9 Alice Yam Sutro

Sutro Biopharma Inc.
Xpress CFTM: A rapid platform for drug development
from antibody discovery to manufacturing
August 2014
Sutro’s Xpress CF™ Platform:
Cell-Free Synthetic Biology
input
(DNA)
Assembled
IgG
+
Energy
+
Accessories*
(AA, proteins, RNAP,
metabolites)
Ribosome
(Catalyst)
output
Engineered cell-free extract
(multi-domain
(E. coli)
eukaryotic proteins)
CONFIDENTIAL
*To be incorporated into strain for commercial production
2
Advantages of Sutro’s Xpress
CF™ Platform
• Protein production is separated from biomass, allowing
–
–
–
–
Off-the-shelf protein production
Rapid expression for short make/test cycle
Expression of toxic proteins
high titer expression at g/L scales
• Xpress CFTM is an “open” system that can be
manipulated, allowing
– Easy incorporation of non-natural amino acids
– Biochemical optimization of proteins via additives
• An ideal platform for antibody discovery!
CONFIDENTIAL
3
Engineered extracts for improved
IgG folding
IgG
CONFIDENTIAL
4
Rapid Execution of Antibody
Discovery Programs
Discover novel antibody fragments using
ribosome display and screening
Express antibodies and fragments with
Sutro protein synthesis system
Reformat fragments in host of different
bispecific and antibody frameworks
Select the best lead candidate based on
in vitro and in vivo activity
5
Ribosome Display
Antigen-biotin
Antibody
Streptavidin
capture
Ribosome
PCR amplify
mRNA
+ Ag
No Ag
Screen output
CDR H3
Ribosome Display
• Ribosome Display is a selection
technology well-suited for CF
• Built-in bias for variants that
express and fold well in CF
• Selection output translates
directly into CF-based screening
Round 1
Round 2
Round 3
Round 4
CONFIDENTIAL
6
Source of Antibodies and
Integration into the Sutro System
•
•
•
•
•
Mouse
Rabbit
Chicken
Humanized Mice
Human B-cells
B-Cell Based
Technologies
Phage
Display
Bacteriophage
Display
CellBased
Display
Ribosome
Display
Bispecifics
Many
Scaffolds
• Bacteria Display
• Yeast Display
• Mammalian
Display
Sutro Cell Free
Antibody
Discovery
Antibody
Engineering
Sutro System
nnAA
Linkerwarhead
ADCs
7
Sutro’s Libraries For Antibody
Discovery
Method
Format
Framework
Diversity
Size
scFv
Optimized Human
Germline
Synthetic
>1012
Fab
Optimized Human
Germline Consensus
Synthetic
>1012
scFv
Optimized Human
Germline
Synthetic
>109
Fab
Naïve Human
Immune Repertoire
Natural
>109
Ribosome
Display
Phage Display
High Throughput Screening
Capabilities
Biacore Affinity
Ligand blocking
Selection
(1012)
Primary Screen
(1000’s)
Secondary Screen
(100’s)
ELISA
Cell binding or
internalization
Epitope Binning
CONFIDENTIAL
9
Screening workflow
Throughput/week
1000’s
Colony picking
DNA prep
Cell-free reaction &
ELISA-based ranking
100’s
Flower Plate Expression
Oasis 600
100’s
10
ELISA and FACS binding
Ligand Blocking
Biacore kinetic analysis
Detailed characterization of
variants
HT purified protein
Protein
quantitation
ELISA
And Cell
Binding
Kinetic
characterization
CONFIDENTIAL
Epitope
binning
Cell Killing
(secondary)
11
Ribosome Display: unique platform for
antibody discovery and optimization
+Ag -Ag
Rd1
Rd2
KD (nM)
Rd3
35.0
30.0
25.0
20.0
15.0
10.0
5.0
0.0
Parent
Improved
clones
New lead
Parent
0
500
1000
1500
2000
Recovered protein (ug/mL)
KD vs protein recovered
Cell Binding
CONFIDENTIAL
ELISA
12
Antibody leads can be combined
in multiple bispecific scaffolds
scFv
scFv1scFv2
scFv1-scFv2- Fc
scFv1-Fc-scFv2
scFv-scFc
scFv-Fc
scFv1-scFc-scFv2
Rapid expression of multiple scaffolds to define product format with
optimal activity and half-life
13
Rapid Screening of Bispecific
Combinations
A
A
HIK
B
B
A
A
Hole x stumpK
knob
CONFIDENTIAL
14
Bispecific-bridging of 2 targets
with AlphaLISA
HIK
A
B
A
B
CONFIDENTIAL
KIH
15
Translation of nnAA-Containing Proteins
Enables Site-Specific Conjugation
input
(DNA)
nnAA
CF Engineered
MJ TyrRS +
NNN
tRNA
mRNA
Stop
NNN
UAA
Ribosome
(Catalyst)
cell-free extract (E. coli)
Sutro Azido nnAA IgG with
DBCO-Based Linker-Warhead
Azido nnAA
Cu Free Click
Conjugation Chemistry
DBCO Linker-Peptide Warhead (MMAF)
Data Driven Design:
Production of Many Variants in Hours
SP Number
Position
TAG site
o
Mutate Sites in IgG: Choose nnAA sites using
rational design, or just make all of them!
o
Produce nnAA IgG: Incorporate nnAA at 100’s of
chosen sites
o
Conjugate: Conjugate nnAA with appropriate
chemistry
o
Purify: Separate conjugated IgG away from
unincorporated linker-warhead
o
Test: Assay conjugated IgG’s for binding and cell
killing
Light Chain: 111 Sites
Heavy Chain: 133 Sites
18
Rapid Selection of Optimal Sites for
nnAA Incorporation
1500
1000
nM
500
DAR
0.4
0.7
1.1
DAR
1.1
1.5
1.6
150
150
0
0.01 0.1
Relative Cell Viability
(Cellular ATP content, % of control)
Conjugation Efficiency (Drug/MAb Ratio)
Transform of
Transform of
Data 1
Data 1
100
50
Relative Cell Viability
MFI
2000
Herceptin
Sutroceptin
E293
K334
Viability
Relative
(Cellular Cell
ATP content,
% of control)
2500
Mean Fluorescence Intensity
SKBR3 Binding Assay
Conjugated Variants Compared to Herceptin
nnAA Incorporation and Expression
0
0.0001
Cell Killing Assay
1
10
100 1000
T359
T359
K360
N361
Q362
K360
K370
Y373
S375
W381
K370
Y373
S375
W381
S383
S383
CF-Trastuzumab
N361
nM
100
50
Q362
Pos N384
Cont ADC
N384
S136, GY S136, GY
DAR=1.6
WT HIC
WT HIC
0
0.0001
0.001
S136 ERB2 S136
ELISAERB2 E
0.001
0.01
ug/ml
0.010.1
ug/ml
mg/mL
0.1 1
1
Best Single Site ADCs Show Differential
Efficacy (single 15 mg/kg IV dose)
1600
Free Drug, 0.54mg/kg Dose equivalence at DAR of 4.0
Tumor Volume, mm3
1400
Vehicle
1200
HC Site 1, 15mg/kg
1000
Unconjugated control
HC Site 1, DAR 1.84
15mg/kg
HC Site 4, DAR 1.97 Trastuzumab-CF
AB4285 (MMAF)
HC Site 5, DAR 1.97
800
600
400
HC Site 6, DAR 1.96
200
0
0
10
20
30
40
Days
50
60
70
HerceptinTM,
1x 30mg/kg (t =0), 3x15mg/kg (weekly)
All MMAF ADC treated groups vs. vehicle
p < 0.0001**** up to d45
•
•
•
All treatments are single dose i.v. @ t=0
Multi-doses of HerceptinTM dosed i.p
No significant weight loss observed in all treatment groups
Xpress CF™
High Titers at Multiple Scales
21
Scalable and Efficient
a
[rhGM-CSF], mg/L
800
250 uL
300 mL
600
4L
100L
400
200
0
0
200
400
600
Time, min
Zawada et al, (2011) Biotech. & Bioeng.
Rapid Production of
Biotherapeutics
2
DAYS
4
6
8
Sutro Technology
Synthetic
DNA
•
•
•
•
50 µ g
protein
HTS plate
format
5g
protein
5-L
reactor
100 g
protein
100-L
reactor
Direct linear scale-up from HTS to production scale
Uses standard bioreactors & downstream equipment
Minimal, rapid process development
Gene sequence to drug substance in days
Speed to the Clinic …