccotto
John Thomas
Julie Frearson
Paul Wyatt
David Gray
Columbia
David Fidock
Marcus Lee
Tara Abraham
Mariana Almedia
Rajshekhar Basak
UCSD
Elizabeth Winzeler
Stephan Meister
MMV
Paul Willis
Sir Simon Campbell
Lidiya Bebrevska
Pete Siegl
Carol Sibley
Didier Leroy
Brice Campo
Sanger
Julian Rayner
Thomas Otto
William Proto
GSK
Francisco Javier Gamo
Inigo Angulo-Barturen
Santiago Ferrer-Bazaga
Maria Santos Martinez
Laura Maria Sanz
Maria Belen Jimenez-Diaz
Imperial College
Robert Sinden
Michael Delves
Andrew Blagborough
Andrea Ruecker
Leanna Upton
Paddy Brock
Tom Churcher
Katarzyna Sala
Sara Zakutansky
SwissTPH
Sergio Wittlin
Monash
Susan Charman
Michael Campbell
TropIQ
Koen Dechering
Robert Sauerwein
Judith Bolscher
Menzies
Ric Price
Jutta Marfurt
Grennady Wirjanata
USF
Dennis Kyle
Anupam Pradhan
Eskitis Institute
Vicky Avery
Funding
MMV
Wellcome Trust
B&MGF
NIH
European Union