1
̶ Targets like an antibody ̶ Performs like a small molecule ̶ Excretes like a peptide ABSTRACT# CHEMICALLY OPTIMISED LEAD 17-69-07-N241 (N241) K D approx. 2nM Plasma stability >>20hrs = stabilising non-natural aa Days after start of dosing Tumour Volume (mm 3 ) 0 2 4 6 8 10 12 14 0 500 1000 1500 Vehicle BT17BDC17 10mg/kg BT1718 10mg/kg BT17BDC19 10mg/kg BT17BDC20 10mg/kg BT17BDC21 10mg/kg ^ ^ ^ ^ ^ ^ Helen Harrison, Peter Park, Gavin Bennett, Diane Blakeley, Amy Brown, Spencer Campbell, Liuhong Chen, Robert Lutz, Silvia Pavan, Catherine Stace, Dan Teufel, Katerine van Rietschoten, Edward Walker, Nicholas Keen Bicycle Therapeutics Limited, Meditrina Building, Babraham Research Campus, Cambridge, CB22 3AT www.bicycletherapeutics.com Discovery and development of BT1718, a novel bicyclic peptide- maytansinoid conjugate targeting MT1-MMP for the treatment of solid tumours: In vitro and in vivo activities ABSTRACT INTRODUCTION Bicycle Therapeutics has developed a proprietary phage display platform technology allowing the selection of high affinity bicyclic peptide binding molecules (Bicycles ®) Membrane type 1-matrix metalloprotease (MT1- MMP/MMP14/MT1) is a promising target for a specific targeted toxin delivery approach in oncology Bicycle peptide N241 binds MT1 with a K D of approximately 2nM, maintains species cross- reactivity and doesn’t bind related MMP proteins Bicycle Drug Conjugates® (BDCs), small molecule drug conjugates with a variety of linkers and cytotoxic payloads were prepared which retain binding affinity to MT1 The anti-tumour activity of select BDCs was demonstrated in vitro and in vivo MT1 is a cell-surface expressed metalloprotease which degrades the extracellular matrix both directly, through collagenolysis, and indirectly via activation of MMP2, promoting cell migration. MT1 is overexpressed in many tumour types including TNBC and NSCLC (fig. 1), is implicated in the invasion and metastasis of many cancers (Zarrabi et al., 2011, Journal of Biological Chemistry, 286(38), 33167– 33177) and is associated with poor patient prognosis. These properties make MT1 an attractive target for targeted tumour payload delivery. METHODS RESULTS Selections were carried out against MT1 hemopexin domain (PEX) using a Bicycle phage library with 6 amino acids in each loop. Output clones were characterised using pyrosequencing & ALPHAscreen® binding assays Peptides were synthesised by Fmoc Solid Phase Synthesis, cleaved and cyclised with a tri-functional small molecule scaffold The lead peptide 17-69-07 underwent chemical modification in which non-natural amino acid mutations were made to increase stability in plasma, creating peptide N241 Peptide N241 was conjugated to maytansinoid toxins via a panel of disulphide linkers varying in steric hindrance or a non-reducible maleimide thioether bond (table 2) Affinity and selectivity of N241 and BDCs were measured using fluorescence polarisation (FP) and Biacore™ standard methods Stability of the linkers was assessed by exposure to 10mM dithiothreitol (DTT) or in vitro human plasma and remaining parent molecule was measured using quantitative UV-LCMS/MS In vitro cytotoxicity was determined by ATP measurement and in vivo efficacy measured in xenograft models in mice implanted with tumours expressing MT1-MMP CONCLUSIONS Figure 1: MT1 over-expression in triple negative breast cancer (TNBC) (Rosse et el., PNAS 111: E1872-1879) and MT1 correlation with prognosis in non-small cell lung carcinoma (NSCLC) (Zhou et al. 2014, Oncology Letters. 7, 1395-1400) 2) Select 3) Amplify 1) Cyclise 4) Hit identification – sequencing and phage binding assay Figure 2: High diversity, high speed and high throughput Bicycle Therapeutics’ phage display and cyclisation platform identifies high affinity Bicycle binders. The platform has now been validated on over 80 targets INTIAL HIT 17-69 K D approx. 500nM BT1718 Bicycle Drug Conjugate K D approx. 3nM AFFINITY MATURATION LEAD 17-69-07 K D approx. 2nM Plasma stability = 6 hrs = newly optimised natural aa Figure 3: Development of BT1718 Cleavable linker Toxin . . Molecular spacer Protein Sequence homology to human MT1 PEX Affinity (K D in nM ± SD) (FP direct) Affinity (K D in nM ± SD) (Biacore™) Human/cyno MT1 PEX 100% 1.6 ± 1.1 (n=20) 2.6 ± 0.4 (n=3) Human/cyno MT1 ectodomain 100% 1.2 ± 0.6 (n=5) Not tested Human/cyno MT1 catalytic domain N/A >> 100 (n=3) Not tested Mouse/rat MT1 PEX 99.5% 1.2 ± 0.1 (n=2) 2.7 ± 0.6 (n=2) Dog MT1 PEX 99.5% 0.9 ± 0.4 (n=3) 2.4± 0.1 (n=2) Human MT2-MMP ectodomain 66% >> 500 (n=4) >10000 (n=1) Human MT3-MMP ectodomain 64% >> 500 (n=5) >10000 (n=1) Human MT5-MMP ectodomain 58% >> 2000 (n=2) Not tested Table 1: The Bicycle binder maintains affinity to human, rodent, dog and primate MT1-MMP and demonstrates impressive selectivity over related MMP proteins. Stability: Increasing stability of the disulphide bond was achieved by introducing methyl groups on carbon atoms adjacent to the disulphide linkage. BT17BDC21 includes a non-reducible maleimide thioether linkage which is proteolytically labile (table 2). More sterically hindered disulphide linkages were more resistant to reductive cleavage by both DTT and plasma, following the same rank order as demonstrated in antibody drug conjugates with the same linkers (Kellogg et al. 2011, Bioconjugate Chemistry, 22, 717-727). Affinity for target was maintained with all BDCs tested. BDC Linker Toxin Relative DTT stability Plasma stability (h) Binding affinity (nM) In vitro cytotoxicity (nM) BT17BDC17 DM-1 1 4 0.7 0.4 BT1718 DM-1 5 13 0.9 1.0 BT17BDC19 DM-4 30 24 0.6 0.2 BT17BDC20 DM-3 93 32 1.1 0.5 BT17BDC21 SMCC-Lys DM-1 N/A Not tested 1.1 8.9 Table 2: Structure, relative stability and affinity of BDCs. Relative DTT stability = rate of thiol-disulphide exchange normalised to that of BT17BDC17. Plasma stability = t 1/2 in in vitro human plasma (hours). Affinity = K i (nM) measured by fluorescence polarisation competition. In vitro cytotoxicity = IC50 (nM) measured in ATP endpoint assay after 72 hours incubation with HT-1080 cells Figure 4: Surface plasmon resonance (Biacore) experiments on MT1-MMP hemopexin domain demonstrate that BT1718 maintains comparable binding affinity and kinetics compared to the unconjugated peptide N241 Ka = 5.7 x 10 5 Kd = 2.8 x 10 -3 K D = 4.9 x 10 -9 Ka = 4.2 x 10 5 Kd = 3.1 x 10 -3 K D = 7.2 x 10 -9 In vitro cytotoxicity: All BDCs tested demonstrated a dose-dependent killing of HT-1080 cells. The least efficacious BDC was BT17BDC21 which would be expected to contain the most stable linker. In vivo efficacy: Treatment with BDCs containing the most labile linkers (BT17BDC17 or BT1718) showed rapid and complete tumour clearance (EBC-1 cells), while BDCs containing more stabilised linkers showed comparatively reduced efficacy (fig. 5) suggesting that target internalisation is not the sole mechanism of action for BDC efficacy and that extracellular cleavage and release of toxin within the local tumour environment likely also contribute. Only the most labile BDC (BT17BDC17) caused any significant toxicity (17% ± 9.7 body weight loss); all others were well tolerated (<10% body weight loss at 10mg/kg tiw). Optimal therapeutic index was achieved with BT1718. Testing of BT1718 in different dosing regimes in an additional model (HT-1080 cells) also demonstrated excellent tumour regression, with 10mg/kg biw leading to complete tumour clearance in all 3 animals within 23 days and no re-growth out to 70 days. Figure 5: A: In vivo efficacy of BDCs with various linkers dosed at 10mg/kg iv tiw in a cell-derived xenograft model in mice implanted with MT1-positive EBC-1 cells. B: In vivo efficacy of BT1718 dosed at 1-10mg/kg iv qw or biw in a cell-derived xenograft model in mice implanted with MT1-positive HT-1080 cells. Dosing began when tumours were approx. 180mm 3. Data represents mean of n=3 animals ± SEM. Days after start of dosing Tumour Volume (mm 3 ) 0 2 4 6 8 10 12 14 0 500 1000 1500 2000 2500 68 70 Vehicle qw BT1718 1 mg/kg qw BT1718 3 mg/kg qw BT1718 10mg/kg qw BT1718 1mg/kg biw BT1718 3mg/kg biw BT1718 10mg/kg biw A B Cys Cys Cys S N-terminus C-terminus Loop 1 Loop 2 Pharmacology Chemical scaffold constraint confers stability and energetically favours positive binding conformations Small molecule (~15 amino acid; 1.5–2 kDa) delivers advantages through rapid penetration and equilibration into the extravascular space Liver metabolism is bypassed as route of elimination is largely renal clearance Chemical diversity Size / symmetry of loops and the chemical scaffold can be altered – offers extremely high diversity in chemical space Fully synthetic nature allows site- specific conjugation to effector molecules such radionuclides High affinity (sub nM) and selectivity usually associated with antibodies N H 2 Bicycles® - A new therapeutic and diagnostic modality Bicycle N241 demonstrates high affinity for the tumour target MT1 and excellent selectivity against other MMPs Conjugation of N241 to DM1 via a range of disulphide linkers produced a panel of Bicycle Drug Conjugates which maintain high affinity for MT1 and demonstrate in vitro and in vivo efficacy, exemplifying proof of concept for an exciting new targeted delivery modality Optimal therapeutic index was achieved using a mono- hindered disulphide bond (BT1718) BT1718 has demonstrated efficacy in a wide range of MT1 positive cell and patient derived xenografts and is currently progressing well through pre-clinical development BT1718 showcases the great potential of Bicycle Therapeutics’ platform for the development of novel and transformational drugs for the treatment of a wide range of cancers PEGS 2017, Boston, MA 2017

Days after start of dosing - Bicycle Therapeutics€¦ · •Bicycle Drug Conjugates® (BDCs), small molecule drug conjugates with a variety of linkers and cytotoxic payloads were

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Days after start of dosing - Bicycle Therapeutics€¦ · •Bicycle Drug Conjugates® (BDCs), small molecule drug conjugates with a variety of linkers and cytotoxic payloads were

Targets like an antibody Performs like a small molecule Excretes like a peptide

AB

STR

AC

T#

CHEMICALLY OPTIMISED LEAD17-69-07-N241 (N241)KD approx. 2nMPlasma stability >>20hrs

= stabilising non-natural aa

Days after start of dosing

Tu

mo

ur

Vo

lum

e (

mm

3)

0 2 4 6 8 10 12 140

500

1000

1500 VehicleBT17BDC17 10mg/kgBT1718 10mg/kgBT17BDC19 10mg/kgBT17BDC20 10mg/kgBT17BDC21 10mg/kg

^ ^ ^ ^ ^ ^

Helen Harrison, Peter Park, Gavin Bennett, Diane Blakeley, Amy Brown, Spencer Campbell, Liuhong Chen, Robert Lutz, Silvia Pavan, Catherine Stace, Dan Teufel, Katerine van Rietschoten, Edward Walker, Nicholas Keen

Bicycle Therapeutics Limited, Meditrina Building, BabrahamResearch Campus, Cambridge, CB22 3ATwww.bicycletherapeutics.com

Discovery and development of BT1718, a novel bicyclic peptide-maytansinoid conjugate targeting MT1-MMP for the treatment of solid tumours: In vitro and in vivo activities

ABSTRACT

INTRODUCTION

• Bicycle Therapeutics has developed a proprietary phagedisplay platform technology allowing the selection of highaffinity bicyclic peptide binding molecules (Bicycles ®)

• Membrane type 1-matrix metalloprotease (MT1-MMP/MMP14/MT1) is a promising target for a specifictargeted toxin delivery approach in oncology

• Bicycle peptide N241 binds MT1 with a KD of approximately2nM, maintains species cross- reactivity and doesn’t bindrelated MMP proteins

• Bicycle Drug Conjugates® (BDCs), small molecule drugconjugates with a variety of linkers and cytotoxic payloadswere prepared which retain binding affinity to MT1

• The anti-tumour activity of select BDCs was demonstratedin vitro and in vivo

MT1 is a cell-surface expressed metalloprotease whichdegrades the extracellular matrix both directly, throughcollagenolysis, and indirectly via activation of MMP2,promoting cell migration. MT1 is overexpressed in manytumour types including TNBC and NSCLC (fig. 1), isimplicated in the invasion and metastasis of many cancers(Zarrabi et al., 2011, Journal of Biological Chemistry, 286(38), 33167–

33177) and is associated with poor patient prognosis. Theseproperties make MT1 an attractive target for targetedtumour payload delivery.

METHODS

RESULTS

• Selections were carried out against MT1 hemopexin domain (PEX)using a Bicycle phage library with 6 amino acids in each loop.Output clones were characterised using pyrosequencing &ALPHAscreen® binding assays

• Peptides were synthesised by Fmoc Solid Phase Synthesis, cleavedand cyclised with a tri-functional small molecule scaffold

• The lead peptide 17-69-07 underwent chemical modification inwhich non-natural amino acid mutations were made to increasestability in plasma, creating peptide N241

• Peptide N241 was conjugated to maytansinoid toxins via a panel ofdisulphide linkers varying in steric hindrance or a non-reduciblemaleimide thioether bond (table 2)

• Affinity and selectivity of N241 and BDCs were measured usingfluorescence polarisation (FP) and Biacore™ standard methods

• Stability of the linkers was assessed by exposure to 10mMdithiothreitol (DTT) or in vitro human plasma and remaining parentmolecule was measured using quantitative UV-LCMS/MS

• In vitro cytotoxicity was determined by ATP measurement and invivo efficacy measured in xenograft models in mice implanted withtumours expressing MT1-MMP

CONCLUSIONS

Figure 1: MT1 over-expression in triple negative breast cancer (TNBC) (Rosse et el.,PNAS 111: E1872-1879) and MT1 correlation with prognosis in non-small cell lungcarcinoma (NSCLC) (Zhou et al. 2014, Oncology Letters. 7, 1395-1400)

2) Select

3) Amplify

1) Cyclise

4) Hit identification – sequencing and phage binding assay

Figure 2: High diversity, high speed and high throughput Bicycle Therapeutics’phage display and cyclisation platform identifies high affinity Bicycle binders. Theplatform has now been validated on over 80 targets

INTIAL HIT17-69KD approx. 500nM

BT1718Bicycle Drug ConjugateKD approx. 3nM

AFFINITY MATURATION LEAD17-69-07KD approx. 2nMPlasma stability = 6 hrs

= newly optimised natural aa

Figure 3: Development of BT1718

Cleavable linker

Toxin.

.

Molecularspacer

Protein

Sequence homology to human MT1

PEX

Affinity (KD in nM ± SD)

(FP direct)

Affinity (KD in nM ± SD)

(Biacore™)

Human/cyno MT1 PEX 100% 1.6 ± 1.1 (n=20) 2.6 ± 0.4 (n=3)

Human/cyno MT1 ectodomain 100% 1.2 ± 0.6 (n=5) Not tested

Human/cyno MT1 catalytic domain

N/A >> 100 (n=3) Not tested

Mouse/rat MT1 PEX 99.5% 1.2 ± 0.1 (n=2) 2.7 ± 0.6 (n=2)

Dog MT1 PEX 99.5% 0.9 ± 0.4 (n=3) 2.4± 0.1 (n=2)

Human MT2-MMP ectodomain 66% >> 500 (n=4) >10000 (n=1)

Human MT3-MMP ectodomain 64% >> 500 (n=5) >10000 (n=1)

Human MT5-MMP ectodomain 58% >> 2000 (n=2) Not tested

Table 1: The Bicycle binder maintains affinity to human, rodent, dog and primate MT1-MMP and demonstrates impressive selectivity over related MMP proteins.

Stability:Increasing stability of the disulphide bond was achieved by introducing methylgroups on carbon atoms adjacent to the disulphide linkage. BT17BDC21 includesa non-reducible maleimide thioether linkage which is proteolytically labile (table2). More sterically hindered disulphide linkages were more resistant to reductivecleavage by both DTT and plasma, following the same rank order asdemonstrated in antibody drug conjugates with the same linkers (Kellogg et al.

2011, Bioconjugate Chemistry, 22, 717-727). Affinity for target was maintained with allBDCs tested.

BDC Linker ToxinRelative

DTT stability

Plasma stability

(h)

Binding affinity

(nM)

In vitro cytotoxicity

(nM)

BT17BDC17 DM-1 1 4 0.7 0.4

BT1718 DM-1 5 13 0.9 1.0

BT17BDC19 DM-4 30 24 0.6 0.2

BT17BDC20 DM-3 93 32 1.1 0.5

BT17BDC21 SMCC-Lys DM-1 N/ANot

tested1.1 8.9

Table 2: Structure, relative stability and affinity of BDCs. Relative DTT stability= rate of thiol-disulphide exchange normalised to that of BT17BDC17. Plasmastability = t1/2 in in vitro human plasma (hours). Affinity = Ki (nM) measured byfluorescence polarisation competition. In vitro cytotoxicity = IC50 (nM)measured in ATP endpoint assay after 72 hours incubation with HT-1080 cells

Figure 4: Surface plasmon resonance (Biacore) experiments on MT1-MMPhemopexin domain demonstrate that BT1718 maintains comparablebinding affinity and kinetics compared to the unconjugated peptide N241

Ka = 5.7 x 105 Kd = 2.8 x 10-3 KD = 4.9 x 10-9 Ka = 4.2 x 105 Kd = 3.1 x 10-3 KD = 7.2 x 10-9

In vitro cytotoxicity: All BDCs tested demonstrated adose-dependent killing of HT-1080 cells. The leastefficacious BDC was BT17BDC21 which would beexpected to contain the most stable linker.

In vivo efficacy: Treatment with BDCs containing the mostlabile linkers (BT17BDC17 or BT1718) showed rapid andcomplete tumour clearance (EBC-1 cells), while BDCscontaining more stabilised linkers showed comparativelyreduced efficacy (fig. 5) suggesting that targetinternalisation is not the sole mechanism of action for BDCefficacy and that extracellular cleavage and release of toxinwithin the local tumour environment likely also contribute.Only the most labile BDC (BT17BDC17) caused anysignificant toxicity (17% ± 9.7 body weight loss); all otherswere well tolerated (<10% body weight loss at 10mg/kgtiw). Optimal therapeutic index was achieved with BT1718.Testing of BT1718 in different dosing regimes in anadditional model (HT-1080 cells) also demonstratedexcellent tumour regression, with 10mg/kg biw leading tocomplete tumour clearance in all 3 animals within 23 daysand no re-growth out to 70 days.

Figure 5: A: In vivo efficacy of BDCs with various linkers dosed at 10mg/kg ivtiw in a cell-derived xenograft model in mice implanted with MT1-positiveEBC-1 cells. B: In vivo efficacy of BT1718 dosed at 1-10mg/kg iv qw or biw ina cell-derived xenograft model in mice implanted with MT1-positive HT-1080cells. Dosing began when tumours were approx. 180mm3. Data representsmean of n=3 animals ± SEM.

Days after start of dosing

Tu

mo

ur

Vo

lum

e (

mm

3)

0 2 4 6 8 10 12 140

500

1000

1500

2000

2500

68 70

Vehicle qw

BT1718 1 mg/kg qw

BT1718 3 mg/kg qw

BT1718 10mg/kg qw

BT1718 1mg/kg biw

BT1718 3mg/kg biw

BT1718 10mg/kg biw

A

B

Cys Cys

Cys

S

N-terminus C-terminus

Loop 1 Loop 2

PharmacologyChemical scaffold constraint confers stability and energetically favours positive binding conformations

Small molecule (~15 amino acid; 1.5–2 kDa) delivers advantages through rapid penetration and equilibration into the extravascular space

Liver metabolism is bypassed as route of elimination is largely renal clearance

Chemical diversitySize / symmetry of loops and the chemical scaffold can be altered – offers extremely high diversity in chemical space

Fully synthetic nature allows site-specific conjugation to effector molecules such radionuclides

High affinity (sub nM) and selectivity usually associated with antibodies

NH2

Bicycles® - A new therapeutic and diagnostic modality• Bicycle N241 demonstrates high affinity for the tumour

target MT1 and excellent selectivity against other MMPs

• Conjugation of N241 to DM1 via a range of disulphidelinkers produced a panel of Bicycle Drug Conjugates whichmaintain high affinity for MT1 and demonstrate in vitro andin vivo efficacy, exemplifying proof of concept for anexciting new targeted delivery modality

• Optimal therapeutic index was achieved using a mono-hindered disulphide bond (BT1718)

• BT1718 has demonstrated efficacy in a wide range of MT1positive cell and patient derived xenografts and is currentlyprogressing well through pre-clinical development

• BT1718 showcases the great potential of BicycleTherapeutics’ platform for the development of novel andtransformational drugs for the treatment of a wide range ofcancers

PEGS 2017, Boston, MA 2017