Treating Tissue Factor-Positive Cancers With Antibody-Drug ......Aug 18, 2018  · maytansinoid DM1,...

29
1 Treating Tissue FactorPositive Cancers With Antibody-Drug Conjugates That Do Not Affect Blood Clotting Authors: Jan-Willem Theunissen 1, , Allen G. Cai 1 , Maryam M. Bhatti 1 , Anthony B. Cooper 2,* , Andrew D. Avery 2 , Ryan Dorfman 3 , Sebastian Guelman 1 , Zoia Levashova 1 , Thi-Sau Migone 1 Affiliations: 1 Iconic Therapeutics, South San Francisco, CA, USA 2 Adimab, Lebanon, NH, USA 3 Haematologic Technologies, Essex Junction, VT, USA * Current Address: Javelin Oncology, Lebanon, NH, USA Running title: A coagulation-inert anti-TF ADC for solid tumors Key words: Tissue Factor, TF, antibody-drug conjugate, ADC, cancer, coagulation Correspondence: Jan-Willem Theunissen, Iconic Therapeutics, 7000 Shoreline Court, Suite 270, South San Francisco, CA 94080. E-mail: [email protected] Disclosure of Potential Conflicts of interest: J.-W.T., A.G.C., M.M.B., S.G., Z.L. and T.-S.M. are employees of Iconic Therapeutics, Inc. No potential conflicts of interest were disclosed by the other authors. on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 20, 2018; DOI: 10.1158/1535-7163.MCT-18-0471

Transcript of Treating Tissue Factor-Positive Cancers With Antibody-Drug ......Aug 18, 2018  · maytansinoid DM1,...

Page 1: Treating Tissue Factor-Positive Cancers With Antibody-Drug ......Aug 18, 2018  · maytansinoid DM1, had anticancer activity, but also inhibited coagulation (19). The TF-specific antibody

1

Treating Tissue Factor–Positive Cancers With Antibody-Drug Conjugates

That Do Not Affect Blood Clotting Authors: Jan-Willem Theunissen

1,, Allen G. Cai

1, Maryam M. Bhatti

1, Anthony B. Cooper

2,*,

Andrew D. Avery2, Ryan Dorfman

3, Sebastian Guelman

1, Zoia Levashova

1, Thi-Sau Migone

1

Affiliations:

1Iconic Therapeutics, South San Francisco, CA, USA

2Adimab, Lebanon, NH, USA

3Haematologic Technologies, Essex Junction, VT, USA

*Current Address: Javelin Oncology, Lebanon, NH, USA

Running title: A coagulation-inert anti-TF ADC for solid tumors

Key words: Tissue Factor, TF, antibody-drug conjugate, ADC, cancer, coagulation

Correspondence: Jan-Willem Theunissen, Iconic Therapeutics, 7000 Shoreline Court, Suite

270, South San Francisco, CA 94080.

E-mail: [email protected]

Disclosure of Potential Conflicts of interest: J.-W.T., A.G.C., M.M.B., S.G., Z.L. and T.-S.M.

are employees of Iconic Therapeutics, Inc. No potential conflicts of interest were disclosed by

the other authors.

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 20, 2018; DOI: 10.1158/1535-7163.MCT-18-0471

Page 2: Treating Tissue Factor-Positive Cancers With Antibody-Drug ......Aug 18, 2018  · maytansinoid DM1, had anticancer activity, but also inhibited coagulation (19). The TF-specific antibody

2

Abstract:

The primary function of Tissue Factor (TF) resides in the vasculature as a cofactor of blood

clotting; however, multiple solid tumors aberrantly express this transmembrane receptor on the

cell surface. Here, we developed anti-TF antibody-drug conjugates (ADCs) that did not interfere

with the coagulation cascade and benchmarked them against previously developed anti-TF

ADCs. After screening an affinity-matured antibody panel of diverse paratopes and affinities, we

identified one primary paratope family that did not inhibit conversion of Factor X (FX) to

activated Factor X (FXa) and did not affect conversion of prothrombin to thrombin. The rest of

the antibody panel and previously developed anti-TF antibodies were found to perturb

coagulation to varying degrees. To compare the anti-cancer activity of coagulation-inert and –

inhibitory antibodies as ADCs, a selection of antibodies was conjugated to the prototypic

cytotoxic agent monomethyl auristatin E (MMAE) through a protease-cleavable linker. The

coagulation-inert and –inhibitory anti-TF ADCs both killed cancer cells effectively. Importantly,

the coagulation-inert ADCs were as efficacious as tisotumab vedotin, a clinical stage ADC that

affected blood clotting, including in patient-derived xenografts from three solid tumor

indications with a need for new therapeutic treatments—squamous cell carcinoma of the Head

and Neck (SCCHN), ovarian and gastric adenocarcinoma. Furthermore, a subset of the anti-TF

antibodies could also be considered for the treatment of other diseases associated with

upregulation of membranous TF expression, such as macular degeneration.

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 20, 2018; DOI: 10.1158/1535-7163.MCT-18-0471

Page 3: Treating Tissue Factor-Positive Cancers With Antibody-Drug ......Aug 18, 2018  · maytansinoid DM1, had anticancer activity, but also inhibited coagulation (19). The TF-specific antibody

3

Introduction

An emerging class of anticancer treatments consists of ADCs. These agents combine the

specificity of an antibody against a membrane-associated protein with the cytotoxicity of a

chemotherapeutic compound. With four ADCs—ado-trastuzumab emtansine, brentuximab

vedotin, inotuzumab ozogamicin and gemtuzumab ozogamicin—approved for treatment (1-3),

more than fifty ADCs are at various stages of clinical development (2,4). Emerging clinical data

suggests that a subset of these ADCs have a relatively narrow therapeutic window (5,6). Novel

linkers and payloads are anticipated to widen this window (4,7,8). Antibodies that do not impact

the normal function of the target could also improve the safety and tolerability of the ADC (6).

For example, the anti-EGFR ADCs depatuximab mafodotin and ABBV-221 target a tumor-

selective EGFR epitope, with low normal tissue binding (9). Furthermore, improvements in

antibody technology enable isolation of candidate antibodies with the desired binding affinity,

species cross-reactivity, cellular internalization, and biological activity or lack thereof (10).

Tissue Factor (TF; also known as coagulation factor III (F3), thromboplastin, CD142) is a

promising ADC target. Under normal physiological conditions, membranous TF expression is

restricted (11,12). Vascular injury results in exposure of cell surface TF to blood. Once Factor

VII (FVII) binds TF, limited proteolysis by plasma proteases results in conversion of FVII into

activated FVII (FVIIa). The TF:FVIIa complex subsequently binds and converts FX into its

activated form, FXa. Cleavage of prothrombin into thrombin by FXa in association with FVa and

divalent calcium is followed by platelet activation and thrombin-mediated cleavage of fibrinogen

to fibrin (11).

In contrast to restricted surface expression in normal tissues, TF exhibits membranous TF

expression on multiple solid tumor indications (13). Surface TF is expressed in gastrointestinal

cancers, urogenital cancers, gliomas, melanomas, lung cancer and breast cancer (13). While TF

has not been identified as an oncogene, its expression is regulated by hypoxia in the tumor

microenvironment and various cancer mutations, such as oncogenic Ras and loss of PTEN

(13,14). In cancer cells, co-expression of TF with its ligand, FVII, and its signaling receptors,

protease activated receptors (PARs), can promote tumor growth by inducing immune-modulating

and pro-angiogenic cytokines (14). Aside from its surface expression in cancer, TF undergoes

efficient internalization and lysosomal targeting (15,16), enabling efficient delivery of ADCs

(15).

TF-specific ADCs from three different preclinical studies have been reported to be

efficacious in cancer xenografts (17-19). The ADC from the first preclinical study, tisotumab

vedotin (17), has advanced into a Phase IIa trial, with a clinical response in one third of patients

with cervical cancer (20). However, adverse events associated with perturbation of coagulation

were detected (20,21). The antibody moiety of tisotumab vedotin competed directly with FVII

for binding to TF and had a modest impact in the selected in vitro coagulation assays (17). In the

second preclinical study, the TF-specific antibody SC1, conjugated to MMAE or the

maytansinoid DM1, had anticancer activity, but also inhibited coagulation (19). The TF-specific

antibody used in the third preclinical study of an ADC interfered with clotting by inhibiting

binding of FVII to TF (18,22).

In this study, we set out to develop a TF-specific ADC with no impact on the coagulation

cascade. An antibody panel with diverse paratopes and affinities was evaluated for perturbation

of coagulation and anti-cancer activity. One paratope family of antibodies in particular did not

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 20, 2018; DOI: 10.1158/1535-7163.MCT-18-0471

Page 4: Treating Tissue Factor-Positive Cancers With Antibody-Drug ......Aug 18, 2018  · maytansinoid DM1, had anticancer activity, but also inhibited coagulation (19). The TF-specific antibody

4

affect blood clotting, and when conjugated to MMAE showed anticancer efficacy comparable to

tisotumab vedotin.

Materials and Methods

Proteins

Human and cynomolgus monkey TF extracellular domain (ECD) fragments were expressed as

C-terminal His or Fc-gamma fragment fusions using recommended procedures (ThermoFisher

Scientific, Waltham, MA, USA). Details are provided in the Supplementary Materials and

Methods section. The sequence identity between the extracellular domains of human and

cynomolgus monkey TF is 94.5 %.

Antibodies

Human antibodies against human TF were isolated from a full-length human IgG library using an

in vitro yeast selection system and associated methods (23). A synthetic naïve human antibody

library of ~1010

in diversity, was designed, generated in yeast, and propagated as described

previously (23,24). TF-binding mAbs were enriched by incubating biotinylated TF-His

monomers at different concentrations with antibody expressing yeast cells followed by magnetic

bead selection (Miltenyi Biotec, Auburn CA) and fluorescence-activated cell sorting on a

FACSAria II cell sorter (BD Biosciences, San Jose CA) using streptavidin secondary reagents in

several successive selection rounds. After the last round of enrichment, yeast cells were plated

onto agar plates, clones were analyzed by DNA sequencing and used for IgG production. Heavy

chains from the naïve outputs were used to prepare light chain diversification libraries, which

were then used for additional selection rounds. In particular, heavy chains were extracted from

the fourth naïve selection round outputs and transformed into a light chain library to create new

libraries ~108 in diversity. Six human TFspecific antibodies (mAb 1, 25, 29, 39, 43 and 54)

were prioritized for affinity maturation in two phases: additional diversification of the heavy

chain followed by additional diversification of the light chain. Further diversification of the

heavy chain was accomplished by diversification of CDR-H1 and CDR-H2, and diversification

of CDR-H3 following CDR-H1 and CDR-H2 diversity pool optimization for improved binding

affinity. CDR-H1 and CDR-H2 were diversified with a premade library with CDR-H1 and CDR-

H2 variants of a diversity of ~108. CDR-H3 was diversified by constructing a library with

oligonucleotide-based NNK degeneracy across the entire CDR-H3. Affinity-matured antibodies

that underwent the first phase of affinity maturation contain a number and letter in their

identifiers, and the number of VH CDR amino acid changes in each antibody relative to the

parent antibody is listed in Supplementary Table S1A. Further diversification of the light chain

was accomplished by diversification of CDR-L3 within CDR-L1 and CDR-L2 diversity pools

selected for improvements in binding affinity. CDR-L3 diversification was accomplished by

constructing a library with oligonucleotide-based NNK degeneracy across the entire CDR-L3.

The antibodies that underwent this second phase of affinity maturation contain an additional

number at the end of their identifiers, and the number of VL CDR amino acid changes in each

antibody relative to the parent antibody is listed in Supplementary Table S1B. While the

screening campaign was conducted with yeast-derived antibody material (Fig. 1), Expi293-

derived antibody material was used for the remainder of this study. Also, a germ-line point

mutation in framework region 3 of 43E was removed in 43Ea.

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 20, 2018; DOI: 10.1158/1535-7163.MCT-18-0471

Page 5: Treating Tissue Factor-Positive Cancers With Antibody-Drug ......Aug 18, 2018  · maytansinoid DM1, had anticancer activity, but also inhibited coagulation (19). The TF-specific antibody

5

Alexa Fluor antibody and FVII-Fc conjugates were generated using Alexa Fluor 488 5-

sulfo-dichlorophenol esters (A488) (ThermoFisher Scientific).

Cell lines

The A431, HCT-116, MDA-MB-231, HPAF-II and RF/6A cell lines were obtained from the

American Tissue Culture Collection (ATCC, Manassas, VA, USA) and were maintained as

recommended. All cell lines from ATCC have been thoroughly authenticated using morphology,

karyotyping and PCR-based approaches to rule out intra- and interspecies contamination. CHO

cells were acquired from ThermoFisher Scientific. Assays were conducted with cells that did not

exceed 15 passages in culture.

FX conversion assay

To evaluate the ability of TF:FVIIa to convert FX into FXa in the presence of human antibodies

against TF, a cell-based FX conversion assay was conducted (25). Briefly, 5x104 MDA-MB-231

cells were plated into 96-well plates (Greiner Bio-One, Monroe, NC, USA) and cultured

overnight. After removal of the cell culture media and addition of FX at a final concentration of

200 nM in a HEPES buffer with 1.5 mM CaCl2, cells were incubated with 50 nM or a titration of

the antibodies for 15 min at 37C. Upon reconstitution of the binary TF:FVIIa complex with a

final concentration of 20 nM of FVIIa, cells were incubated for 5 min at 37C. After quenching

the reaction with ethylenediaminetetraacetic acid (EDTA) in a black 96-well plate, generated

FXa was measured with 50 M of SN-7 6-amino-1-naphthalenesulfonamide-based fluorogenic

substrate (Haematologic Technologies, Essex Junction, VT, USA). Percent FXa generation in the

presence of anti-TF antibody relative to a no-antibody control was reported.

Cell viability assay

To evaluate internalization of the anti-TF antibodies, a secondary ADC cytotoxicity assay was

conducted as previously described (26). Cells were plated in 384-well plates (Greiner Bio-One).

Antibodies and an anti-human Fc Fab conjugated to the tubulin inhibitor mono-methyl auristatin

F (MMAF) (Moradec, San Diego, CA, USA) were serially diluted as shown, starting at 5 and 30

nM, respectively. The anti-human Fc Fab conjugated to MMAF consisted of a polyclonal

antibody specific to the Fc region of human IgGs with a DAR of 1.2 to 1.5. Plates were

incubated for 3 to 5 days, followed by lysis in CellTiter-Glo (CTG) assay reagent (Promega,

Madison, WI, USA). The mean and standard deviation of 4 replicates were graphed in Prism

(GraphPad, La Jolla, CA, USA). For each ADC complex, the IC50 and its associated 95 %

confidence interval were calculated in Prism using a 4-parameter binding model.

Binding assays

Cell-based antibody binding was assessed by flow cytometry as previously described (27).

Details are provided in the Supplementary Materials and Methods. The median fluorescence

intensities (MFIs) at each dilution were plotted and EC50’s derived using a 4-parameter binding

model in Prism.

Kinetic measurements for the anti-TF antibodies were conducted on an Octet QK384

(Pall ForteBio, Fremont, CA, USA) or a Biacore (GE Healthcare Bio-Sciences, Marlborough,

MA, USA). ForteBio affinity measurements were performed during the screening campaign (Fig.

1 and Supplementary Table S1) as previously described (28). Details are provided in the

Supplementary Materials and Methods. For the Biacore-based measurements (Supplementary

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 20, 2018; DOI: 10.1158/1535-7163.MCT-18-0471

Page 6: Treating Tissue Factor-Positive Cancers With Antibody-Drug ......Aug 18, 2018  · maytansinoid DM1, had anticancer activity, but also inhibited coagulation (19). The TF-specific antibody

6

Table S2), the antibody was covalently coupled to a chip using an amine-coupling kit and a five-

point titration of TF-His was analyzed using recommend procedures (GE Healthcare Bio-

Sciences). Kinetic data was analyzed and fitted globally using a 1:1 binding model.

Thrombin Generation Assay (TGA)

The TGA was performed using the calibrated-automated-thrombogram (CAT) instrument

(Diagnostica Stago SAS, Asnières sur Seine, France) (29). The test method design was

equivalent to a standard CAT assay measurement, except that the plasma source was normal

pooled plasma (NPP) collected in 11 mM citrate supplemented with 100 g/mL of corn trypsin

inhibitor. The anti-TF antibodies were diluted and mixed with NPP. Relipidated TF was added to

a 96-well assay plate, followed by addition of the antibody/NPP mixture. Directly after

combining the relipidated TF with the antibody/NPP, thrombin generation was initiated by the

addition of calcium and the thrombin substrate. The STAGO software was used to report the

following parameters: Peak IIa (highest thrombin concentration generated on the thrombin

generation curve [nM]); Lag Time (time from assay start to the moment 10 nM of thrombin is

formed [min]); ETP (endogenous thrombin potential, area under the curve [nM x min]); and

ttPeak (time from assay start to Peak IIa [min]). Percent peak thrombin generation (% Peak IIa),

percent endogenous thrombin potential (% ETP), and percent ttPeak (% ttPeak) in the presence

of each antibody relative to a no-antibody plasma control on the same plate were also reported

(Supplementary Table S3).

FVII competition assay

To evaluate competition between FVII and the human antibodies against TF, TF-positive A431

cells were first incubated for 1 h on ice with a titration of the human antibodies against TF or an

IgG1 isotype control. Subsequently, FVII-Fc conjugated to A488 was added to the antibody-cell

mixture at a final concentration of 20 nM. After another 1 h incubation on ice, cells were

washed, stained with a viability dye, and analyzed by flow cytometry. The A488 fluorescence

data from viable cells was summarized using MFI. FVII-Fc binding was summarized with %

FVII binding = [MFIanti-TF antibody labeled cells – MFIunstained cells] / [MFIIgG1 control labeled cells –

MFIunstained cells].

Generation of ADCs

Antibodies were conjugated to MC-vc-PAB-MMAE (maleimidocaproyl-valine-citrulline-p-

aminobenzyloxycarbonylmonomethyl auristatin E) as previously described (30,31). Details are

provided in the Supplementary Materials and Methods. Hydrophobic interaction chromatography

and size exclusion chromatography were used to corroborate the absorbance-based DAR

estimation and to ensure the ADC preparation was at least 95 % monomeric, respectively.

ADC cytotoxicity assays

To evaluate ADC cytotoxicity, cells were plated in 384-well plates (Greiner Bio-One). Anti-TF

antibodies conjugated to MC-vc-PAB-MMAE were serially diluted as shown. Plates were

incubated for 3 days, followed by lysis in CTG assay reagent. CTG luminescence was measured

and the mean and standard deviation of 4 replicates graphed in Prism. For each ADC, the IC50

and its associated 95 % confidence interval (95 % CI) were calculated in Prism using a 4-

parameter binding model. When ranking the TF-specific ADCs in Supplementary Table S4, the

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 20, 2018; DOI: 10.1158/1535-7163.MCT-18-0471

Page 7: Treating Tissue Factor-Positive Cancers With Antibody-Drug ......Aug 18, 2018  · maytansinoid DM1, had anticancer activity, but also inhibited coagulation (19). The TF-specific antibody

7

overall continuous primary ADC rank was calculated by equally weighing the rank obtained in

A431, MDA-MB-231 and HPAF-II cultures.

Immunofluorescence

Immunofluorescence was conducted as described previously (32). Details are provided in the

Supplementary Materials and Methods. The microtubule networks and nuclei were stained with

anti-tubulin (11H10) rabbit mAb (A488 conjugate) (Cell Signaling Technology, Danvers, MA,

USA) and DAPI, respectively.

TF signaling assay

IL-8 and GM-CSF protein levels were measured as described previously (33). Details are

provided in the Supplementary Materials and Methods. A standard curve using recombinant

GM-CSF or IL-8 (R&D Biosystems, Minneapolis, MN, USA) was used in Prism to calculate

cytokine concentration in the cell culture supernatants. Percent IL-8 and GM-CSF (% IL-8 an %

GM-CSF) at the reported antibody concentration were calculated relative to a no-antibody

control.

Antibody-dependent cellular cytotoxicity (ADCC)

To evaluate ADCC activity, an ADCC Reporter Bioassay Core Kit (Promega) was used

following the manufacturer’s protocol. Details are provided in the Supplementary Materials and

Methods. The mean and standard deviation of 4 replicates were graphed in Prism. For each

antibody and ADC, the EC50 and its associated 95 % confidence interval were calculated in

Prism using a 4-parameter binding model.

Cell line–derived xenograft (CDX) models

To evaluate the efficacy of the ADCs in vivo, xenograft studies in immune compromised mice

were performed as described (26). Details are provided in the Supplementary Materials and

Methods. The animals’ care was in accordance with institutional guidelines. Mean tumor volume

(MTV) with the standard error of the mean (SEM) was plotted over time. Treatment efficacy was

determined by calculating tumor growth inhibition (% TGI = 100 % x [1-(final MTV initial

MTV of a treated group) / (final MTV – initial MTV of the control group)]) before any of the

animals in the vehicle arm were euthanized due to a tumor size ≥ 1200mm3. Statistical

comparisons between the MTVs were conducted using one-way ANOVA followed by Tukey’s

multiple comparisons test comparing all groups. The P-values for each ADC compared to the

vehicle control arm are reported. At the end of the study, efficacy was also determined in each

treatment arm by counting the number of animals with a partial regression (PR) or a complete

regression (CR) of the tumor. In a PR response, the tumor volume was 50 % or less of its day 1

volume for 3 consecutive measurements during the course of study, and equal to or greater than

14mm3 for 1 or more of these measurements. In a CR response, the tumor volume was less than

14mm3 for 3 consecutive measurements. When an animal exhibited a CR response at the end of

the study, it was classified as a tumor-free survivor (TFS) instead of a CR. Throughout the ADC

studies no significant body weight changes due to ADC treatment were observed.

Patient-derived Xenograft (PDX) models

TF-positive PDX models were performed in athymic nude mice (Envigo, Indianapolis, IN) to

evaluate the efficacy of the ADCs in vivo. The animals’ care was in accordance with institutional

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 20, 2018; DOI: 10.1158/1535-7163.MCT-18-0471

Page 8: Treating Tissue Factor-Positive Cancers With Antibody-Drug ......Aug 18, 2018  · maytansinoid DM1, had anticancer activity, but also inhibited coagulation (19). The TF-specific antibody

8

guidelines. Study animals were implanted unilaterally on the left flank with tumor fragments.

Animals were randomized and treated as indicated in the figures. Animals were removed from

study and euthanized once tumor size reached 1200mm3 or skin ulceration was evident. In

addition, the MTV curve for the treatment group in question was no longer shown once an

animal was removed from study due to size. TGI and statistical analyses were conducted in the

same manner as for the CDX studies. The CR and PR response definitions were as follows for

the PDX studies: a PR responder had a MTV ≤ 30 % of MTV at day 1 for 2 consecutive

measurements; a CR responder had an undetectable MTV for 2 consecutive measurements.

Immunohistochemistry

Formalin-fixed paraffin-embedded (FFPE) tissues were sectioned at 4-micron thickness and

mounted onto positive-charged glass slides. The tissue sections were stained with the anti-TF

antibody HTF-1 (ThermoFisher Scientific). Details are provided in the Supplementary Materials

and Methods.

Results

Screen to identify antibodies against tissue factor that do not affect FX conversion and

exhibit cellular internalization

In a discovery campaign with recombinant TF, six human TFspecific antibodies (1, 25, 29, 39,

43 and 54) were prioritized for antibody affinity maturation. Each antibody selected for

optimization was from a different VH lineage, potentially resulting in greater epitope diversity,

and thereby enabling selection of antibodies that do not impact coagulation and internalize

efficiently. Affinity maturation of these six TF antibodies by diversification of the heavy chain

complementary determining regions (CDR) resulted in a panel of fifty-four TF-specific

antibodies with affinities ranging between 0.5 and 181 nM (Supplementary Table S1A). Within

each VH lineage, affinities varied at least eight-fold, prompting bivariate analysis between

affinity and activity for each lineage.

To identify TF-specific antibodies that do not impact conversion of FX into FXa by the

TF:FVIIa complex, we screened the parent and affinity-matured antibodies in a cell-based FX

conversion assay. With a relatively small number of antibodies as a potential caveat for antibody

group 39, a correlation analysis between affinity and inhibition of FX conversion within each VH

lineage indicated that four of the six VH lineages exhibited moderate to strong correlation

between the two parameters (Fig. 1A). In the four antibody groups with a substantial affinity-

activity relationship—groups 1, 29, 39 and 54—antibodies with an affinity less than or equal to 5

nM exhibited more than 45 % inhibition (Figure. 1A, Supplementary Table S1A). In the two

antibody groups without a substantial affinity-activity relationship—groups 25 and 43—the

affinity ranges were 16 -181 nM and 1.714 nM, respectively. While the lack of an affinity-

activity relationship for group 25 antibodies could be attributed to modest affinities, group 43

antibodies with an affinity less than or equal to 5 nM did not impact FX conversion by more than

10 % in the assays used (Figure. 1A, Supplementary Table S1A).

With the intent of developing a therapeutic ADC, internalization properties of the fifty-

four TF-specific antibodies were evaluated in a cell-killing assay (Fig. 1B). Modest to strong

correlation was observed between affinity and ADC IC50 for groups 25, 29, 39, 43 and 54 (Fig.

1B), indicating that improved affinity correlated with more efficacious killing of cancer cells.

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 20, 2018; DOI: 10.1158/1535-7163.MCT-18-0471

Page 9: Treating Tissue Factor-Positive Cancers With Antibody-Drug ......Aug 18, 2018  · maytansinoid DM1, had anticancer activity, but also inhibited coagulation (19). The TF-specific antibody

9

However, bivariate analysis of antibodies from group 1 revealed that improved affinity did not

correlate substantially with improved ADC IC50 for this group (Fig. 1B).

Additional antibody affinity maturation to identify FX conversion–inert high-affinity

antibodies with improved internalization characteristics

Based on the lack of inhibition in the FX conversion assay for antibodies from groups 25 and 43,

two members from each group were selected for additional affinity maturation to test whether

improved binding affinity would affect activity in the FX assay. Affinity maturation by

diversification of the light-chain CDRs resulted in a panel of fifty-two TF-specific antibodies

with affinities ranging from 0.6 to 26 nM (Supplementary Table S1B).

None of the affinity-matured antibodies in this second panel affected FX conversion by

more than 15 % (Fig. 1C). In addition, affinity maturation of group 25 led to a modest

improvement in cell death (Fig. 1D). With some of the affinity-matured antibody clones from

family 25 having binding affinities less than 1 nM, additional affinity maturation was not

pursued.

A panel of ten TF-specific antibodies was selected for further characterization and a

comparison against previously developed anti-TF antibodies. We prioritized antibodies that were

inert or inhibitory in the FX conversion assay: six antibodies with varying affinities from the two

groups that did not substantially affect FX conversion (i.e. 25A, 25A3, 25G1, 43B1, 43D7 and

43E) and the highest affinity antibody from each group that inhibited FX conversion by more

than 50 % (i.e. 1F, 29E, 39A and 54E).

Binding characteristics of anti-TF antibodies

To ensure the difference in coagulation activity between the prioritized antibodies was not driven

by different binding properties, the panel of antibodies was further evaluated in cell- and protein-

based binding assays. The ten antibodies bound the TF-positive cancer cell line HCT-116 in a

dose-dependent manner with a range of EC50 values between 0.04 and 0.53 nM (Fig. 2A,

Supplementary Table S2). The cell binding properties of the FX–inert and FX–inhibitory

antibody categories were equivalent, with a similar range of EC50 values for each category. Only

a two-fold difference was observed between the lowest EC50 in the FX–inert and –inhibitory

antibody category, with 25A3 at 0.08 nM and 39A at 0.04 nM. A two-fold difference was also

observed between the highest EC50 antibody in the FX-inert and –inhibitory category, with 43Ea

at 0.53 nM and 54E at 0.24 nM. Cell-based binding was confirmed in two additional cancer cell

lines (Supplementary Fig. S1A and S1B). Biacore-based affinity measurements with a titration of

human TF indicated that the TF-specific antibodies had affinities between 0.06 and 6.2 nM

(Supplementary Table S2).

Throughout the TF-specific antibody selection campaign, affinity and cell binding were

also evaluated on TF from cynomolgus monkey, the nonhuman primate to be used for

toxicology. The Biacore-based affinity for human and cynomolgus TF differed less than 5-fold

for all the TF-specific antibodies, with 29E as an exception (Supplementary Table S2). With the

protein sequences of Macaca fascicularis and mulatta TF being identical, binding of the TF-

specific antibody panel was also confirmed on the Macaca mulatta RF/6A cell line

(Supplementary Table S2). Finally, while the anti-TF antibodies were selected against

glycosylated TF from a human cell line, the antibodies also bound E. coli-derived TF

(Supplementary Table S2).

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 20, 2018; DOI: 10.1158/1535-7163.MCT-18-0471

Page 10: Treating Tissue Factor-Positive Cancers With Antibody-Drug ......Aug 18, 2018  · maytansinoid DM1, had anticancer activity, but also inhibited coagulation (19). The TF-specific antibody

10

In-depth characterization of anti-TF antibodies in coagulation assays

To corroborate the screening results, the FX conversion assay was performed with an eight-point

antibody titration. As part of this effort, the TF-specific antibodies TF-011 (the antibody used in

tisotumab vedotin), 5G9 and 10H10 were included as comparators (17,34). While 10H10 and the

antibodies from lineage 25 and 43 were considered inert with less than 20 % inhibition at all

concentrations tested, TF-011, 5G9 and the antibodies from lineage 1, 29, 39 and 54 exhibited

higher inhibition (Fig. 2B and Supplementary Fig. S1C). Antibody 29E inhibited FX conversion

by 32-37 % at the three highest concentrations (25, 50 and 100 nM), and 1F, 39A and 54E

inhibited by 56-62 % at these concentrations (Fig. 2B). TF-011 and 5G9 inhibited FX conversion

by 57-59 % and 67-70 % at these three concentrations (Supplementary Fig. S1C).

To understand the impact of diminished FXa generation by the TF-specific antibodies on

the penultimate step in the coagulation cascade, a thrombin generation assay (TGA) was

conducted (29). The TGA is considered a promising tool for monitoring patients on oral

anticoagulants or replacement therapies and predicting thrombosis risk (35). Briefly, to mimic

injury, we added recombinant TF to normal human plasma premixed with a four-point titration

of each TF-specific antibody. Subsequent addition of Ca2+

and a fluorescent thrombin substrate

triggered TF-dependent coagulation and thrombin generation, respectively (Fig. 2C and 2D,

Supplementary Table S3). At the 100 nM antibody concentration, 1F, 29E, 39A, 54E diminished

the peak IIa concentration by 92, 76, 91 and 70 %, respectively (Fig. 2D). Similarly, 100 nM of

5G9 and TF-011 inhibited peak IIa concentration by 92 % and 91 %, respectively (Fig. 2D,

Supplementary Table S3). Severely reduced thrombin generation in the presence of the two

highest concentrations of 1F, 39A, 5G9 and TF-011 hampered endogenous thrombin generation

(ETP) calculations and increased time to Peak IIa/thrombin generation (ttPeak) by at least 284 %

at 50 nM (Supplementary Table S3). On the other hand, antibodies from group 25 did not impact

the peak IIa concentration or ttPeak by more than single-digit percentage points (Fig. 2D and

Supplementary Table S3). Group 43 antibodies and 10H10 exhibited mild interference with the

peak IIa concentration: 100 nM of 43B1, 43D7, 43Ea and 10H10 reduced the peak IIa

concentration by 33, 44, 13 and 34 %, respectively (Fig. 2D). In addition, 100 nM of 43B1, 43D7

and 10H10 showed at least a 29 % increase in ttPeak (Supplementary Table S3). However, the

observed decline in peak IIa concentration and delayed ttPeak for group 43 antibodies and

10H10 did not result in more than a 10 % decline in the ETP (Supplementary Table S3). Overall,

group 25 antibodies were completely inert in the penultimate step of the coagulation cascade

when all three TGA parameters (ETP, Peak IIa concentration and ttPeak) were taken into

consideration.

To establish whether the TF-specific antibodies interfered with binding of FVII to TF,

competition between FVII and the TF-specific antibodies was evaluated. While the FX

conversion–inert antibodies did not compete with FVII binding by more than 20 %, all the FX

conversion–inhibitory antibodies (1F, 29E, 39A and 54E) competed with FVII binding (Fig. 2E).

The previously described TF-specific antibodies TF-011, 5G9 and 10H10 (17,34) were also

tested. TF-011 effectively competed with FVII, whereas 5G9 and 10H10 showed less than 25 %

and 10 % competition at the highest concentration of antibody, respectively (Supplementary Fig.

S1D). Thus, the FVII-competing antibodies 1F, 29E, 39A, 54E and TF-011 likely impaired FXa

and thrombin generation by inhibiting the formation of an active TF:FVIIa complex. The results

are also consistent with previous reports that indicated that 5G9 predominantly competes with

substrate FX binding (36,37), resulting in the observed inhibition of FX conversion and thrombin

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 20, 2018; DOI: 10.1158/1535-7163.MCT-18-0471

Page 11: Treating Tissue Factor-Positive Cancers With Antibody-Drug ......Aug 18, 2018  · maytansinoid DM1, had anticancer activity, but also inhibited coagulation (19). The TF-specific antibody

11

generation, while 10H10 inhibits TF-FVIIa mediated signaling without substantially affecting

binding of FVIIa to TF (38).

The dichotomy in absence or presence of FVII competition suggested the TF-specific

antibodies bound distinct epitopes on TF. Competition experiments using antibody pairs showed

that antibodies from groups 25 and 43 bound overlapping epitopes, while the antibodies from

groups 1, 29, 39 and 54 bound epitopes that did not overlap with the epitopes of groups 25 and

43 (Supplementary Fig. 2A–C). Furthermore, TF-011 did not compete with 25A3 and 43D7, but

did compete with 39A (Supplementary Fig. 2A–C). However, possible overlap between 5G9’s

epitope and those of group 25 and 43 antibodies illustrates the utility of high-resolution epitope

mapping to further understand how antibodies from group 25 and 43 avoid interference with the

coagulation cascade (Supplementary Fig. 2A-B).

Anti-TF antibody internalization and toxin delivery

To verify the TF-specific antibodies can mediate internalization and toxin delivery into cells, we

conducted a cell-based assay in which the TF-specific antibodies were complexed with or

without a Fab fragment conjugated to monomethyl auristatin F (Fab:MMAF). Because

Fab:MMAF binds the Fc region of the TF-specific antibodies, cellular uptake of these complexes

can trigger cell death, as previously shown in Fig. 1. While the TF-specific antibodies alone had

no impact on cell viability in three-day cultures of TF-positive A431 cells (Supplementary Fig.

S3A), the TF-specific antibodies in complex with Fab:MMAF showed dose-dependent cell

killing with IC50 values ranging between 0.07 and 0.14 nM (Supplementary Fig. S3B).

Cellular uptake was corroborated with fluorescently labeled TF-specific antibodies. In a

quantitative assay based on internalized fluorescence and quenched surface-fluorescence, the TF-

specific antibodies showed between 28 and 37 % internalization after a 4 h incubation

(Supplementary Fig. S3C).

We then evaluated direct toxin delivery to cells by generating ADCs. MMAE was

conjugated to cysteine residues on the TF-specific antibodies via a cleavable maleimide-

containing linker with an average DAR of 3 to 4. The conjugation process did not alter the cell-

binding properties of the TF-specific antibody moiety of the ADC (Supplementary Fig. S3D).

Titrations of the TF-specific ADCs were added to A431 cells, with either a 72 h incubation or a 4

h incubation followed by removal of excess ADC and culture for another 68 h (Fig. 3A). Both

treatments resulted in efficacious cell killing, with a 2.4 to 4.7-fold increase in IC50 when excess

ADC was removed from the culture after the 4 h incubation compared to the 72 h incubation

(Fig. 3A). Removal of excess 25A3 and 39A ADC had the smallest impact on IC50, with a 2.7

and 2.4-fold increase from 0.07 and 0.05 nM, respectively. The antibody internalization and

short-term ADC exposure data were in agreement with previous reports in which TF and anti-TF

antibodies undergo substantial cellular internalization (15,16).

Tumor angiogenesis, growth and metastasis have been reported to be regulated by FVIIa-

dependent TF signaling (14). To understand whether FVIIa interfered with the activity of the TF-

specific ADC, we treated A431 cells for 4 h with the TF-specific ADCs in the absence or

presence of FVIIa and measured cell viability 68 h later. While the ADCs that competed with

FVII (29E, 39A, 54E and TF-011) were negatively affected by the presence of FVIIa by at least

2.3-fold, the ADCs that did not compete with FVII (group 25 and 43 antibodies) were equally

efficacious in the absence or presence of FVIIa (Supplementary Fig. S4A).

The activity of the TF-specific ADCs was confirmed on additional cancer cell lines with

different levels of surface-exposed TF. The level of surface TF ranged from 1.9x105 to 5.7x10

5

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 20, 2018; DOI: 10.1158/1535-7163.MCT-18-0471

Page 12: Treating Tissue Factor-Positive Cancers With Antibody-Drug ......Aug 18, 2018  · maytansinoid DM1, had anticancer activity, but also inhibited coagulation (19). The TF-specific antibody

12

copies in A431, MDA-MB-231 and HPAF-II cells (Supplementary Fig. S4B). HCT-116 cells

expressed 2.2x104 copies of surface TF, and CHO cells were used as a negative control for

human TF (Supplementary Fig. S4B). While the TF-specific ADCs were also active on MDA-

MB-231 and HPAF-II cells (Fig. 3B and 3C), the ADCs were less efficacious on HCT-116 cells,

with some activity at the highest concentration and no reportable IC50 value (Supplementary Fig.

S4C). The TF-specific ADCs did not affect the viability of the CHO cultures (Supplementary

Fig. S4D). When ranking the cell-killing efficacy of the ADCs on A431, HPAF-II and MDA-

MB-231 cells, the top four ADCs in descending order were 39A, 29E, 25G1 and 25A3

(Supplementary Table S4A). When A431 cells were incubated for 4 h with the TF-specific

ADCs followed by a washout, the top four ADCs in descending order were 39A, 25G1, 25A3

and 29E (Supplementary Table S4B). Thus, the top 2 ranking ADCs with no impact on

coagulation were 25G1 and 25A3.

To illustrate the mechanism of action of the MMAE-based ADCs, we evaluated the

disruption of the intracellular microtubule network in response to treatment. While the isotype

control ADC did not affect the microtubule network, the 25A3 ADC disrupted the microtubule

network effectively in A431 and HPAF-II cells (Fig. 3D).

In response to injury, inflammatory and angiogenic factors transiently increase

expression of surface TF in the vasculature (39). We mimicked the transient upregulation of TF

in cell culture by treating human umbilical vein endothelial cells (HUVECs) with a combination

of inflammatory cytokines. Surface TF levels increased from 2.4x103 copies in the absence of

inflammatory cytokines to 1.2x104 copies after 6 h of cytokine treatment (Supplementary Fig.

S5A). In agreement with the transient nature of TF upregulation, surface TF was ~3-fold lower

after 20 h of cytokine treatment relative to 6 h of treatment. Cytokine-treated endothelial cell

cultures appeared resistant to anti-TF ADCs, as viability was unaffected in a 4-day viability

assay (Supplementary Fig. S5B). To further understand the apparent resistance of endothelial

cells to anti-TF ADCs, cell cycle progression was evaluated 24 h after addition of the cytokines

and TF-specific ADCs. While the TF-specific ADCs induced an arrest at the G2/M phase of the

cell cycle in HCT-116 cells, the ADCs did not impact cell cycle progression in HUVECs treated

with or without inflammatory cytokines (Supplementary Fig. S5C and S5D). An increase in

phosphorylated histone H3 was observed in both HCT-116 cells and HUVECs upon treatment

with unconjugated MMAE, indicating that the resistance in endothelial cells is specific for the

MMAE-based ADC (Supplementary Fig. S5E).

Additional functional properties of TF-specific antibodies

Signaling by tumor cell–expressed TF:FVIIa complex induces proangiogenic and immune-

modulating cytokines (14). To determine whether the TF-specific antibodies had an impact on

TF signaling, cytokine secretion and Erk phosphorylation were evaluated in cell cultures treated

with FVIIa. All the TF-specific antibodies inhibited IL-8 and GM-CSF protein levels with an

IC50 value ranging between 1 and 2.7 nM (Fig. 4A). Erk phosphorylation was ablated by

pretreatment with 1F, 39A and 54E (fold induction between 0.8 and 1.2) and attenuated by 29E

and the members of groups 25 and 43 (induction between 2.0 and 3.4) (Supplementary Fig. S6).

Both the TF-specific antibodies and ADCs can exert antibody-dependent cellular

cytotoxicity (ADCC) via the IgG1 Fc domain of the antibody. In an ADCC assay with TF-

positive A431 cells in which primary NK cells were replaced with a Jurkat cell line stably

expressing human FcγRIIIa V158 and NFAT-induced luciferase, all the TF-specific antibodies

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 20, 2018; DOI: 10.1158/1535-7163.MCT-18-0471

Page 13: Treating Tissue Factor-Positive Cancers With Antibody-Drug ......Aug 18, 2018  · maytansinoid DM1, had anticancer activity, but also inhibited coagulation (19). The TF-specific antibody

13

and ADCs exerted induction of luciferase-dependent luminescence with EC50 values ranging

between 0.18 and 0.43 nM (Fig. 4B).

ADCs against tissue factor exhibit antitumor activity in cell line– and patient-derived

xenograft models

The in vivo efficacy of the TF-specific ADCs was evaluated in multiple cell-line (CDX) and

patient-derived xenografts (PDX). The target-specific ADCs were efficacious in the HPAF-II

xenograft model using various dosing regimens (Fig. 5). In the first HPAF-II study, we

compared TF-011 with two representative clones from the two groups that did not impact FX

conversion (i.e. 25A and 43Ea). Tumor-bearing mice were treated with 5 mg/kg of ADC on day

1, 8 and 15 after randomization. Twenty-one days after randomization the efficacy of the 25A,

43Ea and TF-011 ADCs was equivalent, with tumor growth inhibition ranging between 131 and

136 % (Fig. 5A). In the second HPAF-II study, the highest affinity antibody that affected

coagulation (i.e. 39A) and six antibodies with varying affinities from groups 25 and 43 (i.e. 25A,

25A3, 25G1, 43Ea, 43B1 and 43D7) were equally efficacious when dosed twice at 2 mg/kg (Fig.

5B). Tumor growth inhibition for the TF-specific ADCs ranged between 129 and 139 % on day

21, and 6 to 9 out of 10 animals per treatment arm were classified as tumor-free survivors at the

end of the study (Fig. 5B).

In the MDA-MB-231 xenograft model, the ADCs were administered on day 1 and 8 post-

randomization at 4 or 2 mg/kg (Fig. 5C and 5D). All the TF-specific ADCs were active at 4

mg/kg, with tumor growth inhibition ranging between 69 and 100 %, and a significant difference

in mean tumor volume for each TF-specific ADC compared to the vehicle control arm (Fig. 5C).

While a notable difference was observed in mean tumor volume between 25G1 and the other TF-

specific ADCs, it was not statistically significant (P > 0.05, Fig. 5C). At 2 mg/kg of ADC, all the

TF-specific antibodies showed suboptimal activity with varying degrees of significance in mean

tumor volume compared to the vehicle control arm. 25A3, 39A and 43B1 showed the greatest

degree of significance in mean tumor volume compared to the vehicle control arm (P < 1x10-4

)

(Fig. 5D). The difference in mean tumor volume between 39A and the other antibodies was only

significant for the comparison between 39A and 43Ea (P < 0.05, Fig. 5D).

Next, we benchmarked the activity of the 25A and 43Ea ADCs against tisotumab vedotin

in three PDX models (Fig. 6). PDX tumor models reflect human tumor cell heterogeneity to a

greater degree than cancer cell lines and CDX models (40). After profiling TF expression in

PDX tumor models from multiple cancer indications—colorectal, esophageal, gastric, head and

neck, lung, melanoma, ovarian, pancreatic, and sarcoma—we prioritized PDX models from three

indications with varying levels of expression. While the squamous cell carcinoma of the head

and neck (SCCHN) and ovarian adenocarcinoma PDX had H-scores of 250 and 220,

respectively, the gastric adenocarcinoma PDX had an H-score of 155 (Fig. 6). Upon

randomization of tumor-bearing mice, treatment occurred on a weekly basis either twice or three

times with the dose ranging between 2.5 and 5 mg/kg (Fig. 6). In all the PDX models a

significant reduction in mean tumor volume was observed for each TF-specific ADC compared

to the isotype control arm (P < 1x10-4

), with no significant difference between the various TF-

specific ADCs (P > 0.05, Fig. 6). In the head and neck and ovarian PDX model, the number of

complete responders and tumor-free survivors did not exceed 2 out of 10 animals at the end of

the study in any of the treatment groups (Fig. 6A and 6B). However, in the gastric PDX the 25A

treatment arm had 2 partial responders, 2 complete responders and 3 tumor-free survivors, and

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 20, 2018; DOI: 10.1158/1535-7163.MCT-18-0471

Page 14: Treating Tissue Factor-Positive Cancers With Antibody-Drug ......Aug 18, 2018  · maytansinoid DM1, had anticancer activity, but also inhibited coagulation (19). The TF-specific antibody

14

the TF-011 arm contained 1 complete responder and 5 tumor-free survivors at the end of the

study (Fig. 6C).

To understand whether the activity of the ADC was solely attributable to the delivery of

toxin by the anti-TF antibody to the xenograft, the activity of a subset of naked antibodies was

evaluated in the HPAF-II xenograft model. Unlike the 25A, 25A3 and 43Ea ADCs, the naked

antibody equivalents lacked substantial anticancer activity when dosed twice at 10 mg/kg (Fig.

6D).

Overall, the coagulation-inert and -inhibitory ADCs were equally efficacious in CDX and

PDX models. A member of group 25 has been selected for further preclinical and clinical

development as an ADC.

Discussion

Here, coagulation-inert TF-specific ADCs with potent anticancer activity were developed. First,

a panel of anti-TF antibodies was evaluated for perturbation of coagulation and cellular

internalization. To ensure lack of coagulation interference was not due to poor binding affinity,

the antibody panel underwent affinity maturation and coagulation-inert antibodies were

identified. Affinity maturation also improved the anticancer efficacy of the coagulation-inert

antibodies as ADCs. The prioritized coagulation-inert ADCs from antibody family 25 induced

tumor growth inhibition in TF-positive solid tumor xenografts at levels comparable to tisotumab

vedotin, a clinical stage ADC (17,20,21).

Tisotumab vedotin validated TF as a promising clinical-stage target (20,21). In a phase

I/II dose-escalation study of tisotumab vedotin, fourteen out of twenty-seven patients achieved

stable disease or presented with a better outcome (20,21). While tisotumab vedotin showed

anticancer activity, some of the adverse events could be attributed to the biological activity of

TF-011, the antibody used for the development of tisotumab vedotin. In the current study, TF-

011 competed with FVII, partially inhibited the conversion of FX into FXa, and severely

impacted thrombin generation at clinically relevant concentrations. In the phase I dose escalation

cohorts from 0.3 to 2.2 mg/kg of tisotumab vedotin, the Cmax concentrations ranged from 32 up

to 370 nM (21). One of the most common adverse events recorded in the trial was epistaxis, with

an incidence of 48 % (20,21). We postulate that these nosebleeds were triggered by TF-011-

induced perturbation of coagulation. In addition, one SCCHN patient died from tumor-related

bleeding at 0.6 mg/kg of ADC. While this death could be related to the cancer itself, the

frequency of epistaxis suggests that tisotumab vedotin may affect hemostasis.

Aside from mediating hemostasis, TF can also trigger thrombosis (11). Another TF-

specific antibody, Sunol-cH36 underwent clinical development as an inhibitor of coronary

thrombosis. In this naked antibody trial, dose-dependent spontaneous minor bleeding was

observed (41). Sunol-cH36 had a putative mechanism of action similar to that of 5G9, with both

antibodies inhibiting binding of FX to the TF:FVIIa complex (41,42). In the current study, 5G9

inhibited conversion of FX into FXa, and also interfered with thrombin generation. Therefore,

the FX conversion and thrombin generation assays described herein might be predictive assays

for clotting-related adverse events in the clinic. Indeed, the TGA is considered a bridge between

in vivo and ex vivo coagulation, and an automated version can be used in the clinic on a large

number of patients (35). We prioritized group 25 for further preclinical and clinical development,

because none of the TGA parameters (i.e., ETP, Peak IIa concentration and ttPeak) were affected

by group 25 antibodies. The automated TGA could be used during clinical development to

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 20, 2018; DOI: 10.1158/1535-7163.MCT-18-0471

Page 15: Treating Tissue Factor-Positive Cancers With Antibody-Drug ......Aug 18, 2018  · maytansinoid DM1, had anticancer activity, but also inhibited coagulation (19). The TF-specific antibody

15

understand whether the TGA parameters are also unaffected after dosing patients with a group 25

ADC.

In a population-based case-control study, the baseline risk of thrombosis in cancer was

four-fold compared to a healthy population, with an additional seven-fold increase after

treatment with chemotherapy (43). In cancer indications with a higher incidence of venous

thromboembolism (VTE), such as pancreatic and brain cancer, a low dose of an antibody-based

inhibitor of coagulation might attenuate thrombotic events (44). Indeed, treatment of human

patients with tisotumab vedotin or Sunol-cH36 did not augment VTE. Furthermore, in light of

the coagulation-inert properties of antibody family 25, we predict the clinical-stage ADC from

this family not to elicit thrombosis.

The anticancer activity of the anti-TF ADCs was attributable to the delivery of the toxin

to the TF-positive xenografts. While the TF-specific ADCs were efficacious in various xenograft

models, the isotype control ADC and a high dose of naked anti-TF antibody showed little to no

impact on tumor growth. ADC-dependent toxin delivery was consistent with the observation that

cancer cells internalized about a third of fluorescently labeled anti-TF antibody over a 4 h time

course. Because TF undergoes such rapid internalization, the anticancer activity of the anti-TF

ADC described herein will likely not improve with a biparatopic anti-TF ADC or a bispecific

ADC that bridges TF with another surface antigen that also undergoes rapid internalization, like

the prolactin receptor or CD63 (45,46).

In the current study a high dose of naked antibody did not significantly impact tumor

growth. However, co-injection of a cancer cell line with 10H10 resulted in tumor growth

inhibition (34). In this prophylactic treatment setting, tumor growth inhibition was attributed to

10H10-dependent inhibition of TF:FVIIa signaling. Like 10H10, all the antibodies isolated from

the screening campaign inhibited TF:FVIIa signaling by downmodulating Erk phosphorylation

and cytokine production. Importantly, in the context of an ADC, the antibody’s ability to inhibit

TF:FVIIa signaling could be masked by the anticancer activity of the internalized toxin.

The anti-signaling activity of the anti-TF antibody might be more relevant in cell

populations resistant to MMAE-based ADCs. While the anti-TF ADCs affected cell cycle

progression and viability of cancer cells, the ADCs did not alter cell cycle progression and

viability of endothelial cells, consistent with previous reports in which endothelial cells were

resistant to MMAE-based ADCs (47). This resistance was likely unrelated to TF copy number,

as the difference in TF surface copy number between the cytokine-treated endothelial cells and

the MMAE-based ADC–sensitive HCT-116 cancer cells was only two-fold. However, the lack of

activity of the ADCs on endothelial cell cultures does not preclude a safe toxicity profile in

normal tissue.

Stabilization of disease with tisotumab vedotin validated TF as an anti-cancer target for

an ADC. To enable appropriate benchmarking against tisotumab vedotin, the prototypic linker-

toxin vc-MMAE was used for all the in vivo efficacy studies. In the phase I/II study of tisotumab

vedotin, the adverse events of peripheral neuropathy and neutropenia were consistent with

MMAE-based ADCs, with a maximum tolerated dose (MTD) between 2–3 mg/kg (5). With the

advent of safer linker-payloads such as XMT-1267 (48), we anticipate lack of MMAE-associated

toxicities for the clinical-stage group 25 ADC.

Finally, the antibodies described herein could also be prioritized for the treatment of other

diseases associated with upregulation of membranous TF expression, such as macular

degeneration (49). A FVII-Fc fusion protein that binds TF reduces the size of choroid

neovascular (CNV) lesions in various animal models (50,51). With the binding affinities of a

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 20, 2018; DOI: 10.1158/1535-7163.MCT-18-0471

Page 16: Treating Tissue Factor-Positive Cancers With Antibody-Drug ......Aug 18, 2018  · maytansinoid DM1, had anticancer activity, but also inhibited coagulation (19). The TF-specific antibody

16

subset of the anti-TF antibodies below the affinity of FVII for TF (52), some of the antibodies

could be more efficacious than the FVII-Fc fusion protein. A subset of the anti-TF antibodies

will be evaluated in various CNV animal models.

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 20, 2018; DOI: 10.1158/1535-7163.MCT-18-0471

Page 17: Treating Tissue Factor-Positive Cancers With Antibody-Drug ......Aug 18, 2018  · maytansinoid DM1, had anticancer activity, but also inhibited coagulation (19). The TF-specific antibody

17

Acknowledgments

The authors thank Ashley McLaren and Justin Meyette for acquisition of the TGA data and

Helen Mao for generation of the ADCs. This work was supported by Iconic Therapeutics, Inc., a

privately held and financed company.

References

1. Kantarjian HM, DeAngelo DJ, Stelljes M, Martinelli G, Liedtke M, Stock W, et al. Inotuzumab

Ozogamicin versus Standard Therapy for Acute Lymphoblastic Leukemia. N Engl J Med 2016;375(8):740-

53 doi 10.1056/NEJMoa1509277.

2. Lambert JM, Morris CQ. Antibody-Drug Conjugates (ADCs) for Personalized Treatment of Solid Tumors:

A Review. Adv Ther 2017;34(5):1015-35 doi 10.1007/s12325-017-0519-6.

3. Senter PD, Sievers EL. The discovery and development of brentuximab vedotin for use in relapsed

Hodgkin lymphoma and systemic anaplastic large cell lymphoma. Nat Biotechnol 2012;30(7):631-7 doi

10.1038/nbt.2289.

4. Beck A, Goetsch L, Dumontet C, Corvaia N. Strategies and challenges for the next generation of antibody-

drug conjugates. Nat Rev Drug Discov 2017;16(5):315-37 doi 10.1038/nrd.2016.268.

5. de Goeij BE, Lambert JM. New developments for antibody-drug conjugate-based therapeutic approaches.

Curr Opin Immunol 2016;40:14-23 doi 10.1016/j.coi.2016.02.008.

6. Donaghy H. Effects of antibody, drug and linker on the preclinical and clinical toxicities of antibody-drug

conjugates. MAbs 2016;8(4):659-71 doi 10.1080/19420862.2016.1156829.

7. Diamantis N, Banerji U. Antibody-drug conjugates--an emerging class of cancer treatment. Br J Cancer

2016;114(4):362-7 doi 10.1038/bjc.2015.435.

8. Sau S, Alsaab HO, Kashaw SK, Tatiparti K, Iyer AK. Advances in antibody-drug conjugates: A new era of

targeted cancer therapy. Drug Discov Today 2017;22(10):1547-56 doi 10.1016/j.drudis.2017.05.011.

9. Phillips AC, Boghaert ER, Vaidya KS, Falls HD, Mitten MJ, DeVries PJ, et al. Characterization of ABBV-

221, a Tumor-Selective EGFR-Targeting Antibody Drug Conjugate. Mol Cancer Ther 2018;17(4):795-805

doi 10.1158/1535-7163.MCT-17-0710.

10. Bradbury AR, Sidhu S, Dubel S, McCafferty J. Beyond natural antibodies: the power of in vitro display

technologies. Nat Biotechnol 2011;29(3):245-54 doi 10.1038/nbt.1791.

11. Grover SP, Mackman N. Tissue Factor: An Essential Mediator of Hemostasis and Trigger of Thrombosis.

Arterioscler Thromb Vasc Biol 2018 doi 10.1161/ATVBAHA.117.309846.

12. Drake TA, Morrissey JH, Edgington TS. Selective cellular expression of tissue factor in human tissues.

Implications for disorders of hemostasis and thrombosis. Am J Pathol 1989;134(5):1087-97.

13. van den Berg YW, Osanto S, Reitsma PH, Versteeg HH. The relationship between tissue factor and cancer

progression: insights from bench and bedside. Blood 2012;119(4):924-32 doi 10.1182/blood-2011-06-

317685.

14. Ruf W, Disse J, Carneiro-Lobo TC, Yokota N, Schaffner F. Tissue factor and cell signalling in cancer

progression and thrombosis. J Thromb Haemost 2011;9 Suppl 1:306-15 doi 10.1111/j.1538-

7836.2011.04318.x.

15. de Goeij BE, Satijn D, Freitag CM, Wubbolts R, Bleeker WK, Khasanov A, et al. High turnover of tissue

factor enables efficient intracellular delivery of antibody-drug conjugates. Mol Cancer Ther

2015;14(5):1130-40 doi 10.1158/1535-7163.MCT-14-0798.

16. Mandal SK, Pendurthi UR, Rao LV. Cellular localization and trafficking of tissue factor. Blood

2006;107(12):4746-53 doi 10.1182/blood-2005-11-4674.

17. Breij EC, de Goeij BE, Verploegen S, Schuurhuis DH, Amirkhosravi A, Francis J, et al. An antibody-drug

conjugate that targets tissue factor exhibits potent therapeutic activity against a broad range of solid tumors.

Cancer Res 2014;74(4):1214-26 doi 10.1158/0008-5472.CAN-13-2440.

18. Koga Y, Manabe S, Aihara Y, Sato R, Tsumura R, Iwafuji H, et al. Antitumor effect of antitissue factor

antibody-MMAE conjugate in human pancreatic tumor xenografts. Int J Cancer 2015;137(6):1457-66 doi

10.1002/ijc.29492.

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 20, 2018; DOI: 10.1158/1535-7163.MCT-18-0471

Page 18: Treating Tissue Factor-Positive Cancers With Antibody-Drug ......Aug 18, 2018  · maytansinoid DM1, had anticancer activity, but also inhibited coagulation (19). The TF-specific antibody

18

19. Zhang X, Li Q, Zhao H, Ma L, Meng T, Qian J, et al. Pathological expression of tissue factor confers

promising antitumor response to a novel therapeutic antibody SC1 in triple negative breast cancer and

pancreatic adenocarcinoma. Oncotarget 2017;8(35):59086-102 doi 10.18632/oncotarget.19175.

20. Vergote I, Dean E, Lassen U, de Bono J, Drew Y, Machiels J, et al. A Phase IIa Study of Tisotumab

Vedotin (HuMax®-TF-ADC) in Patients With Relapsed, Recurrent and/or Metastatic Cervical Cancer.

Annals of Oncology 2017;28(suppl_5):v330-v54. 10.1093/annonc/mdx372.

21. Chenard-Poirier M, Hong DS, Coleman R, de Bono J, Mau-Sorensen M, Collins D, et al. A Phase I/II

Safety Study of Tisotumab Vedotin (HuMax®-TF-ADC) in Patients With Solid Tumors. Annals of

Oncology 2017;28 (suppl_5):v403-v27. 10.1093/annonc/mdx376.

22. Saito Y, Hashimoto Y, Kuroda J, Yasunaga M, Koga Y, Takahashi A, et al. The inhibition of pancreatic

cancer invasion-metastasis cascade in both cellular signal and blood coagulation cascade of tissue factor by

its neutralisation antibody. Eur J Cancer 2011;47(14):2230-9 doi 10.1016/j.ejca.2011.04.028.

23. Rouha H, Badarau A, Visram ZC, Battles MB, Prinz B, Magyarics Z, et al. Five birds, one stone:

neutralization of alpha-hemolysin and 4 bi-component leukocidins of Staphylococcus aureus with a single

human monoclonal antibody. MAbs 2015;7(1):243-54 doi 10.4161/19420862.2014.985132.

24. Xu Y, Roach W, Sun T, Jain T, Prinz B, Yu TY, et al. Addressing polyspecificity of antibodies selected

from an in vitro yeast presentation system: a FACS-based, high-throughput selection and analytical tool.

Protein Eng Des Sel 2013;26(10):663-70 doi 10.1093/protein/gzt047.

25. Larsen KS, Ostergaard H, Olsen OH, Bjelke JR, Ruf W, Petersen LC. Engineering of substrate selectivity

for tissue factor.factor VIIa complex signaling through protease-activated receptor 2. J Biol Chem

2010;285(26):19959-66 doi 10.1074/jbc.M110.101030.

26. Kim SY, Theunissen JW, Balibalos J, Liao-Chan S, Babcock MC, Wong T, et al. A novel antibody-drug

conjugate targeting SAIL for the treatment of hematologic malignancies. Blood Cancer J 2015;5:e316 doi

10.1038/bcj.2015.39.

27. Liao-Chan S, Daine-Matsuoka B, Heald N, Wong T, Lin T, Cai AG, et al. Quantitative assessment of

antibody internalization with novel monoclonal antibodies against Alexa fluorophores. PLoS One

2015;10(4):e0124708 doi 10.1371/journal.pone.0124708.

28. Estep P, Reid F, Nauman C, Liu Y, Sun T, Sun J, et al. High throughput solution-based measurement of

antibody-antigen affinity and epitope binning. MAbs 2013;5(2):270-8 doi 10.4161/mabs.23049.

29. Samama MM, Mendell J, Guinet C, Le Flem L, Kunitada S. In vitro study of the anticoagulant effects of

edoxaban and its effect on thrombin generation in comparison to fondaparinux. Thromb Res

2012;129(4):e77-82 doi 10.1016/j.thromres.2011.07.026.

30. Behrens CR, Ha EH, Chinn LL, Bowers S, Probst G, Fitch-Bruhns M, et al. Antibody-Drug Conjugates

(ADCs) Derived from Interchain Cysteine Cross-Linking Demonstrate Improved Homogeneity and Other

Pharmacological Properties over Conventional Heterogeneous ADCs. Mol Pharm 2015;12(11):3986-98 doi

10.1021/acs.molpharmaceut.5b00432.

31. Doronina SO, Mendelsohn BA, Bovee TD, Cerveny CG, Alley SC, Meyer DL, et al. Enhanced activity of

monomethylauristatin F through monoclonal antibody delivery: effects of linker technology on efficacy and

toxicity. Bioconjug Chem 2006;17(1):114-24 doi 10.1021/bc0502917.

32. Theunissen JW, Petrini JH. Methods for studying the cellular response to DNA damage: influence of the

Mre11 complex on chromosome metabolism. Methods Enzymol 2006;409:251-84 doi 10.1016/S0076-

6879(05)09015-4.

33. Hjortoe GM, Petersen LC, Albrektsen T, Sorensen BB, Norby PL, Mandal SK, et al. Tissue factor-factor

VIIa-specific up-regulation of IL-8 expression in MDA-MB-231 cells is mediated by PAR-2 and results in

increased cell migration. Blood 2004;103(8):3029-37 doi 10.1182/blood-2003-10-3417.

34. Versteeg HH, Schaffner F, Kerver M, Petersen HH, Ahamed J, Felding-Habermann B, et al. Inhibition of

tissue factor signaling suppresses tumor growth. Blood 2008;111(1):190-9 doi 10.1182/blood-2007-07-

101048.

35. Tripodi A. Thrombin Generation Assay and Its Application in the Clinical Laboratory. Clin Chem

2016;62(5):699-707 doi 10.1373/clinchem.2015.248625.

36. Huang M, Syed R, Stura EA, Stone MJ, Stefanko RS, Ruf W, et al. The mechanism of an inhibitory

antibody on TF-initiated blood coagulation revealed by the crystal structures of human tissue factor, Fab

5G9 and TF.G9 complex. J Mol Biol 1998;275(5):873-94.

37. Ruf W, Rehemtulla A, Edgington TS. Antibody mapping of tissue factor implicates two different exon-

encoded regions in function. Biochem J 1991;278 ( Pt 3):729-33.

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 20, 2018; DOI: 10.1158/1535-7163.MCT-18-0471

Page 19: Treating Tissue Factor-Positive Cancers With Antibody-Drug ......Aug 18, 2018  · maytansinoid DM1, had anticancer activity, but also inhibited coagulation (19). The TF-specific antibody

19

38. Teplyakov A, Obmolova G, Malia TJ, Wu B, Zhao Y, Taudte S, et al. Crystal structure of tissue factor in

complex with antibody 10H10 reveals the signaling epitope. Cell Signal 2017;36:139-44 doi

10.1016/j.cellsig.2017.05.004.

39. Holy EW, Tanner FC. Tissue factor in cardiovascular disease pathophysiology and pharmacological

intervention. Adv Pharmacol 2010;59:259-92 doi 10.1016/S1054-3589(10)59009-4.

40. Byrne AT, Alferez DG, Amant F, Annibali D, Arribas J, Biankin AV, et al. Interrogating open issues in

cancer precision medicine with patient-derived xenografts. Nat Rev Cancer 2017;17(4):254-68 doi

10.1038/nrc.2016.140.

41. Morrow DA, Murphy SA, McCabe CH, Mackman N, Wong HC, Antman EM. Potent inhibition of

thrombin with a monoclonal antibody against tissue factor (Sunol-cH36): results of the PROXIMATE-

TIMI 27 trial. Eur Heart J 2005;26(7):682-8 doi 10.1093/eurheartj/ehi094.

42. Ngo CV, Picha K, McCabe F, Millar H, Tawadros R, Tam SH, et al. CNTO 859, a humanized anti-tissue

factor monoclonal antibody, is a potent inhibitor of breast cancer metastasis and tumor growth in xenograft

models. Int J Cancer 2007;120(6):1261-7 doi 10.1002/ijc.22426.

43. Heit JA, Silverstein MD, Mohr DN, Petterson TM, O'Fallon WM, Melton LJ, 3rd. Risk factors for deep

vein thrombosis and pulmonary embolism: a population-based case-control study. Arch Intern Med

2000;160(6):809-15.

44. Jiao JA, Kelly AB, Marzec UM, Nieves E, Acevedo J, Burkhardt M, et al. Inhibition of acute vascular

thrombosis in chimpanzees by an anti-human tissue factor antibody targeting the factor X binding site.

Thromb Haemost 2010;103(1):224-33 doi 10.1160/TH09-06-0400.

45. Andreev J, Thambi N, Perez Bay AE, Delfino F, Martin J, Kelly MP, et al. Bispecific Antibodies and

Antibody-Drug Conjugates (ADCs) Bridging HER2 and Prolactin Receptor Improve Efficacy of HER2

ADCs. Mol Cancer Ther 2017;16(4):681-93 doi 10.1158/1535-7163.MCT-16-0658.

46. de Goeij BE, Vink T, Ten Napel H, Breij EC, Satijn D, Wubbolts R, et al. Efficient Payload Delivery by a

Bispecific Antibody-Drug Conjugate Targeting HER2 and CD63. Mol Cancer Ther 2016;15(11):2688-97

doi 10.1158/1535-7163.MCT-16-0364.

47. Seaman S, Zhu Z, Saha S, Zhang XM, Yang MY, Hilton MB, et al. Eradication of Tumors through

Simultaneous Ablation of CD276/B7-H3-Positive Tumor Cells and Tumor Vasculature. Cancer Cell

2017;31(4):501-15 e8 doi 10.1016/j.ccell.2017.03.005.

48. Trail PA, Dubowchik GM, Lowinger TB. Antibody drug conjugates for treatment of breast cancer: Novel

targets and diverse approaches in ADC design. Pharmacol Ther 2018;181:126-42 doi

10.1016/j.pharmthera.2017.07.013.

49. Cho Y, Cao X, Shen D, Tuo J, Parver LM, Rickles FR, et al. Evidence for enhanced tissue factor

expression in age-related macular degeneration. Lab Invest 2011;91(4):519-26 doi

10.1038/labinvest.2010.184.

50. Bora PS, Hu Z, Tezel TH, Sohn JH, Kang SG, Cruz JM, et al. Immunotherapy for choroidal

neovascularization in a laser-induced mouse model simulating exudative (wet) macular degeneration. Proc

Natl Acad Sci U S A 2003;100(5):2679-84 doi 10.1073/pnas.0438014100.

51. Tran J, Craven C, Wabner K, Schmit J, Matter B, Kompella U, et al. A Pharmacodynamic Analysis of

Choroidal Neovascularization in a Porcine Model Using Three Targeted Drugs. Invest Ophthalmol Vis Sci

2017;58(9):3732-40 doi 10.1167/iovs.16-21230.

52. Ruf W, Edgington TS. Two sites in the tissue factor extracellular domain mediate the recognition of the

ligand factor VIIa. Proc Natl Acad Sci U S A 1991;88(19):8430-4.

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 20, 2018; DOI: 10.1158/1535-7163.MCT-18-0471

Page 20: Treating Tissue Factor-Positive Cancers With Antibody-Drug ......Aug 18, 2018  · maytansinoid DM1, had anticancer activity, but also inhibited coagulation (19). The TF-specific antibody

20

Figure Legends

Fig. 1. Screen to identify anti-TF antibodies that do not impact coagulation and exhibit

cellular internalization

(A) A diverse panel of anti-TF antibodies was evaluated in a binding and coagulation assay. The

relationship between affinity and inhibition of coagulation, expressed as binding affinity of the

anti-TF antibody to human TF (human KD) versus inhibition of FX conversion by the anti-TF

antibody (% FXa generation when cells were pretreated with 50 nM of antibody). Six antibody

groups, each with a different symbol for the parent antibody and its associated affinity-matured

antibody clones, are presented. In the legend, the parent antibody group identifier, the number of

affinity-matured antibodies from each parent antibody, the affinity range within each group, and

the bivariate equation and its associated correlation coefficient that capture the relationship

between binding affinity and inhibition of FX conversion are shown (poor fit: R2 < 0.40).

Average % FXa generation was derived from three independent experiments, with each

experiment conducted in duplicate.

(B) The panel of anti-TF antibodies was also evaluated in a cell killing–based internalization

assay. The relationship between affinity and cell-killing activity in A431 cells, expressed as

human KD (as in panel A) versus the cell-killing IC50 value from a nine-point 3.5-fold titration

starting at 10 nM of the anti-TF antibody complexed with 60 nM of an anti-human Fab

conjugated to the auristatin MMAF. In the legend, the parent antibody group identifier, and the

bivariate equation and its associated correlation coefficient that capture the relationship between

binding affinity and cell-killing IC50 are listed.

(C) Additional affinity maturation of antibody clones 25A, 25G, 43B and 43D did not result in

inhibition of FX conversion. The relationship between antibody affinity and inhibition of

coagulation is presented as in panel A. Average % FXa generation was derived from two

independent experiments, with each experiment performed in duplicate.

(D) Additional affinity maturation of antibody clones 25A, 25G, 43B and 43D improved cell-

killing activity of antibodies in group 25. The relationship between antibody affinity and cell-

killing IC50 is presented as in panel B. The cell-killing IC50 values were derived from a nine-

point 2-fold antibody titration starting at 4.5 nM of anti-TF antibody and 27 nM of Fab:MMAF

in A431 cells.

Fig. 2. In-depth characterization of TF-specific antibodies in binding and coagulation

assays

(A) Binding of anti-TF antibodies to human TF–positive HCT-116 cells. For each anti-TF

antibody, the median fluorescence intensity (MFI) of antibody bound to cells is plotted against

antibody concentration. Reportable cell EC50’s are listed. The 95 % confidence intervals are

reported in Supplementary Table S2. An IgG1 isotype control did not bind cells (no binding, nb).

Representative data from one of three experiments are shown.

(B) TF:FVIIa–dependent conversion of FX into FXa on the cell surface of MDA-MB-231 cells

in the absence or presence of an anti-TF antibody titration. Percent FX conversion to FXa (%

FXa) is plotted against the anti-TF antibody concentration, with % FXa calculated relative to an

antibody-free FX conversion reaction. The standard deviation of the mean % FXa is also

graphed. Representative data from one of two experiments are shown.

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 20, 2018; DOI: 10.1158/1535-7163.MCT-18-0471

Page 21: Treating Tissue Factor-Positive Cancers With Antibody-Drug ......Aug 18, 2018  · maytansinoid DM1, had anticancer activity, but also inhibited coagulation (19). The TF-specific antibody

21

(C) Thrombin generation in the presence of anti-TF antibody. Factor IIa/Thrombin generation in

the absence or presence of 100 nM anti-TF antibody is plotted against time. The mean of a

triplicate data set is shown. The standard deviation of the mean is listed in Supplementary Table

S3. Representative data from one of two experiments are shown.

(D) The peak IIa/thrombin concentration associated with the time course in panel C is shown

against a titration of anti-TF antibody. The mean of a triplicate data set is shown. Other

parameters associated with the real-time IIa/thrombin generation measurement shown in panel C

are summarized in Supplementary Table S3.

(E) FVII binding in the presence of anti-TF antibody. After pre-incubation of MDA-MB-231

cells with a titration of anti-TF antibody, the amount of FVII-Fc that bound cells is plotted as

percent FVII binding (% FVII binding) relative to FVII binding in the presence of an IgG1

isotype control. An IC50 was not reported for antibodies that did not compete with FVII by more

than 20% (no competition, nc). Representative data from one of two experiments are shown.

Fig. 3. In vitro efficacy of anti-TF ADCs

(A) Cell viability of A431 cell cultures after addition of a titration of anti-TF antibody

conjugated directly to MMAE via a cleavable linker (MC-vc-PAB-MMAE). Prior to the cell

viability measurement, the ADC titration was incubated continuously with the cells for 72 h, or

the titration was washed out after a 4 h incubation and the cells were cultured for another 68 h.

The isotype control lacked a reportable IC50 value (not applicable, na). The 95 % confidence

intervals for the IC50’s are reported in Supplementary Table S4.

(B, C) Cell viability of 5-day old cultures of MDA-MB-231 (B) or HPAF-II (C) cells incubated

with a titration of anti-TF antibody conjugated to MC-vc-PAB-MMAE. Isotype control as in

panel A. The 95 % confidence intervals for the IC50’s are reported in Supplementary Table S4A.

(D) Staining of the microtubule network of A431 and HPAF-II cells treated for 20 h with

MMAE-based ADCs. Representative images of tubulin from one of two experiments are shown

in white or in green when overlaid with nuclei in blue. Scale bar, 10 microns.

Fig. 4. Additional functional properties of anti-TF antibodies

(A) FVIIa-dependent TF-signaling in MDA-MB-231 cells pretreated with an anti-TF antibody

titration was evaluated by measuring the concentration of IL-8 and GM-CSF in cell culture

supernatants. The IgG1 isotype control does not inhibit signaling and lacks a reportable IC50 (no

inhibition, ni). Representative data from one of two experiments are shown.

(B) Antibody-dependent cellular cytotoxicity (ADCC) reporter luminescence was evaluated after

a 6 h incubation of the reporter Jurkat cell line with TF-positive A431 cells and a titration of anti-

TF antibody (left panel) or ADC (right panel). The ADCC reporter luminescence EC50 values for

each anti-TF antibody or ADC are listed. An isotype control antibody and ADC lacked activity

(EC50 value not applicable, na).

Fig. 5. In vivo efficacy of anti-TF ADCs in cell line–derived xenograft models

(A, B) Efficacy evaluation of anti-TF ADCs in HPAF-II xenograft model dosed weekly at 5

mg/kg for 3 weeks (A) or 2 mg/kg for 2 weeks (B). The treatment started on day 1 after animals

with a tumor size of ~ 190 mm3 were randomized. The mean tumor volumes (Mean) and tumor

growth inhibition (TGI) percentages on day 21 for all the animals on study are listed. The P-

values for the mean tumor volume comparison between each ADC and the vehicle control arm

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 20, 2018; DOI: 10.1158/1535-7163.MCT-18-0471

Page 22: Treating Tissue Factor-Positive Cancers With Antibody-Drug ......Aug 18, 2018  · maytansinoid DM1, had anticancer activity, but also inhibited coagulation (19). The TF-specific antibody

22

are also listed. At the end of the study (day 59 for (A) and day 39 for (B)), the number of PR, CR

and TFS animals were counted.

(C, D) Efficacy evaluation of anti-TF ADCs in MDA-MB-231 xenograft model dosed weekly at

4 mg/kg for 2 weeks (C) or 2 mg/kg for 2 weeks (D). The treatment started on day 1 after

animals with a tumor size of ~ 160 mm3 were randomized. The mean tumor volume and tumor

growth inhibition on day 25 (C) and day 27 (D) for the animals on study are reported. The P-

values for the mean tumor volume comparison between each ADC and the vehicle control arm

are also listed. At the end of the study, the number of PR, CR and TFS animals were counted.

Fig. 6. In vivo efficacy of anti-TF ADCs in patient-derived xenograft (PDX) models and

lack of anticancer activity of unconjugated anti-TF antibodies.

Efficacy evaluation of anti-TF ADCs dosed as indicated in PDX models of a head and neck

carcinoma (A), an ovarian carcinoma (B), and a gastric adenocarcinoma (C).

Immunohistochemistry (IHC) analysis indicated TF expression in each PDX, with an H-score of

250 for the head and neck carcinoma (A), 220 for the ovarian carcinoma (B) and 155 for the

gastric adenocarcinoma (C). (D) The lack of anticancer activity of unconjugated anti-TF

antibodies was evaluated in the HPAF-II xenograft model. ADC or naked antibody treatment

started on day 1 after animals with a tumor size of ~210 mm3 (A), ~190 mm

3 (B), ~140 mm

3 (C),

and ~190 mm3 (D) were randomized. Mean tumor volume, tumor growth inhibition and P-value

calculations, and PR/CR/TFS enumerations were performed as in Fig. 5.

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 20, 2018; DOI: 10.1158/1535-7163.MCT-18-0471

Page 23: Treating Tissue Factor-Positive Cancers With Antibody-Drug ......Aug 18, 2018  · maytansinoid DM1, had anticancer activity, but also inhibited coagulation (19). The TF-specific antibody

Figure 1

A

C

B

D

Human KD (nM) Human K

D (nM)

1

25

29

39

43

54

Parent

antibody

group

Number of

affinity matured

antibodies

Bivariate

equation

Affinity

range (nM)R2

1 10 y = 34x0.30 0.96

25 12 poor fit

29 6 y = 46x0.12 0.62

39 4 y = 37x0.23 0.98

43 9 poor fit

54 7

1.5 - 46

16 - 181

1.7 - 100

0.5 - 50

1.7 - 14

1.1 - 35 y = 44x0.18 0.92

Parent

antibody

group

Bivariate

equationR2

1 poor fit

25 y = 0.003x + 0.09 0.98

29 y = 0.009x + 0.11 0.99

39 y = 0.003x + 0.11 0.98

43 y = 0.008x + 0.18 0.63

54 y = 0.002x + 0.17 0.46

Daughter

antibody

group

Bivariate

equationR2

25A y = 0.003x + 0.10 0.89

25G y = 0.003x + 0.09 0.60

43B poor fit

43D poor fit

Daughter

antibody

group

Number of clones

from 2nd affinity

maturation phase

25A

13

25G

16

43B

10

43D

9

Affinity

range (nM)

1.1 - 1.8

1.3 - 2.0

0.6 - 14

2.3 - 26

0.1

1.0

2.0

0.1 1 10 100 1000

AD

C IC

50

(n

M)

Human KD (nM) Human K

D (nM)

AD

C IC

50

(n

M)

0

20

40

60

80

100

120

0.1 1 10 100 1000

%F

Xa

ge

ne

rati

on

at

50

nM

of

an

tib

od

y

%F

Xa

ge

ne

rati

on

at

50

nM

of

an

tib

od

y

0

20

40

60

80

100

120

0.1 1 10 100

43B

43D

25A

25G

43B

43D

25A

25G

0.075

0.15

0.225

0.1 1 10 100

1

25

29

39

43

54

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 20, 2018; DOI: 10.1158/1535-7163.MCT-18-0471

Page 24: Treating Tissue Factor-Positive Cancers With Antibody-Drug ......Aug 18, 2018  · maytansinoid DM1, had anticancer activity, but also inhibited coagulation (19). The TF-specific antibody

0

3x104

2x104

1x104

A

C D

B

E

1x10-31x10-4 1x10-2 1x10-1 1x100 1x101 1x102

Antibody concentration (nM) Antibody concentration (nM)

Med

ian

Flu

ore

scen

ce In

ten

sit

y (

MF

I)

1F

AntibodyEC50

(nM)

Antibody

25A

25A3

25G1

29E

39A

43B1

43D7

43Ea

54E

Isotype

0.05

0.17

0.08

0.09

0.15

0.04

0.28

0.09

0.53

0.24

nb

IC50

(nM)

1.15

1.54

0.69

2.13

Antibody

1F

Antibody

25A

25A3

25G1

29E

39A

43B1

43D7

43Ea

nc

nc

nc

nc

nc

nc

nc

54E

Isotype

1 10 1000

20

40

60

80

100

120

140

% F

Xa g

en

era

tio

n

Antibody concentration (nM)

Peak IIa

/Th

rom

bin

(% r

ela

tive t

o p

lasm

a)

0 10 20 30 40 50 60 70 80 90 1000

25

50

75

100

125

% F

VII b

ind

ing

Figure 2

Time (min)

IIa/T

hro

mb

in (

nM

)

1x10-3 1x10-2 1x10-1 1x100 1x101 1x102 1x1030

25

50

75

100

125

Antibody concentration (nM)

0 20 40 600

100

200

300

400

500

500

1F

25A

25A3

25G1

29E

39A

43B1

43D7

43Ea

54E

Isotype

TF-011

5G9

10H10

Plasma

1F

25A

25A3

25G1

29E

39A

43B1

43D7

43Ea

54E

Isotype

TF-011

5G9

10H10

1F

Antibody

25A

25A3

25G1

29E

39A

43B1

43D7

43Ea

54E

Isotype

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 20, 2018; DOI: 10.1158/1535-7163.MCT-18-0471

Page 25: Treating Tissue Factor-Positive Cancers With Antibody-Drug ......Aug 18, 2018  · maytansinoid DM1, had anticancer activity, but also inhibited coagulation (19). The TF-specific antibody

Figure 3

C

A

Antibody concentration (nM) Antibody concentration (nM)

Lu

min

escen

ce

B

D

ADCIC50

(nM)

25A

25A3

25G1

29E

39A

43B1

43D7

43Ea

Isotype

0.14

0.11

0.09

0.07

0.05

0.14

0.14

0.15

na na

ADCIC50

(nM)

25A

25A3

25G1

29E

39A

43B1

43D7

43Ea

Isotype

0.06

0.05

0.04

0.04

0.04

0.05

0.05

0.06

Antibody concentration (nM) Antibody concentration (nM)

Lu

min

escen

ce

Lu

min

escen

ce

0

1x105

2x105

1x105

2x105

1x10-3 1x10-2 1x10-1 1x100 1x101 1x10-3 1x10-2 1x10-1 1x100 1x1010

0

1x105

2x105

3x105

25A-vc-MMAE

25A3-vc-MMAE

25G1-vc-MMAE

29E-vc-MMAE

39A-vc-MMAE

43B1-vc-MMAE

43D7-vc-MMAE

43Ea-vc-MMAE

54E-vc-MMAE

Isotype-vc-MMAE

TF-011-vc-MMAE

0.09

0.07

0.06

0.06

0.05

0.08

0.06

0.09

0.07

na

0.05

0.35

0.19

0.19

0.20

0.12

0.36

0.28

0.43

0.26

na

0.17

continuous 72 h incubation

and culture4 h incubation, followed by

washout and 68 h culture

Cont.ADC

IC50 (nM)

Washout

1x10-3 1x10-2 1x10-1 1x100 1x101 1x10-3 1x10-2 1x10-1 1x100 1x101

Isotype ctrl-vc-MMAE

Tu

bu

lin

Tu

bu

lin

+ D

AP

I

A431

25A3-vc-MMAE Isotype ctrl-vc-MMAE 25A3-vc-MMAE

HPAF-II

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 20, 2018; DOI: 10.1158/1535-7163.MCT-18-0471

Page 26: Treating Tissue Factor-Positive Cancers With Antibody-Drug ......Aug 18, 2018  · maytansinoid DM1, had anticancer activity, but also inhibited coagulation (19). The TF-specific antibody

Figure 4

Antibody concentration (nM)

0.001 0.01 0.1 1 10 1000

1x105

2x105

3x105

4x105

Lu

min

escen

ce

Antibody concentration (nM)

0.001 0.01 0.1 1 10 1000

1x105

2x105

3x105

4x105

Lu

min

escen

ce

25A

NakedAntibody ADCUnconjugated antibody (naked) ADC

25A3

25G1

29E

39A

43B1

43D7

43Ea

54E

Isotype

0.26

0.24

0.18

0.28

0.26

0.42

0.27

0.43

0.30

na

0.31

EC50 (nM)

0.22

0.22

0.29

0.26

0.36

0.23

0.40

0.31

na

A

B

0.01 0.1 1 10 100 10000

500

1000

1500

IL8 c

on

c (

pg

/mL

)

0.01 0.1 1 10 100 10000

100

200

300

400

500

Antibody concentration (nM)Antibody concentration (nM)

GM

-CS

F c

on

c (

pg

/mL

)

1F

Antibody IC50

(nM)

25A

25A3

25G1

29E

39A

43B1

43D7

43Ea

54E

Isotype

1.0

2.7

2.3

1.9

1.5

1.0

1.7

1.5

2.1

2.2

ni

1F

Antibody IC50

(nM)

25A

25A3

25G1

29E

39A

43B1

43D7

43Ea

54E

Isotype

1.3

2.5

2.3

1.7

1.5

1.7

1.8

1.3

2.6

2.4

ni

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 20, 2018; DOI: 10.1158/1535-7163.MCT-18-0471

Page 27: Treating Tissue Factor-Positive Cancers With Antibody-Drug ......Aug 18, 2018  · maytansinoid DM1, had anticancer activity, but also inhibited coagulation (19). The TF-specific antibody

A B

Figure 5

Day

Mean

tu

mo

r vo

lum

e, S

EM

(m

m3)

25A-vc-MMAE

25A3-vc-MMAE

2 mg/kg, qwk x 2, IV

25G1-vc-MMAE

39A-vc-MMAE

43B1-vc-MMAE

43D7-vc-MMAE

Isotype-vc-MMAE

Vehicle

43Ea-vc-MMAE

DayMean

tu

mo

r vo

lum

e, S

EM

(m

m3)

25A-vc-MMAE

5 mg/kg, qwk x 3, IP

43Ea-vc-MMAE

Isotype-vc-MMAE

TF-011-vc-MMAE

TreatmentDay 21

(n) P %TGI

10 8 [2] <0.0001 132

10 8 [3] <0.0001 131

10 434 [76] NS 56

10 7 [3] <0.0001 132

0/8/2

0/6/4

0/0/0

0/5/5

Vehicle 10 749 [68] - 0 0/0/0

Mean

[SEM] (mm3)

+- Mean

[SEM] (mm3)

+-Day 59

PR/CR/TFS

Day 39

PR/CR/TFS

Day 41

PR/CR/TFS

TreatmentDay 21

(n) P %TGI

10 4 [1] <0.001 139

10 47 [40] <0.001 129

10 4 [1] <0.001 138

10 8 [2] <0.001 137

10 10 [3] <0.001 137

10 9 [3] <0.001 137

9 23 [22] <0.001 135

10 598 [62] NS 22

10 708 [65] - 0

1/1/8

2/0/7

1/2/7

1/0/9

3/1/6

3/1/6

0/0/8

0/0/0

0/0/0

0 10 20 30 400

500

1000

1500

0 10 20 30 40 50 600

500

1000

1500

C D

Day Day

Mean

tu

mo

r vo

lum

e, S

EM

(m

m3)

25A-vc-MMAE

25A3-vc-MMAE

4 mg/kg, qwk x 2, IV

25G1-vc-MMAE

29E-vc-MMAE

39A-vc-MMAE

43B1-vc-MMAE

43D7-vc-MMAE

43Ea-vc-MMAE

Isotype-vc-MMAE

Vehicle

TreatmentDay 25

(n) P %TGI

Mean

[SEM] (mm3)

+- Day 49

PR/CR/TFS

10 174 [43] <0.0001 98

10 153 [43] <0.0001 100

10 449 [102] <0.0001 69

10 180 [78] <0.0001 97

10 160 [45] <0.0001 99

10 232 [66] <0.0001 92

10 217 [49] <0.0001 93

10 203 [56] <0.0001 95

10 873 [114] NS 24

10 1098 [133] - 0

3/0/1

1/1/1

1/1/1

1/0/3

0/4/1

1/0/0

0/0/1

2/0/0

0/0/0

0/0/0

0/0/0

0/0/0

0/0/0

2/0/0

0/0/0

0/0/0

0/0/0

0/0/0

0/0/0

Mean

tu

mo

r vo

lum

e, S

EM

(m

m3)

25A-vc-MMAE

25A3-vc-MMAE

2 mg/kg, qwk x 2, IV

25G1-vc-MMAE

39A-vc-MMAE

43B1-vc-MMAE

43D7-vc-MMAE

43Ea-vc-MMAE

Isotype-vc-MMAE

Vehicle

TreatmentDay 27

(n) P %TGI

Mean

[SEM] (mm3)

+-

7 517 [52] <0.001 67

7 495 [69] <0.0001 68

7 550 [53] <0.001 63

7 223 [46] <0.0001 92

7 472 [58] <0.0001 70

7 708 [104] <0.01 49

7 728 [188] <0.05 48

7 1197 [88] NS 9

7 1292 [180] - 0

0 10 20 30 40 500

500

1000

1500

0 10 20 30 40 500

500

1000

1500

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 20, 2018; DOI: 10.1158/1535-7163.MCT-18-0471

Page 28: Treating Tissue Factor-Positive Cancers With Antibody-Drug ......Aug 18, 2018  · maytansinoid DM1, had anticancer activity, but also inhibited coagulation (19). The TF-specific antibody

AFigure 6

C

B

25A-vc-MMAE

Isotype-vc-MMAE

43Ea-vc-MMAE

10

10

10

<0.001

-

<0.001

124

0

119

5/0/1

0/0/0

6/0/0

74 [16]

780 [154]TF-011-vc-MMAE 10 <0.001 128 6/0/249 [8]

100 [18]

Day

Mean

tu

mo

r vo

lum

e, S

EM

(m

m3)

5 mg/kg, qwk x 2, IP

Day

Mean

tu

mo

r vo

lum

e, S

EM

(m

m3)

2.5 mg/kg, qwk x 3, IP

Day

Mean

tu

mo

r vo

lum

e, S

EM

(m

m3)

4 mg/kg, qwk x 2, IV

Head &

Neck

H-s

core

250

Ovarian

H-s

core

220

Gastr

ic

H-s

core

155

TreatmentDay 44

(n) P %TGI

Mean

[SEM] (mm3)

+- Day 60

PR/CR/TFS

25A-vc-MMAE

Isotype-vc-MMAE

9

9

<0.001

-

112

0

2/2/3

0/0/0

24 [7]

1093 [143]

TF-011-vc-MMAE 9 <0.001 112 0/1/546 [12]

TreatmentDay 25

(n) P %TGI

Mean

[SEM] (mm3)

+- Day 46

PR/CR/TFS

25A-vc-MMAE

Isotype-vc-MMAE

43Ea-vc-MMAE

10

10

10

<0.001

-

<0.001

71

0

74

0/0/0

0/0/0

0/0/0

464 [35]

1131 [118]

TF-011-vc-MMAE 10 <0.001 72 0/0/0452 [50]

439 [23]

TreatmentDay 15

(n) P %TGI

Mean

[SEM] (mm3)

+- Day 46

PR/CR/TFS

0 10 20 30 40 500

500

1000

1500

0 10 20 30 40 50 600

500

1000

1500

0 10 20 30 40 500

500

1000

1500 D

Mean

tu

mo

r vo

lum

e, S

EM

(m

m3)

TreatmentDay 15

(n) P

Mean

[SEM] (mm3)

+-

10 mg/kg, qwk x 2, IV

Day

Vehicle

Isotype

25A

43Ea

25A3

8

9

9

9

8

954 [86]

938 [78]

976 [114]

898 [73]

829 [67] -

NS

NS

NS

NS

0 10 200

500

1000

1500

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 20, 2018; DOI: 10.1158/1535-7163.MCT-18-0471

Page 29: Treating Tissue Factor-Positive Cancers With Antibody-Drug ......Aug 18, 2018  · maytansinoid DM1, had anticancer activity, but also inhibited coagulation (19). The TF-specific antibody

Published OnlineFirst August 20, 2018.Mol Cancer Ther   Jan-Willem Theunissen, Allen G Cai, Maryam M Bhatti, et al.   Conjugates That Do Not Affect Blood ClottingTreating Tissue Factor-Positive Cancers With Antibody-Drug

  Updated version

  10.1158/1535-7163.MCT-18-0471doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://mct.aacrjournals.org/content/suppl/2018/08/18/1535-7163.MCT-18-0471.DC1

Access the most recent supplemental material at:

  Manuscript

Authoredited. Author manuscripts have been peer reviewed and accepted for publication but have not yet been

   

   

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected] at

To order reprints of this article or to subscribe to the journal, contact the AACR Publications

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://mct.aacrjournals.org/content/early/2018/08/18/1535-7163.MCT-18-0471To request permission to re-use all or part of this article, use this link

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 20, 2018; DOI: 10.1158/1535-7163.MCT-18-0471