Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
MULTISPECIFIC ANTIBODY PRODUCT THAT BINDS TO DIFFERENT ROR1 EPITOPES
Document Type and Number:
WIPO Patent Application WO/2019/016381
Kind Code:
A1
Abstract:
The present invention relates to a multi-specific product comprising a first entity comprising an antigen-binding domain that binds to the same RORl epitope as and/or competes for RORl binding with an antibody comprising a heavy chain variable region sequence shown in SEQ ID NO. 2 and a light chain variable region sequence shown in SEQ ID NO. 3; and a second entity comprising an antigen-binding domain that binds to a different target or RORl epitope than, and/or does not compete for binding with the antibody comprising a heavy chain variable region sequence shown in SEQ ID NO. 2 and a light chain variable region sequence shown in SEQ ID NO. 3.

Inventors:
GRAWUNDER ULF (CH)
BEERLI ROGER (CH)
WALDMEIER LORENZ (CH)
Application Number:
PCT/EP2018/069798
Publication Date:
January 24, 2019
Filing Date:
July 20, 2018
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
NBE THERAPEUTICS AG (CH)
International Classes:
C07K16/28; A61K47/68; A61P35/00
Domestic Patent References:
WO2016187220A22016-11-24
WO2015184203A12015-12-03
WO2016055592A12016-04-14
WO2016055593A12016-04-14
WO2016094873A22016-06-16
WO2014167022A12014-10-16
WO2017053469A22017-03-30
WO2017127499A12017-07-27
WO2017142928A12017-08-24
WO2014140317A22014-09-18
WO2016102697A12016-06-30
WO2016102679A12016-06-30
WO2014140317A22014-09-18
Other References:
R. E. KONTERMANN: "Bispecific Antibodies", 2011, SPRINGER, ISBN: 978-3-642-20909-3, article R. E. KONTERMANN: "Chapter 1 Bispecific Antibodies: Developments and Current Perspectives", pages: 4, 8 - 19, XP002785676, DOI: 10.1007/978-3-642-20910-9
BEERLI ROGER R ET AL: "Sortase Enzyme-Mediated Generation of Site-Specifically Conjugated Antibody Drug Conjugates with High In Vitro and In Vivo Potency.", PLOS ONE, vol. 10, no. 7, E0131177, 1 July 2015 (2015-07-01), pages 1 - 17, XP002785677, ISSN: 1932-6203, DOI: 10.1371/journal.pone.0131177
REBAGAY ET AL., FRONT ONCOL., vol. 2, no. 34, 2012, pages 1 - 8
BALAKRISHNAN ET AL., CLIN CANCER RES., 2016
REBAGAY ET AL., FRONTIERS ONCOL., vol. 2, no. 34, 2012, pages 1 - 8
SHATZ ET AL., MABS, vol. 5, no. 6, 2013, pages 872 - 881
WARD ET AL., NATURE, vol. 341, 1989, pages 544 - 546
BIRD ET AL., SCIENCE, vol. 242, 1988, pages 423 - 426
HUSTON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 5879 - 5883
SKERRA; PLUCKTHUN, SCIENCE, vol. 240, 1988, pages 1038 - 41
REITER ET AL., INT. J. CANCER, vol. 67, 1996, pages 113 - 23
CAI; GAREN, PROC. NATL. ACAD. SCI. USA, vol. 93, 1996, pages 6280 - 85
DUMOULIN ET AL., NAT. STRUCT. BIOL., vol. 11, 2002, pages 500 - 515
GHAHROUDI ET AL., FEBS LETTERS, vol. 414, 1997, pages 521 - 526
BOND ET AL., J. MOL. BIOL., vol. 332, 2003, pages 643 - 55
HARLOW; LANE: "Using Antibodies, A Laboratory Manual", 1998, COLD SPRING HARBOR LABORATORY PRESS
BEERLI ET AL., PLOS ONE, vol. 10, 2015, pages e131177
SHINKAI ET AL., CELL, vol. 68, 1992, pages 855 - 67
YANG ET AL., PLOS ONE, vol. 6, 2011, pages e21018
KWONG; RADER: "Curr Protoc Protein Sci", 2009, article "Chapter 6:Unit 6 10"
MCCARTNEY-FRANCIS ET AL., PROC. NATL. ACAD. SCI. USA, vol. 81, 1984, pages 1794 - 1798
POPKOV ET AL., J. MOL. BIOL., vol. 325, 2003, pages 325 - 335
RADER, METHODS MOL BIOL, vol. 525, 2009, pages 101 - 128
HOFER ET AL., J IMMUNOL METH, vol. 318, 2007, pages 75 - 87
YANG; RADER, METHODS MOL BIOL, vol. 901, 2012, pages 209 - 232
BASKAR ET AL., INT J MED SCI., vol. 9, no. 3, 2012, pages 193 - 9
SKERRA; PLUCKTHUN, SCIENCE, 1988, pages 2401038 - 41
LEFRANC MP ET AL., NUCLEIC ACIDS RES., vol. 43, pages D413 - 22
BOND ET AL., J. MOL. BIOL., 2003, pages 332643 - 55
CHEN ET AL., PNAS, vol. 108, no. 28, 2011, pages 11399 - 11404
DORR BM ET AL., PNAS, vol. 111, 2014, pages 13343 - 8
BEERLI ET AL., PLOS ONE, vol. 10, 2015, pages e131177
QUINTIERI ET AL., CLIN. CANCER RES, vol. 11, 2005, pages 1608 - 1617
YANG ET AL., PLOS ONE, vol. 6, 2011, pages e21018
MCCARTNEY-FRANCIS ET AL., PROC. NATL. ACAD. SCI. USA, vol. 81, pages 1794 - 1798
GAREN, PROC. NATL. ACAD. SCI. USA, vol. 93, 1996, pages 6280 - 85
Attorney, Agent or Firm:
MICHALSKI HÜTTERMANN & PARTNER, PATENTANWÄLTE MBB et al. (DE)
Download PDF:
Claims:
What is claimed is:

1. A multi-specific product comprising

(a) a first entity comprising an antigen-binding domain that

• binds to the same ROR1 epitope as and/or

• competes for ROR1 binding with antibody ERRl-324 comprising a heavy chain variable region sequence shown in SEQ ID NO. 2 and a light chain variable region sequence shown in SEQ ID NO. 3; and

(b) a second entity comprising an antigen-binding domain that binds to

• to a different ROR1 epitope than, and/or

• does not compete for binding with antibody ERRl-324 comprising a heavy chain variable region sequence shown in SEQ ID NO. 2 and a light chain variable region sequence shown in SEQ ID NO. 3.

2. The product according to claim 1, which is a multi-specific antibody, alternative scaffold or antibody mimetic, wherein

(a) the first entity is a first antigen-binding domain that

• binds to the same ROR1 epitope as and/or

• competes for ROR1 binding with antibody ERRl-324 comprising a heavy chain variable region sequence shown in SEQ ID NO. 2 and a light chain variable region sequence shown in SEQ ID NO. 3; and

(b) the second entity is a second antigen-binding domain that • binds to a different ROR1 epitope than, and/or

• does not compete for binding with antibody ERRl-324 comprising a heavy chain variable region sequence shown in SEQ ID NO. 2 and a light chain variable region sequence shown in SEQ ID NO. 3.

3. The product according to claim 1, which comprises two or more antibodies, alternative scaffolds or antibody mimetics, wherein

(a) the first entity is a first antibody comprising an antigen-binding domain that

• binds to the same ROR1 epitope as and/or

• competes for ROR1 binding with antibody ERRl-324 comprising a heavy chain variable region sequence shown in SEQ ID NO. 2 and a light chain variable region sequence shown in SEQ ID NO. 3; and

(b) the second entity is a second antibody comprising an antigen-binding domain that

• binds to a different ROR1 epitope than, and/or

• does not compete for binding with antibody ERRl-324 comprising a heavy chain variable region sequence shown in SEQ ID NO. 2 and a light chain variable region sequence shown in SEQ ID NO. 3.

4. The product according to any one of claims 1 - 3, wherein the entity comprising an antigen-binding domain is at least one selected from the group consisting of an antibody, an antibody-based binding protein, a bi-epitope-reactive antibody, a modified antibody format retaining target binding capacity, an antibody derivative or a fragment retaining target binding capacity, an alternative scaffold and/or an antibody mimetic.

5. The product according to any one of claims 1 - 4, wherein the antibody is at least one selected from the group consisting of an antibody, an antibody-based binding protein, a bi- epitope-reactive antibody, a modified antibody format retaining target binding capacity, an antibody derivative or a fragment retaining target binding capacity.

6. The product according to any one of claims 3 - 5, wherein the first and/or the second antibody, alternative scaffold or antibody mimetic is monospecific or mono-epitope reactive.

7. The product according to any one of claims 1 - 6, wherein the second antibody, alternative scaffold or antibody mimetic, or the second antigen-binding domain binds to ROR1, yet

• to a different epitope than, and/or

• does not compete for binding with antibody ERRl-324 comprising a heavy chain variable region sequence shown in SEQ ID NO. 2 and a light chain variable region sequence shown in SEQ ID NO. 3.

8. The product according to claim 7, wherein the second antibody, alternative scaffold or antibody mimetic, or second antigen-binding domain

• binds to the same ROR1 epitope as and/or

• competes for ROR1 binding with at least one antibody selected from the group consisting of a) R12, comprising a heavy chain variable region sequence shown in SEQ ID NO. 20 and a light chain variable region sequence shown in SEQ ID NO. 21,

b) ERR1-TOP43, comprising a heavy chain variable region sequence shown in SEQ ID NO. 12 and a light chain variable region sequence shown in SEQ ID NO. 13, c) ERRl-301, comprising a heavy chain variable region sequence shown in SEQ ID NO.

4 and a light chain variable region sequence shown in SEQ ID NO. 5,

d) ERR1-402, comprising a heavy chain variable region sequence shown in SEQ ID NO.

8 and a light chain variable region sequence shown in SEQ ID NO. 9, and/or e) 2A2, comprising a heavy chain variable region sequence shown in SEQ ID NO. 22 and a light chain variable region sequence shown in SEQ ID NO. 23.

9. The product according to any one of claims 1 - 8, wherein ROR1 is human ROR1 (hRORl).

10. The product according to any one of claims 2 and 4 - 9, which is in a format selected from the group consisting of

• bispecific or bi-epitope reactive scFv-Fc,

• bispecifc or bi-epitope reactive scFv-Ig, and/or

• DVD-Ig.

11. The multi-specific product according to any one of claims 1 - 10, wherein the first entity comprises the following CDRs:

• QASQSVYGNNELA (VL CDR1 , SEQ ID NO. 69),

• RASILTS (VL CDR2, SEQ ID NO. 70),

• LGGYVSQSYRAA (VL CDR3, SEQ ID NO. 71),

• RNGMT (VH CDRl SEQ ID NO. 72),

• IITSSGDKYYATWAKG (VH CDR2, SEQ ID NO. 73), and

• GTVSSDI (VH CDR3, SEQ ID NO. 74), and wherein the CDRs are comprised in a suitable protein framework so as to be capable to bind to RORl.

12. The multi-specific product according to any one of claims 1 - 10, wherein the first entity comprises the heavy chain variable region sequence of antibody ERRl-324 shown in SEQ ID NO. 2 and the light chain variable region sequence of antibody ERRl-324 shown in SEQ ID NO. 3.

13. The multi-specific product according to any one of claims 1

entity comprises at least one of the following sequence pairs: a) the heavy chain variable region sequence of antibody R12 shown in SEQ ID NO. 20 and the light chain variable region sequence shown in SEQ ID NO. 21,

b) the heavy chain variable region sequence of antibody ERR1-TOP43 shown in SEQ ID NO. 12 and the light chain variable region sequence of antibody ERR1-TOP43 shown in SEQ ID NO. 13,

c) the heavy chain variable region sequence of antibody ERR 1-301 shown in SEQ ID NO. 4 and the light chain variable region sequence of antibody ERR 1-301 shown in SEQ ID NO. 5,

d) the heavy chain variable region sequence of antibody ERRl-402 shown in SEQ ID NO. 8 and the light chain variable region sequence of antibody ERRl-402 shown in SEQ ID NO. 9, and/or

e) the heavy chain variable region sequence of antibody 2A2 shown in SEQ ID NO. 22 and the light chain variable region sequence of antibody 2A2 shown in SEQ ID NO. 23.

14. An antibody drug conjugate (ADC) having the general formula A - (L)n - (T)m, in which

• A is at least one antigen binding domain, antibody, alternative scaffold or antibody mimetic according to any one of claims 2 - 13,

• L is a linker,

• T is a toxin. and in which n and m are integers between >1 and < 10.

15. An antibody effector conjugate (AEC) having the general formula A - (L)n - (E)m, in which

• A is at least one antigen binding domain, antibody, alternative scaffold or antibody mimetic according to any one of claims 2 - 13,

• L is a linker,

• E is a label and in which n and m are integers between >1 and < 10.

16. The conjugate according to any of claims 14 - 15, wherein the antibody is a multispecific antibody according to any of claims 2 and 4 - 13.

17. A composition comprising at least two antibody effector conjugates (AEC) or antibody drug conjugates (ADC) according to any one of claims 14 - 16, wherein each of the two conjugates comprises one of the monospecific antibodies, alternative scaffolds or antibody mimetics of any of claims 3 - 8.

18. The conjugate according to any one of claims 14 - 16, wherein the linker is at least one selected from the group consisting of

• an oligopeptide linker

• a maleimide linker, optionally comprising cleavable spacers, that may be cleaved by changes in pH, redox potential and or specific intracellular or extracellular enzymes.

19. The conjugate or composition according to any one of claims 14 - 18, wherein the linker has at least one of the following amino acid sequences: - LPXTGn-, -LPXAGn-, -LPXSGn-, - LAXTGn-, -LPXTGn-, -LPXTAn- or -NPQTGn-, with Gn being an oligo- or polyglycine or polyalanine with n being an integer between > 1 and < 21, , and X being any conceivable amino acid sequence.

20. The conjugate or composition according to any one of claims 14 - 19, wherein the linker is conjugated to the C-terminus of at least one subdomain of the antibody.

21. The conjugate or composition according to any one of claims 14 - 20, wherein, prior to conjugation

• the antibody bears a sortase recognition tag used or conjugated to the C-terminus of at least one subdomain thereof, and

• the toxin or label comprises a glycine stretch with a length of between > 1 and < 20 glycine residues, preferably with a length of > 2 and < 5 glycine residues.

22. The conjugate or composition according to any one of claims 14 - 21, wherein, the toxin, or derivative thereof, is at least one selected from the group consisting of:

• maytansinoids,

• auristatins,

• anthracyclins, preferably PNU-159682 derived anthracyclins

• calicheamicins,

• tubulysins

• duocarmycins

• radioisotopes

• liposomes comprising a toxid payload

• protein toxins

• taxanes, and/or

• pyrrolobenzodiazepines.

23. The conjugate or composition according to any one of claims 14 - 22, which is created by sortase-mediated conjugation of (i) an antibody carrying one or more sortase recognition tags and (ii) one or more toxins or labels carrying an oligoglycine tag.

24. A method of producing a conjugate according to any one of claims 14 - 23, which method comprises the following steps: a) providing an antibody, alternative scaffold or antibody mimetic according to any one of claims 2 - 13, which antibody carries a sortase recognition tag, b) providing one or more toxins or labels carrying an oligoglycine tag, and c) conjugating the antibody and the toxin or label by means of sortase-mediated

conjugation.

25. Use of the multispecific product according to any one of claims 1 - 13 or the antibody drug conjugate according to any one of claims 14 and 16 - 23, for the treatment of a patient that is

• suffering from,

• at risk of developing, and/or

• being diagnosed for a neoplastic disease.

26. A pharmaceutical composition comprising the multispecific product according to any one of claims 1 - 13 or the antibody drug conjugate according to any one of claims 14 and 16 - 23, together with one or more pharmaceutically acceptable ingredients.

27. A method of killing or inhibiting the growth of a cell expressing RORl in vitro or in a patient, which method comprises administering to the cell a pharmaceutically effective amount or dose of the multispecific product according to any one of claims 1 - 13, the antibody drug conjugate according to any one of claims 14 and 16 - 23, or of the

pharmaceutical composition according to claim 26.

28. The method according to claim 27, wherein the cell expressing RORl is a cancer cell.

Description:
multispecific antibody product that binds to different ROR1 epitopes

FIELD OF THE INVENTION

The present invention relates to a multispecific antibody product that binds to a first ROR1 epitope and to at least one other epitope of ROR1, and conjugates thereof, as well as to uses thereof.

BACKGROUND OF THE INVENTION

Cancer is one of the leading causes of death. It is a class of diseases caused by malignant transformation of healthy cells, resulting from genetic alterations, like chromosomal translocations, mutations in tumor suppressor genes, transcription factors or growth-factor receptors, leading to the immortalization of the cells. If the immortalization is combined with excessive proliferation, the immortalized cells generate tumors, with or without metastasis (in case of solid tumors), or leukemias and lymphomas (cancers of the blood). Defective apoptosis, or programmed cell death, can further contribute to malignant transformation of cells leading to cancer.

A family of membrane associated receptor tyrosine kinases, consisting of the receptor tyrosine kinase orphan receptors- 1 and -2 (ROR1 and ROR2) have been described as specifically associated with particular cancers (Rebagay et al. (2012) Front Oncol. 2(34): 1-8; doi 10.3389/onc.2012.00034), while being largely absent in expression on healthy tissue

with, a few exceptions e.g. in case of RORl (Balakrishnan et al. (2016) Clin Cancer Res. doi: 10.1158/1078-0432). Whether or not ROR expression is functionally associated with tumorigenesis remains unclear. However, due to the very tumor-selective expression of ROR family members, they represent relevant targets for targeted cancer therapies.

Receptor tyrosine kinase orphan receptors- 1 and -2, RORl and ROR2, are the only two family members defining a new receptor tyrosine kinase family, based on the overall structural design and some functional similarities. Both RORl and ROR2 proteins are type I- single pass trans-membrane receptors with an extracellular domain (ECD) consisting of an immunoglobulin domain, a cysteine rich frizzled domain and a Kringle domain. These three extracellular domains are followed by a trans-membrane domain connecting the ECD to an intracellular portion of the protein comprising kinase domains (Rebagay et al. (2012) Frontiers Oncol. 2(34):l-8; doi 10.3389/onc.2012.00034).

The human RORl and ROR2 proteins are 58% homologous between each other, but each of the ROR proteins is highly conserved between species. This represents a challenge for the development of human RORl specific monoclonal antibodies and very few antibodies are known.

Further, it appears that anti-RORl antibodies, and antibody drug conjugates (ADCs) that encompass anti-RORl antibodies, show only limited efficacy, in particular on cell lines and tumors with low expression levels of RORl .

It is hence one object of the present invention to provide antibody-based products that target RORl and demonstrate a better efficacy, in particular on cell lines and tumors with low expression levels of RORl.

It is another object of the present invention to provide antibody drug conjugates (ADCs) that target RORl and demonstrate a better efficacy, in particular on cell lines with low expression levels.

These and further objects are met with methods and means according to the independent claims of the present invention. The dependent claims are related to specific

embodiments. Summary of the Invention The present invention relates to a multi-specific product that binds to a first ROR1 epitope and to at least one other epitope on ROR1, and conjugates thereof, as well as the uses thereof. The invention and general advantages of its features will be discussed in detail below.

Detailed Description of the Figures

Fig. 1 shows the amino acid sequences of variable immunoglobulin heavy and light chains of novel rabbit anti-human ROR1 (hRORl) mAbs, as indicated. The amino acid sequence alignment of the rabbit variable domains (V K , Υχ, and VH) is shown with framework regions (FR) and complementarity determining regions (CDR) using Kabat numbering. Shown in the figure are the heavy chain variable domain sequences and the light chain variable domain sequences of 13 antibodies designated XBR1-402, ERR1-301, ERR1-306, ERRl-316, ERRl- 324, ERRl-403, ERRl-409, ERR1-TOP4, ERR1-TOP15, ERR1-TOP22, ERR1-TOP40, ERR1-TOP43, and ERR1-TOP54. As indicated in the figure clones XBR1-402, ERRl-301, ERR1-306, ERRl -316, ERRl -403, ERRl-409, ERR1-TOP4, ERR1-TOP15, ERR1-TOP22, ERR1-TOP43, and ERR1-TOP54 are variable domains of immunoglobulin λ light chains, while antibodies ERRl-324 and ERR1-TOP40 are variable domains of immunoglobulin κ light chains.

Fig. 2 shows the binding activity of chimeric rabbit/human Fabs to human ROR1 (hRORl) and mouse ROR1 (mRORl) expressed as fusion proteins of the extracellular domain (ECD) of hRORl and mRORl to the human Fc domain of a human IgGl antibody. The binding of each chimeric rabbit/human Fab to hRORl and mRORl fused with human IgGl Fc (hFc- hRORl and hFc-mRORl) was analyzed by ELISA. hFc-RORl or hFc-mRORl were captured by anti-human IgGl Fc antibody immobilized on plate and then incubated with hRORl specific Fabs comprising a His-tag via detection with mouse anti-His tag. Specificity of the Fabs was confirmed by using fusion proteins of the extracellular domain (ECD) of hROR2 with the human Fc domain of a human IgGl antibody (hFc-hROR2) and with bovine serum albumin (BSA) as control.

Fig. 3 shows binding activity of chimeric rabbit/human Fabs to native human ROR1 protein expressed on the cell surface of murine preB cell line 63-12 (see Example 1). The binding of each chimeric rabbit/human Fab to the ectopically expressed human ROR1 on mouse pre-B cell (63-12) surface was analyzed by flow cytometry. ERR2-TOP35 is a mAb against hROR2 that served as an isotype-matched control.

Fig. 4 shows epitope mapping studies for chimeric rabbit/human Fabs on six different immobilized IgGl-Fc fusion proteins that comprise different parts of the extracellular domain of human ROR1: hFc-hRORl-Ig (comprising the Immunoglobulin-domain of hRORl), hFc- hRORl-Fr (comprising the Frizzled domain of hRORl), hFc-hRORl-Kr (comprising the Kringle domain of hRORl), hFc-hRORl-Ig-Fr (comprising the Immunoglobulin and Frizzled domains of hRORl), hFc-hRORl-Fr-Ki (comprising the Frizzled and Kringle domains of hRORl) and hFc-hRORl (comprising the entire extracellular domain (ECD) of hRORl).

Fig. 5 shows epitope binding studies performed by surface plasmon resonance. Shown are SPR sensorgrams obtained for the binding of different Fabs to hFc-hRORl captured by anti- human Fey antibody immobilized on a CM5 chip. Fabs were injected in different orders to identify independent and overlapping epitopes. Resonance unit (RU, y axis) increases that exceeded the values found for previously injected Fabs indicated independent epitopes because they allow simultaneous binding. For example, the increase found for the binding of Fab Rl 1 exceeded the values found for XBR1-402 alone, indicating that Fab Rl 1 and XBR1- 402 can bind simultaneously to human ROR1. By contrast, the epitope of Fab XBR1-402 overlaps with the epitopes of ERR1-301, ERR1-403 and R12 (left graph); the epitope of Fab ERR1-TOP43 overlaps with the epitope of ERR1-306, XBR1-402 and ERR1-TOP40. The x- axis depicts the time in seconds (s).

Fig. 6 shows sensorgrams of affinity measurements of anti-hRORl specific Fabs to hRORl ECD by surface plasmon resonance (SPR). (A) Shown are Biacore XI 00 sensorgrams obtained for the binding of each Fab to hFc-hRORl captured by anti-human Fey antibody immobilized on CM5 chip after instantaneous background depletion. Fabs were injected at five different concentrations with the highest concentration indicated in Figure 6(B), one of the five concentrations was tested in duplicates. (B) Monospecific affinities of each Fab are shown in the table. The equilibrium dissociation constant (_¾) was calculated from k 0 fj/k on (k ori , association rate constant; k 0 /f, dissociation rate constant).

Fig. 7 shows the binding analyzed by ELISA of selected hRORl specific rabbit-human-Fc chimeric antibodies of selected clones ERRl-301, XBR1-402, ERR1-306, ERRl-324, ERR1- 403 and ER l-Top43 to recombinant, purified hRORl (panel A) and to recombinant, purified hROR2 as a negative control (panel B).

Fig. 8 shows FACS-based cell staining of hRORl on various human cancer cell lines with anti-human ROR1 antibody 2A2 as described in Example 9. Cell lines analyzed include 697 (human acute lymphocytic leukemia, ALL), human triple-negative breast cancer cell lines MDA-MB-468 and HS-578T, human lung cancer cell line A549, human colon cancer cell line HT-29, as well as human breast cancer cell line T47D. Except for the T47D human breast cancer cell line, all of the evaluated cells are positive for hRORl expression.

Fig. 9 shows schematically how site-specifically conjugated ADCs disclosed in this invention have been generated. (A) schematically shows the mechanism of sortase-enzyme mediated antibody conjugation (SMAC-technology) as disclosed in WO2014140317. In order to generate site-specifically conjugated ADCs, recombinant antibodies need to be expressed with the C-terminal pentapeptide motif LPXTG, which serve as recognition sites for the sortase A enzyme from Staphylococcus aureus (SrtA). When a glycine modified toxin substrate is incubated with pentapeptide motif LPXTG containing antibody and sortase A enzyme, the sortase A enzyme catalyzes a transpeptidation reaction by which the glycine- modified toxin replaces the C-terminal glycine of the LPXTG motif and is covalently coupled to the threonine of the remaining LPXT sequence. This way C-terminally toxin-conjugated ADCs can be generated with high efficiency. (B) shows the structure of the preferred toxin, a PNU- 159682 derivative comprising an ethylene-diamino (EDA) linker connecting a 5x glycine stretch to the carbonyl group at CI 3 of the anthracycline structure, as disclosed in WO2016102697.

Fig. 10 shows in vitro cell killing assays performed on human ALL cancer cell line 697 with individual antibody-based ADCs and mixtures thereof comprising ERRl-324-based ADCs as per Example 10. For the same total dose of ADC, the mixtures of selected ADCs of the invention provide synergistic killing of 697 cancer cells that is superior to the individual ADCs.

Fig. 11 shows evaluation of in vitro cell killing performed on ROR1 positive human ALL cancer cell line 697 with individual scFv-Fc-G5-PNU-toxin conjugates based on anti-RORl antibody XBR1-402 and anti-RORl antibody ERR1-324 and also mixtures of these two scFv-Fcs at the combined same concentration as used for the single scFv-Fcs, as well as a bi- epitope-reactive scFv-Fc-based ADCs (BETR-ADC™), in which the two binding domains are combined in a single, bi-specific antibody molecule, as indicated. For the same total dose of ADC, the mixtures of selected ADCs and the bi-epitope-reactive scFv-Fc-based ADC (BETR-ADC™) of the invention provide improved cell killing activity of 697 cancer cells that is superior to the cell killing activity on RORl positive 697 cells of individual ADCs at equivalent total concentration.

Fig. 12 shows evaluation of in vitro cell killing performed on human ALL cancer cell line 697 with individual DVD-Ig-based bi-epitope-reactive anti-RORl ADCs (BETR-ADC™) based on anti-RORl antibody XBRl-402 and anti-RORl antibody ERRl-324 and mixtures of ADCs based on anti-RORl antibody XBRl-402 and anti-RORl antibody ERRl-324. For the same total dose of ADC, the bi-epitope-reactive DVD-Ig-based anti-RORl ADC of the invention show cell killing activity of 697 cancer cells that is superior to the cell killing activity on RORl positive 697 cells of individual ADCs at equivalent total concentration. Trastuzumab-PNU ADC (Tras-G5-PNU) was used as an isotype-matched control ADC.

Fig. 13 shows evaluation of in vitro cell killing performed on RORl -positive human cancer cell lines (colon cancer cell line HT-29, triple-negative breast cancer cell lines MDA-MB-468 and HS-578T, lung cancer cell line A549,) with individual anti-RORl specific antibody- based ADCs (A) and mixtures thereof (B), as indicated, and comprising anti-RORl ERRl- 324-based ADCs as per Example 13. For the same total dose of ADC, the mixtures of selected ADCs of the invention provide synergistic target cell killing on RORl -positive cell lines that is superior to the target cell killing with individual ADCs. CD30-specific PNU- ADC based on brentuximab (clone AclO) was used as an isotype-matched control ADC.

Fig. 14 shows three different embodiments of the invention that have experimentally been evaluated. Fig 14 A shows the use of mixtures of two ADCs comprising a monospecific anti- RORl antibody each that bind to different epitopes of a RORl target molecule. The two monospecific antibodies target two different epitopes of RORl, one of which competes for binding to RORl with an antibody having SEQ ID NOs 2 and 3. Fig 14 B shows the use of a bi-epitope-reactive ADC (BETR-ADC™) comprising a bi-epitope-reactive antibody in the scFv-Fc format. The two target binding domains of the two scFv subunits bind to two different epitopes of RORl, one of which competes for binding to RORl with an antibody having SEQ ID NOs 2 and 3. Fig 14 C shows the use of a bi-epitope-reactive ADC (BETR- ADC™) comprising a bi-epitope-reactive antibody in the DVD-Ig format. The two target binding domains bind to two different epitopes of ROR1, one of which competes for binding to ROR1 with an antibody having SEQ ID NOs 2 and 3.

Fig. 15 schematically shows the structures of the three bi-epitope-reactive antibody formats scFv-Fc, DVD-Ig and bispecific scFv-Ig. Note that in a knob-in-hole embodiment, the two Fc domains can be structurally modified, as, e.g., shown in Shatz et al., mAbs 5:6, 872- 881(2013). Note also the respectively modified sequences in the sequence listing.

Fig. 16 shows how recognition of two independent epitopes by bi-epitope-reactive ADC (BETR-ADC™) can not only induce the formation of target-homodimers (A), but lead to extensive clustering of the target protein (B). This in turn may enhance internalization of said product, a more efficient transport to intracellular lysosomes, and, in the case of an ADC, ultimately facilitate degradation-dependent release of the payload, thus leading to an increase of potency for killing of target expressing cells.

Fig. 17 shows evaluation of in vitro cell killing performed, on murine cancer cell line EMT-6 engineered to overexpress ROR1, with an scFv-IgG-based bi-epitope-reactive anti-RORl ADC (BETR-ADC™) based on anti-RORl antibody XBR1-402 and anti-RORl antibody ERR1-324. For the same total dose of ADC, the bi-epitope-reactive scFv-IgG-based anti- RORl ADC of the invention show cell killing activity on engineered EMT-6 cancer cells that is superior to the cell killing activity of individual ADCs at equivalent total concentration. Trastuzumab-PNU ADC (Tras-G3-PNU) was used as an isotype-matched control ADC.

Detailed Description of the Invention

Before the invention is described in detail, it is to be understood that this invention is not limited to the particular component parts of the devices described or process steps of the methods described as such devices and methods may vary. It is also to be understood that the terminology used herein is for purposes of describing particular embodiments only, and is not intended to be limiting. It must be noted that, as used in the specification and the appended claims, the singular forms "a", "an", and "the" include singular and/or plural referents unless the context clearly dictates otherwise. It is moreover to be understood that, in case parameter ranges are given which are delimited by numeric values, the ranges are deemed to include these limitation values.

It is further to be understood that embodiments disclosed herein are not meant to be understood as individual embodiments which would not relate to one another. Features discussed with one embodiment are meant to be disclosed also in connection with other embodiments shown herein. If, in one case, a specific feature is not disclosed with one embodiment, but with another, the skilled person would understand that does not necessarily mean that said feature is not meant to be disclosed with said other embodiment. The skilled person would understand that it is the gist of this application to disclose said feature also for the other embodiment, but that just for purposes of clarity and to keep the specification in a manageable volume this has not been done.

Furthermore, the content of the prior art documents referred to herein is incorporated by reference. This refers, particularly, for prior art documents that disclose standard or routine methods. In that case, the incorporation by reference has mainly the purpose to provide sufficient enabling disclosure, and avoid lengthy repetitions.

According to a first aspect of the invention, a multispecific antibody-based product comprising

(a) a first entity comprising an antigen-binding domain that

• binds to the same RO 1 epitope as and/or

• competes for ROR1 binding with an antibody comprising a heavy chain variable region sequence shown in SEQ ID NO. 2 and a light chain variable region sequence shown in SEQ ID NO. 3; and

(b) a second entity comprising an antigen-binding domain that binds to

• to a different ROR1 epitope than, and/or

• does not compete for binding with the antibody comprising a heavy chain variable region sequence shown in SEQ ID NO. 2 and a light chain variable region sequence shown in SEQ ID NO. 3.

As herein, the term "multispecific" means a product which has specificity for two or more different epitopes of ROR-1 antigens. In the context of the present invention, the terms "multispecific" and "multi-epitope reactive" are hence used simultaneously. In one embodiment, the multi-epitope reactive product is bi-epitope reactive.

The term "variable region", as used herein, refers to the respective regions of the heavy and light chain of an antibody, abbreviated VH and VL, (sometimes also written VH and VL, or HCVD and LCVD), as opposed to the constant domains CHI , ¾2 and CR3 of the heavy chain and CL of the light chain. The variable regions encompass the complementarity determining regions (CDRs). The term "variable domain" is used interchangeably with the term "variable region" herein.

While an antibody comprising an Fc region has specificity not only for antigen epitopes, via its VH/VL domains, but also binds, via its Fc region, to an Fc receptor. In case its VH/VL domains bind two different epitopes, such antibody will still be called "bispecific" or "bi- epitope reactive", in case its VH/VL domains bind two different epitopes, despite the fact that its actually binds another target, namely an Fc receptor. In case its VH VL domains bind only one epitope, such antibody will still be called "monospecific" or "mono-epitope reactive", in case its VH/VL domains bind two different epitopes, despite the fact that its actually binds another target, namely an Fc receptor.

According to one embodiment of the respective aspect of the invention, the product is a multispecific antibody, alternative scaffold or antibody mimetic wherein

(a) the first entity is a first antigen-binding domain that

• binds to the same ROR1 epitope as and/or

• competes for ROR1 binding with antibody ERRl-324 comprising a heavy chain variable region sequence shown in SEQ ID NO. 2 and a light chain variable region sequence shown in SEQ ID NO. 3; and

(b) the second entity is a second antigen-binding domain that

• binds to a different ROR1 epitope than, and/or

• does not compete for binding with antibody ERRl-324 comprising a heavy chain variable region sequence shown in SEQ ID NO. 2 and a light chain variable region sequence shown in SEQ ID NO. 3.

According to another embodiment of the respective aspect of the invention, the product comprises two or more antibodies, alternative scaffolds or antibody mimetics wherein

(a) the first entity is a first antibody comprising an antigen-binding domain that

• binds to the same ROR1 epitope as and/or

• competes for ROR1 binding with antibody ERRl-324 comprising a heavy chain variable region sequence shown in SEQ ID NO. 2 and a light chain variable region sequence shown in SEQ ID NO. 3; and

(b) the second entity is a second antibody comprising an antigen-binding domain that

• binds to a different ROR1 epitope than, and/or

• does not compete for binding with antibody ERRl-324 comprising a heavy chain variable region sequence shown in SEQ ID NO. 2 and a light chain variable region sequence shown in SEQ ID NO. 3.

The two embodiments discussed above are shown, in principle, in Figure 14. Both have in common that they relate to a multispecific product wherein • a first entity binds to the same ROR1 epitope as and/or competes for ROR1 binding with an antibody having SEQ ID NO. 2 (HCVD) and SEQ ID NO. 3 (LCVD) and

• a second entity binds to a different ROR1 epitope than, and/or does not compete for ROR1 binding with the antibody having SEQ ID NO. 2 (HCVD) and SEQ ID NO. 3 (LCVD).

These two entities can either be on two different antibodies, or on a single multi-specific antibody molecule.

Additionally, the invention provides bi- or multispecific antibodies targeting ROR1 as well as at least one binding domain specific for another target, for instance, but not limited to targets that recruit and/or activate cells of the immune system, like T cells or NK cells. Such other binding domains may be specific for CD3, CD16, CD32, CD56, CD64 or other markers specific for T and NK cells.

SEQ ID NOs 2 and 3 belong to an anti-RORl antibody called ERRl-324. This antibody binds a specific epitope of ROR1, including human ROR1 (hRORl). In a preferred embodiment, the first entity comprises the CDRs of ERRl-324, as given in Figure 1, i.e., the first entity comprises the following CDRs: QASQSVYGNNELA (V K CDR1), RASILTS (V K CDR2), LGGYVSQSYRAA (V K CDR3), RNGMT (V H CDR1), IITSSGDKYYATWAKG (V H CDR2), and GTVSSDI (V H CDR3).

In a preferred embodiment, the first entity comprises the variable domain sequences of ERRl-324, i.e., comprises the variable domain sequences of SEQ ID NO. 2 (HCVD) and SEQ ID NO. 3 (LCVD).

The invention shows that such multi-specific product has significant advantages over a product which only has a single epitope reactivity for the ROR1 target, like e.g. the epitope that an antibody having SEQ ID NO. 2 (HCVD) and SEQ ID NO. 3 (LCVD).

Without being bound to theory, it is conceivable that the bi-epitope reactivity may induce the formation of target-homodimers or even clusters of the target as schematically presented in Figure 16. This may increase the induction of internalization of the said product by receptor- mediated endocytosis, and therefore a more efficient transport of such bi-epitope reactive ADCs (BETR-ADC™) to intracellular lysosomes leading to higher potency for killing of target expressing cells.

According to one embodiment, the entity comprising an antigen-binding domain is at least one selected from the group consisting of an antibody, an antibody-based binding protein, a modified antibody format retaining target binding capacity, an antibody derivative or a fragment retaining target binding capacity, an alternative scaffold and/or an antibody mimetic.

According to one other embodiment, the antibody is at least one selected from the group consisting of an an antibody, an antibody-based binding protein, a bi-epitope-reactive antibody, a modified antibody format retaining target binding capacity, an antibody derivative or a fragment retaining target binding capacity, an alternative scaffold and/or an antibody mimetic.

According to one other embodiment, the antibody is at least one selected from the group consisting of an antibody, an antibody-based binding protein, a bi-epitope-reactive antibody, a modified antibody format retaining target binding capacity, an antibody derivative or a fragment retaining target binding capacity.

The term "fully human antibody" refers to an antibody, antibody-based binding protein or antigen-binding fragment that contains sequences derived from human immunoglobulin genes, such that substantially all of the heavy and light chain CDR1 and CDR2 regions are of human origin, and substantially all of the heavy and light chain FR regions 1, 2, 3, and 4 correspond to those of a human immunoglobulin sequence either with or without a limited number of somatic mutations that may be introduced into individual heavy and light chain CDR1 and CDR2 and FR1, 2, 3, and 4 variable domain sequences .

The terms "antibody", "antibody-based binding protein", "modified antibody format retaining target binding capacity", "antibody derivative or fragment retaining target binding capacity" refers to polypeptide chain(s) which exhibit a strong monovalent, bivalent or polyvalent binding to a given antigen, epitope or epitopes. Antibodies, antibody-based binding proteins and antigen-binding fragments used in the invention can be generated using any suitable technology, e.g., hybridoma technology, ribosome display, phage display, gene shuffling libraries, semi-synthetic or fully synthetic libraries or combinations thereof. Antibodies, antibody-based binding proteins and antigen-binding fragments of the invention include intact antibodies and antibody fragments or antigen-binding fragments that contain the antigen-binding portions of an intact antibody and retain the capacity to bind the cognate antigen. Unless otherwise specified herein, all peptide sequences, including all antibody and antigen-binding fragment sequences are referred to in N -> C order.

An intact antibody typically comprises at least two heavy (H) chains (about 50-70 kD) and two light (L) chains (about 25 kD) inter-connected by disulfide bonds. The recognized immunoglobulin genes encoding antibody chains include the kappa, lambda, alpha, gamma, delta, epsilon, and mu constant region genes, as well as the myriad immunoglobulin variable region genes. Light chains are classified as either kappa or lambda. Heavy chains are classified as gamma, mu, alpha, delta, or epsilon, which in turn define the immunoglobulin classes, IgG, IgM, IgA, IgD and IgE, respectively. Each heavy chain of an antibody is comprised of a heavy chain variable region (VH) and a heavy chain constant region. In the case of IgG, the heavy chain constant region is comprised of three domains, CHI, CH2 and CH3. Each light chain is comprised of a light chain variable region (VL) and a light chain constant region. The light chain constant region is comprised of one domain, CL. The variable regions of the heavy and light chains contain a binding domain that interacts with an antigen. The constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system and the first component (Clq) of the classical complement system. Monoclonal antibodies (niAbs) consist of identical antibodies molecules.

The VH and VL regions of an antibody can be further subdivided into regions of hypervariability, also termed complementarity-determining regions (CDRs), which are interspersed with the more conserved framework regions (FRs). Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxyl-terminus in the following order: FRl, CDRl, FR2, CDR2, FR3, CDR3, FR4. The locations of CDR and FR regions and a numbering system have been defined, e.g., the IMGT system (Lefranc MP et al., 2015), or the Kabat numbering scheme. Antibodies, antibody-based binding proteins and antigen-binding fragments of the invention also encompass single chain antibodies. The term "single chain antibody" refers to a polypeptide comprising a VH domain and a VL domain in polypeptide linkage, generally linked via a spacer peptide, and which may comprise additional domains or amino acid sequences at the amino- and/or carboxyl-termini. For example, a single-chain antibody may comprise a tether segment for linking to the encoding polynucleotide. As an example, a single chain variable region fragment (scFv) is a single-chain antibody. Compared to the VL and VH domains of the Fv fragment that are coded for by separate genes, a scFv has the two domains joined (e.g., via recombinant methods) by a synthetic linker. This enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules.

Examples of antibody-based binding proteins are polypeptides in which the binding domains of the antibodies are combined with other polypeptides or polypeptide domains, e.g. alternative molecular scaffolds, Fc-regions, other functional or binding domains of other polypeptides or antibodies resulting in molecules with addition binding properties, e.g. bi- or multispecific proteins or antibodies. Such polypeptides can create an arrangement of binding or functional domains normally not found in naturally occurring antibodies or antibody fragments.

Examples of antigen-binding fragments include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CHI domains; (ii) a F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CHI domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an intact antibody; (v) disulfide stabilized Fvs (dsFvs) which have an interchain disulfide bond engineered between structurally conserved framework regions; (vi) a single domain antibody (dAb) which consists of a VH or VL domain (see, e.g., Ward et al., Nature 341:544-546, 1989); and (vii) an isolated complementarity determining region (CDR) as a linear or cyclic peptide.

Antigen-binding fragments of the present invention also encompass single domain antigen- binding units that have a camelid scaffold. Animals in the camelid family include camels, llamas, and alpacas. Camelids produce functional antibodies devoid of light chains. The heavy chain variable (VH) domain folds autonomously and functions independently as an antigen-binding unit. Its binding surface involves only three CDRs as compared to the six CDRs in classical antigen-binding molecules (Fabs) or single chain variable fragments (scFvs). Camelid antibodies are capable of attaining binding affinities comparable to those of conventional antibodies.

The terms "alternative scaffold" and "antibody mimetic" refer to proteins not belonging to the immunoglobulin family, and even non-proteins such as aptamers, or synthetic polymers. Some types have an antibody-like beta-sheet structure. Potential advantages of "antibody mimetics" or "alternative scaffolds" over antibodies are better solubility, higher tissue penetration, higher stability towards heat and enzymes, and comparatively low production costs.

Some antibody mimetics can be provided in large libraries, which offer specific binding candidates against every conceivable target. Just like with antibodies, target specific antibody mimetics can be developed by use of High Throughput Screening (HTS) technologies as well as with established display technologies, just like phage display, bacterial display, yeast or mammalian display. Currently developed antibody mimetics encompass, for example, ankyrin repeat proteins (called DARPins), C-type lectins, A- domain proteins of S. aureus, transferrins, lipocalins, 10th type III domains of fibronectin, Kunitz domain protease inhibitors, ubiquitin derived binders (called affilins), gamma crystallin derived binders, cysteine knots or knottins, thioredoxin A scaffold based binders, nucleic acid aptamers, artificial antibodies produced by molecular imprinting of polymers, peptide libraries from bacterial genomes, SH-3 domains, stradobodies, "A domains" of membrane receptors stabilised by disulfide bonds and Ca2+, CTLA4-based compounds, Fyn SH3, and aptamers (oligonucleic acid or peptide molecules that bind to a specific target molecules)

The anti-RORl antibodies, antibody-based binding proteins and antigen-binding fragments described herein can be produced by enzymatic or chemical modification of the intact antibodies, or synthesized de novo using recombinant DNA methodologies. Methods for generating these antibodies, antibody-based binding proteins and antigen-binding molecules are all well known in the art. In particular, scFv antibodies can be obtained using methods described in, e.g., Bird et al, Science 242:423-426, 1988; and Huston et al, Proc. Natl. Acad. Sci. USA 85:5879-5883, 1988. Fv antibody fragments can be generated as described in Skerra and Pluckthun, Science 240:1038-41, 1988. Disulfide-stabilized Fv fragments (dsFvs) can be made using methods described in, e.g., Reiter et al., Int. J. Cancer 67:113-23, 1996. Similarly, single domain antibodies (dAbs) can be produced by a variety of methods described in, e.g., Ward et al., Nature 341 :544-546, 1989; and Cai and Garen, Proc. Natl. Acad. Sci. USA 93:6280-85, 1996. Camelid single domain antibodies can be produced using methods well known in the art, e.g., Dumoulin et al., Nat. Struct. Biol. 11 :500-515, 2002; Ghahroudi et al., FEBS Letters 414:521-526, 1997; and Bond et al., J. Mol. Biol. 332:643-55, 2003. Other types of antigen-binding fragments (e.g., Fab, F(ab')2 or Fd fragments) can also be readily produced with routinely practiced immunology methods. See, e.g., Harlow & Lane, Using Antibodies, A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, 1998.

The anti-RORl antibodies, antibody-based binding proteins or antigen-binding fragments of the invention can be produced by any suitable technique, for example, using any suitable eukaryotic or non-eukaryotic expression system. In certain embodiments, the antibody, antibody-based binding protein or antigen-binding fragment is produced using a mammalian expression system. Some specific techniques for generating the antibodies, antibody-based binding proteins or antigen-binding fragments or antigen-binding fragments of the invention are exemplified herein. In some embodiments, the antibodies, antibody-based binding proteins or antigen-binding fragments of the invention can be produced using a suitable non- eukaryotic expression system such as a bacterial expression system. Bacterial expression systems can be used to produce fragments such as a F(ab)2, Fv, scFv, IgGACH2, F(ab')2, scFv2CH3, Fab, VL, VH, scFv4, scFv3, scFv2, dsFv, Fv, scFv-Fc, (scFv)2, and diabodies. Techniques for altering DNA coding sequences to produce such fragments are known in the art.

According to one preferred embodiment, the first and/or the second antibody, alternative scaffold or antibody mimetic is monospecific or mono epitope-reactive.

According to one embodiment of the respective aspect of the invention, the second antibody, alternative scaffold or antibody mimetic, or the second antigen-binding domain binds to RORl, yet

• to a different epitope than, and/or

• does not compete for binding with the antibody comprising a heavy chain variable region sequence shown in SEQ ID NO. 2 and a light chain variable region sequence shown in SEQ ID NO. 3.

According to one embodiment of the respective aspect of the invention, the second antibody, alternative scaffold or antibody mimetic, or antigen-binding domain

• binds to the same ROR1 epitope as and/or

• competes for ROR1 binding with at least one antibody selected from the group consisting of a) R12, comprising a heavy chain variable region sequence shown in SEQ ID NO. 20 and a light chain variable region sequence shown in SEQ ID NO. 21,

b) ERR1-TOP43, comprising a heavy chain variable region sequence shown in SEQ ID NO. 12 and a light chain variable region sequence shown in SEQ ID NO. 13, c) ERRl-301, comprising a heavy chain variable region sequence shown in SEQ ID NO.

4 and a light chain variable region sequence shown in SEQ ID NO. 5,

d) ERRl-402, comprising a heavy chain variable region sequence shown in SEQ ID NO.

8 and a light chain variable region sequence shown in SEQ ID NO. 9, and/or e) 2A2, comprising a heavy chain variable region sequence shown in SEQ ID NO. 22 and a light chain variable region sequence shown in SEQ ID NO. 23.

According to one embodiment of the invention, ROR1 as mentioned herein is human ROR1 (hRORl).

According to one embodiment of the respective aspect of the invention, the multispecific product is in a format selected from the group consisting of

• bispecific or bi-epitope reactive scFv-Fc,

• bispecific or bi- epitope reactive scFv-IgG, and/or

• DVD-Ig.

The bispecific scFv-Fc format consists of two scFv fragments of different specificity genetically fused to an Fc fragment. The bispecific scFv-IgG format consists of an IgG shaped antibody with a given specificity with two scFv fragments of different specificity fused to the N-terminus of the VH domain of the IgG. The DVD-Ig format consists of an Ig shaped antibody with a given specificity, wherein each VL/VH pair carries, N-terminally, another VH/VL pair of different specificity.

Examples for the three formats are shown in the following table, and in Figure 15. bispecific V L 1 V H 1 V H 2 V L 2

scFv-Fc C H 2 C H 2

C H 3 C H 3

DVD-Ig V L 2 V H 2 V H 2 V L 2

v L i v H i v H i VLI

C L C H 1 c H i CL

C H 2 C H 2

C H 3 C H 3

bispecific V L 2 V H 2 V H 2 V L 2

scFv IgG v L i VHI v H i V L 1

C L C H 1 CHI CL

C H 2 C H 2

C H 3 C H 3

According to another embodiment, the first entity comprises the following CDRs:

• QASQSVYGNNELA (V L CDR1, SEQ ID NO. 69),

• RASILTS (V L CDR2, SEQ ID NO. 70),

• LGGYVSQSYRAA (V L CDR3, SEQ ID NO. 71),

• RNGMT (V H CDRl SEQ ID NO. 72),

• IITSSGDKYYATWAKG (V H CDR2, SEQ ID NO. 73), and

• GTVSSDI (V H CDR3, SEQ ID NO. 74),

These are the CDRs of antibody ERR1-324. The CDRs are comprised in a suitable protein framework so as to be capable to bind to ROR1. According to another embodiment, the second entity comprises one of the following CDR sets:

Again, the CDRs are comprised in a suitable protein framework so as to be capable to bind to ROR1.

According to another embodiment, the first entity comprises the heavy chain variable region sequence of antibody ERRl-324 shown in SEQ ID NO. 2 and the light chain variable region sequence of antibody ERRl-324 shown in SEQ ID NO. 3.

According to another embodiment, the second entity comprises at least one of the following sequence pairs:

a) the heavy chain variable region sequence of antibody R12 shown in SEQ ID NO. 20 and the light chain variable region sequence shown in SEQ ID NO. 21,

b) the heavy chain variable region sequence of antibody ERR1-TOP43 shown in SEQ ID NO. 12 and the light chain variable region sequence of antibody ERR1-TOP43 shown in SEQ ID NO. 13, c) the heavy chain variable region sequence of antibody ERR1-301 shown in SEQ ID NO. 4 and the light chain variable region sequence of antibody ERR1-301 shown in SEQ ID NO. 5,

d) the heavy chain variable region sequence of antibody ERRl-402 shown in SEQ ID NO. 8 and the light chain variable region sequence of antibody ERRl-402 shown in SEQ ID NO. 9, and/or

e) the heavy chain variable region sequence of antibody 2A2 shown in SEQ ID NO. 22 and the light chain variable region sequence of antibody 2A2 shown in SEQ ID NO. 23.

According to another aspect of the invention, an antibody drug conjugate (ADC) having the general formula A - (L)n - (T)m is provided, in which

• A is at least one antigen binding domain, antibody, alternative scaffold or antibody mimetic according to the above description,

• L is a linker,

• T is a toxin and in which n and m are integers between >1 and < 10.

According to another aspect of the invention, an antibody effector conjugate (AEC) having the general formula A - (L)n - (E)m is provided, in which

• A is at least one antigen binding domain, antibody, alternative scaffold or antibody mimetic according to the above description,

• L is a linker,

• E is a label and in which n and m are integers between >1 and < 10.

Such label can be a detectable label, can be at least one selected from the group consisting of: a fluorescent label (including a fluorescent dye or a fluorescent protein), a chromophore label, a radi *oisotope label contai *ning iodine (e.g., 125 I), galli *um ( 67 Ga), indium ( 111 I), technetium ( 99m Tc), phosphorus ( 32 P), carbon ( 14 C), tritium ( 3 H), other radioisotope (e.g., a radioactive ion), and/or a protein label such as avidin or streptavidin.

According to one embodiment of the respective aspect of the invention, the antibody is a multi-specific, preferably a bi-epitope reactive antibody according to the above description.

According to another aspect of the invention, a composition comprising at least two antibody effector conjugates (AEC) or antibody drug conjugates (ADC) according to the above description, wherein each of the two conjugates comprises one of the monospecific antibodies, alternative scaffolds or antibody mimetics according to the above description.

According to yet another embodiment of the invention, the ADC is a bi-epitope reactive ADC (abbreviated BETR-ADC™) binding to two different epitopes of the ROR1 target.

According to one embodiment of the respective aspect of the invention, the linker is at least one selected from the group consisting of

• an oligopeptide linker

• a maleimide linker, optionally comprising cleavable spacers, that may be cleaved by changes in pH, redox potential and or specific intracellular or extracellular enzymes.

According to one embodiment, the linker has at least one of the following amino acid sequences: -LPXTGn-, -LPXAGn-, -LPXSGn-, -LAXTGn-, -LPXTGn-, -LPXTAn- or - NPQTGn-, with Gn being an oligo- or polyglycine with n being an integer between > 1 and < 21, An being an oligo-or polyalanine with n being an integer between > 1 and < 21, and X being any conceivable amino acid sequence.

Gn (also called Gly(n)) is the oligoglycin discussed elsewhere herein In a preferred embodiment its length n can be between > 1 and < 21, preferably between > 1 and < 5.

It is important to understand that, in one specific embodiment (where Streptococcus pyogenes sortase A is used, see below), the oligo-glycine (Gly) n can optionally be replaced by an oligo- alanine (Ala) n . According to one embodiment of the respective aspect of the invention, the linker is conjugated to the C-terminus of at least one subdomain of the antibody.

According to one embodiment of the respective aspect of the invention, prior to conjugation

• the antibody bears a sortase recognition tag used or conjugated to the C-terminus of at least one subdomain thereof, and

• the toxin or label comprises a glycine stretch with a length of between > 1 and < 20 glycine residues, preferably with a length of > 2 and < 5 glycine residues.

According to another embodiment, said sortase enzyme recognition motif comprises at least one of the following amino acid sequences: LPXTG, LPXAG, LPXSG, LAXTG, LPXTA or NPQTN, with X being any conceivable amino acid sequence.

The following table shows the recognition tags and the peptides derived therefrom to be part of the linker:

Engineered sortases, including but not limited to sortase A mutant 2A-9 and sortase A mutant 4S-9 from Staphylococcus aureus, are described in Dorr et al. (2014) and mutants described in Chen et al. (2011).

As background and to exemplify the general concept of sortase transpeptidation, Sortase A uses an oligo-glycine-stretch as a nucleophile to catalyze a transpeptidation by which the terminal amino group of the oligo-glycine effects a nucleophilic attack on the peptide bond joining the last two C-terminal residues of the sortase tag. This results in breakage of that peptide bond and formation of a new peptide bond between the C-terminally second-to-last residue of the sortase tag and the N-terminal glycine of the oligo-glycine peptide, i.e. resulting in a transpeptidation.

It is important to understand that, in one specific embodiment (where Streptococcus pyogenes sortase A is used, see above), the oligo-glycine (Gly)n can optionally be replaced by an oligo- alanine (Ala)n.

Prior to sortase conjugation, the sortase recognition motif may, at its C-terminus, furthermore carry other tags, like His-tags, Myc-tags or Strep-tags (see Fig. 4a of WO 2014/140317, the content of which is incorporated by reference herein). However, because the peptide bond between the 4th and 5th amino acid of the sortase tag is cleaved upon sortase A mediated conjugation, these additional tags do not appear in the conjugated product.

The sortase tag may, for example, be fused to a C-terminus of a binding protein, or to a domain or subunit thereof, by genetic fusion and co-expressed therewith. In another preferred embodiment, the sortase tag may directly be appended to the last naturally occurring C- terminal amino acid of the immunoglobulin light chains or heavy chains, which in case of the human immunoglobulin kappa light chain is the C-terminal cysteine residue, and which in the case of the human immunoglobulin IgGl heavy chain may be the C-terminal lysine residue encoded by human Fcyl cDNA. However, another preferred embodiment is also to directly append the sortase tag to the second last C-terminal glycine residue encoded by human Fcyl cDNA, because usually terminal lysine residues of antibody heavy chains are clipped off by posttranslational modification in mammalian cells. Therefore, in more than 90% of the cases naturally occurring human IgGl lacks the C-terminal lysine residues of the IgGl heavy chains.

Therefore, one preferred embodiment of the invention is to omit the C-terminal lysine amino acid residues of human IgGl heavy chain constant regions in expression constructs for sortase recognition-motif tagged Igyl heavy chains. Another preferred embodiment is to include the C-terminal lysine amino acid residues of human IgGl heavy chain constant regions in expression constructs for sortase recognition-motif tagged Igyl heavy chains.

In another preferred embodiment the sortase or oligoglycine tag may be appended to the C- terminus of a human immunoglobulin IgGl heavy chain where the C-terminal lysine residue encoded by human Fcyl cDNA is replaced by an amino acid residue other than lysine to prevent unproductive reactions of sortase with the ε -amino group of said C-terminal lysine residue leading to inter-heavy chain crosslinking.

We have described previously that in some cases (e.g. at the C-terminus of the Ig kappa light chains, see: Beerli et al. (2015) PloS One 10, el31177) it is beneficial to add additional amino acids between the C-terminus of the binding protein and the sortase tag. This has been shown to improve sortase enzyme conjugation efficiencies of payloads to the binding protein. In the case of Ig kappa light chains, it was observed that by adding 5 amino acids between the last C-terminal cysteine amino acid of the Ig kappa light chain and the sortase pentapeptide motif improved the kinetic of conjugation, so that the C-termini of Ig kappa light chains and Ig heavy chains could be conjugated with similar kinetics (see: Beerli et al. (2015) PloS One 10, el31177). Therefore, it is another preferred embodiment that optionally > 1 and < 11 amino acids are added in between the last C-terminal amino acid of a binding protein or antibody subunit and the sortase tag. In a preferred embodiment, a G n S peptide (wherein n is from 1 to 10, preferably 1 to 5) is added between the last C-terminal amino acid of a binding protein or antibody subunit and the sortase tag. Finally, in another preferred embodiment, additional amino acids between the C-terminus of the binding protein and the sortase or oligoglycine tag may beneficially be included that comprise a sequence and/or linker that is cleavable by hydrolysis, by a pH change or by a change in redox potential, or that cleavable by a non-sortase enzyme, e.g., by proteases.

According to one embodiment of the respective aspect of the invention, the toxin derivative thereof is at least one selected from the group consisting of: maytansinoids,

auristatins,

anthracyclins, preferably PNU-derived anthracyclins

calicheamicins,

tubulysins

duocarmycins

radioisotopes

liposomes comprising a toxid payload

protein toxins

taxanes, and/or

pyrrolobenzodiazepines.

In a preferred embodiment of the ADC, the toxin is selected from PNU- 159682 as described in Quintieri et al. (2005) and derivatives thereof, maytansine, monomethyl auristatin MMAE, and monomethyl auristatin MMAF. In a preferred embodiment of the ADC, the toxin, joined to the linker at its wavy line, is of formula (i), as described in WO 2016/102679:

formula (i)

In the embodiment where the toxin is of formula (i), the linker may optionally comprise an alkyldiamino group of the form NH 2 -(CH 2 ) m -NH 2 , where m > 1 and < 11, preferably m=2, such that one amino group is directly linked at the wavy line of formula (i) to form an amide bond. It is moreover preferred that the second amino group is linked to an oligopeptide linker, which is more preferably an oligoglycine.

According to one embodiment of the respective aspect of the invention, the conjugate is created by sortase-mediated conjugation of (i) an antibody carrying one or more sortase recognition tags and (ii) one or more toxins or labels carrying an oligoglycine tag.

According to another aspect of the invention, a method of producing a conjugate according to the above description is provided, which method comprises the following steps: a) providing an antibody, alternative scaffold or antibody mimetic according to the above description, which antibody carries a sortase recognition tag,

b) providing one or more toxins or labels carrying an oligoglycine tag, and

c) conjugating the antibody, alternative scaffold or antibody mimetic and the toxin or label by means of sortase-mediated

conjugation.

According to another embodiment of the invention, the use of the multispecific product according to the above description or the antibody drug conjugate according to the above description, for the treatment of a patient that is

• suffering from,

• at risk of developing, and/or

• being diagnosed for a neoplastic disease is provided.

As an alternative, a method of treating a patient suffering from, at risk of developing, and/or being diagnosed for a neoplastic disease is provided, which method comprises the administration of one or more therapeutically active doses of the multispecific product according to the above description or the antibody drug conjugate according to the above description.

According to another aspect of the invention, the neoplastic disease is a neoplastic disease characterized by expression of RORl. In one embodiment, RORl is overexpressed in said neoplastic disease.

As used herein, the term "overexpression of RORl " refers to the expression level of RORl mRNA and/or protein expressed in cells of a given tissue being elevated in comparison to the levels of RORl as measured in normal cells (free from disease) of the same type of tissue, under analogous conditions. Said RORl mRNA and/or protein expression level may be determined by a number of techniques known in the art including, but not limited to, quantitative RT-PCR, western blotting, immunohistochemistry, and suitable derivatives of the above.

According to another aspect of the invention, the neoplastic disease is at least one selected from the group consisting of sarcoma, renal cell carcinoma, breast cancer, inch triple- negative breast cancer, lung cancer, colon carcinoma, testicular cancer, ovarian cancer, pancreatic cancer, kidney cancer, gastric cancer, prostate cancer, head and neck cancer, melanoma, squamous cell carcinoma, multiple myeloma and other cancers, mesothelioma, chronic lymphoblastic leukemia, mantle cell lymphoma, non-Hodgkin lymphoma, Hodgkin lymphoma, preB acute lymphocytic leukemia, acute myeloid leukemia, multiple myeloma, and other types of leukemias and lymphomas as well as solid tumors.

According to another aspect of the invention, a pharmaceutical composition comprising the multi-specific product according to the above description, the antibody drug conjugate according to the above description together with one or more pharmaceutically acceptable ingredients, is provided.

According to another aspect of the invention, a method of killing or inhibiting the growth of a cell expressing or overexpressing RORl in vitro or in a patient is provided, which method comprises administering to the cell a pharmaceutically effective amount or dose of the multi- specific or bi-epitope reactive product according to the above description, the antibody drug conjugate according to the above description, or of the pharmaceutical composition according to the above description.

According to one embodiment, the cell expressing ROR1 is a cancer cell. Examples

While the invention has been illustrated and described in detail in the drawings and foregoing description, such illustration and description are to be considered illustrative or exemplary and not restrictive; the invention is not limited to the disclosed embodiments. Other variations to the disclosed embodiments can be understood and effected by those skilled in the art in practicing the claimed invention, from a study of the drawings, the disclosure, and the appended claims. In the claims, the word "comprising" does not exclude other elements or steps, and the indefinite article "a" or "an" does not exclude a plurality. The mere fact that certain measures are recited in mutually different dependent claims does not indicate that a combination of these measures cannot be used to advantage. Any reference signs in the claims should not be construed as limiting the scope.

All amino acid sequences disclosed herein are shown from N-terminus to C-terminus; all nucleic acid sequences disclosed herein are shown 5'->3'.

Example 1. Establishment of 63-12 cells stably expressing hRORl or hROR2

The mouse Abelson murine pre-B cell line 63-12 (Shinkai et al. (1992) Cell 68:855-67) was cultured in culture media (17.7g/L Gibco® IMDM (Life Technologies, 42200-030), 3.024g/L NaHC0 3 (Sigma-Aldrich, p. a., >99.7%), lOmL/L lOOx non-essential amino acids (Life Technologies, 11140035), 5mg/L insulin (Sigma-Aldrich, 1-5500), 3mL/L of 10% primatone RL/UF in H 2 0 (Sheffield Bioscience), and lmL/L of 50mM 2-mercaptoethanol (Sigma- Aldrich, M-3148) in H 2 0), supplemented with 2% (v/v) FCS, 100 IU/mL Pen/Strep/Fungizone (Amimed, 4-02F00-H), 200mM L-glutamine (Amimed, 5-10K00-H) and 50 μΜ 2-mercaptoethanol (Amresco, 0482) at 37°C and 7.5% C0 2 .

Cells were engineered to overexpress hRORl and hROR2 by transposition as follows: cells were centrifuged (6min, 1200rpm, 4°C) and resuspended in RPMI-1640 media (5x10 6 cells/mL). 400 μΐ, of cell suspension was then added to 400 μΐ of RPMI containing 10μg of transposable vector pPB-PGK-Puro-RORl (directing co-expression of full-length ROR1 (NP 005003.2) and the puromycin-resistance gene), or 10μg of transposable vector pPB- PGK-Puro-ROR2 (directing co-expression of full-length ROR2 (NP 004551.2) and the puromycin-resistance gene), along with 10μg of transposase-containing vector pCDNA3.1_hy_mPB. DNA/63-12 cell mixtures were transferred to electroporation cuvettes (0.4cm-gap, 165-2088, BioRad, Cressier, Switzerland) and electroporated using the Biorad Gene Pulser II with capacitance extender at 300V and 950μΡ. Then, cells were incubated for 5-10min at room temperature. Following the incubation, cells were centrifuged at 1200rpm for 6min (4°C), washed once and subsequently resuspended in aqueous culture media (17.7g/L Gibco® IMDM (Life Technologies, 42200-030), 3.024g/L NaHC0 3 (Sigma- Aldrich, p. a., >99.7%), lOmL/L lOOx non-essential amino acids (Life Technologies, 11140035), 5mg/L insulin (Sigma-Aldrich, 1-5500), 3mL/L of 10% primatone RL/UF in H 2 0 (Sheffield Bioscience), and 1 mL/L of 50mM 2-mercaptoethanol (Sigma-Aldrich, M-3148) in H 2 0), supplemented with 2% (v/v) FCS, 100 IU/mL Pen/Strep/Fungizone (Amimed, 4- 02F00-H), 200mM L-glutamine (Amimed, 5-10K00-H) and 50μΜ 2-mercaptoethanol (Amresco, 0482). After two days incubation at 37°C in a humidified incubator at 5% C0 2 atmosphere, cell pools stably expressing hRORl or hROR2 were selected by adding 2μg/mL puromycin (Sigma-Aldrich, P8833).

After 4 to 5 days, hRORl or hROR2 expression on engineered cells were confirmed by flow cytometry. Briefly, following trypzinization, 10 6 cells were centrifuged in FACS tubes; obtained pellets were resuspended in buffer (PBS with 2% (v/v) FCS). In the case of hRORl - engineered cells, cells were then incubated with 2A2 (mAb066 antibody targeting ROR1, final concentration 2 μg/mL) for 30min at 4°C, followed by centrifugation and washing. Cells were resuspended as previously and incubated with anti-human IgG antibody (Fc gamma- specific) PE (eBioscience, Vienna, Austria, 12-4998-82), at a 1 :100 dilution, in the dark (30min, 4°C), washed once in buffer and kept on ice until FACS sorting. For hROR2- engineered 63-12 cells, the same protocol was followed but using EPR3779 (Abeam antibody targeting ROR2; 1:100 dilution) as primary antibody and allophycocyanin-conjugated AffmiPure F(ab')2 goat anti-rabbit IgG (H+L) (Jackson Immunoresearch, 111-136-144) as secondary antibody. In the case of hRORl -engineered 63-12 cells, cells were single cell sorted into 96-well flat- bottom plates containing 200μί of supplemented culture media per well using a FACS Aria II. Plates were incubated at 37°C and clones were expanded to 6-well plates before analysis. Target expression was confirmed by flow cytometry using a FACSCalibur instrument (BD Biosciences) and FlowJo analytical software (Tree Star, Ashland, OR).

63-12, 63-12/hRORl and 63-12/hROR2 transfectants were cultured in DMEM (Invitrogen; Carlsbad, CA) supplemented with 10% (v/v) heat inactivated FBS (Thermo Scientific; Logan, UT), lOOIU/mL penicillin, and lOOmg/mL streptomycin (Invitrogen). HEK 293F cells were purchased from Invitrogen and maintained in FreeStyle Medium supplemented with 1% (v/v) heat inactivated FBS (Thermo Scientific) to support adherent culture or without FBS for suspension culture, 100 U/mL penicillin, and 100 mg/mL streptomycin (Invitrogen).

Example 2. Generation of high-complexity rabbit Fab library and reagents for screening

Construction, expression, and purification of recombinant human RORl proteins: Construction, expression, purification and biotinylation of hFc fusion proteins containing different domains of human RORl or mouse RORl were described (Yang et al., PloS One 6:e21018, 2011). For hRORl -AVI-6XHIS fusion protein, the extracellular domain of human RORl (24-403) was PCR amplified with primers pCEP4-hRORl-F and pCEP4-hRORl-Avi tag-R (note that the AVI tag was introduced to the C terminus of RORl by primer pCEP4- hRORl-Avi tag-R), followed by extension PCR with primers pCEP4-signal-F-KpnI and pCEP4-6HIS-R-XhoI to add a signal peptide and 6XHIS tag to the N and C terminus separately before cloning into pCEP4 via KpnVXhol. This construct was then transiently transfected into HEK 293F cells (Invitrogen) using 293fectin (Invitrogen), and the protein was purified by Immobilized Metal Ion Affinity Chromatography using a 1-mL HisTrap column (GE Healthcare) as described in Kwong and Rader, Curr Protoc Protein Sci Chapter 6:Unit 6 10, 2009. The quality and quantity of purified hRORl -AVI-6XHIS was analyzed by SDS-PAGE and A 2 0 absorbance, respectively. Subsequently, the fusion protein was biotinylated by BirA enzyme kit from Avidity (Aurora, Colorado) following the protocol. Briefly, 2 mg ROR1-AVI-6XHIS at 40μΜ in lOmM Tris-HCl (pH 8) was biotinylated in the presence of biotin using 10 μg BirA after incubation for 30min at 37°C, followed by purification again using a 1-mL HisTrap column (GE Healthcare) as described above.

pCEP4-hRORl-F:

5'-atcctgtttctcgtagctgctgcaactggagcacactccgcccggggcgccgccgcc cag-3' (SEQ ID NO:26); pCEP4-hRORl- Avi-tag-R:

5'-ccactcgatcttctgggcctcgaagatgtcgttcaggccctccatcttgttcttctc ctt-3' (SEQ ID NO:27);

pCEP4-signal-F-KpnI:

5' gctgggtaccggcgcgccaccatggactggacttggagaatcctgtttctcgtagctgct -3' (SEQ ID NO:28);

pCEP4-6HIS-R-XhoI:

5'-gccggcctcgagtcagtgatggtgatggtggtgctcgtgccactcgatcttctgggc ctc-3' (SEQ ID NO:29). Construction, expression, and purification of recombinant human ROR1 (hRORl-His) and human ROR2 (hROR2-His) proteins: hRORl-His was PCR-amplified with primers

SP-hRORl_F

(5'gctgggtaccggcgcgccaccatggactggacttggagaatcctgtttctcgtagct gctgcaactggagcacactccgcccggggcgccgccgcccag

3') (SEQ ID NO:50) and

hRORl-His_R

(5' cggcctcgagtcagtgatggtgatggtggtgctccatcttgttcttctcctt 3') (SEQ ID NO: 30) using pCEP4-hFc-hRORl (Yang et al, PloS One 6:e21018, 2011) as template, while hROR2- His was PCR-amplified with primers

SP-hROR2 F

(gctgggtaccggcgcgccaccatggactggacttggagaatcctgtttctcgtagctgc tgcaactggagcacactccgaagtggaggttctggatccg)

(SEQ ID NO:31) and

hROR2-His_R

(cggcctcgagtcagtgatggtgatggtggtgccccatcttgctgctgtctcg) (SEQ ID NO:32) using pCEP4-hFc-hROR2 as template. Then they are cloned into pCEP4 (Invitrogen) separately via KpnVXhol. These constructs were then separately and transiently transfected into HEK 293F cells (Invitrogen) using 293fectin (Invitrogen), and the corresponding proteins were purified by Immobilized Metal Ion Affinity Chromatography using a 1-mL HisTrap column (GE Healthcare) as described in Kwong and Rader, Curr Protoc Protein Sci Chapter 6:Unit 6 10, 2009. The quality and quantity of purified hRORl-His and hROR2-His were analyzed by SDS-PAGE and A 280 absorbance, respectively.

Generation and selection of naive chimeric rabbit/human Fab libraries: All rabbit handling was carried out by veterinary personnel at Pocono Rabbit Farm & Laboratory (Canadensis, PA) or R & R Research (Stanwood, WA). A total of nine rabbits (ages 3-4 months) were used. Five of these rabbits were of the New Zealand White (NZW) strain, with three obtained from Pocono Rabbit Farm & Laboratory (Canadensis, PA) and two obtained from R & R Research (Stanwood, WA). Four b9 wild-type rabbits were derived from a separate R & R Research colony that originated from a pedigreed colony developed and characterized at the National Institute of Allergy and Infectious Diseases (NIAID) (McCartney-Francis et al., Proc. Natl. Acad. Sci. U S A 81 :1794-1798, 1984; and Popkov et al., J. Mol. Biol. 325:325-335, 2003. Spleen and bone marrow from each rabbit were collected and processed for total RNA preparation and RT-PCR amplification of rabbit V K , V^, and VH encoding sequences using established protocols (Rader, Methods Mol Biol 525:101-128, xiv, 2009. Rabbit (rb) V K /human (hu) C K /rbVn and rbVx/huCx/rbNH segments, respectively, were assembled in one fusion step based on 3 -fragment overlap extension PCR. Note that the VL derived from b9 rabbits were also assembled with VH from NZW rabbits. The Fab-encoding fragments were digested with Sfil and ligated with ^l-treated phage display vector pC3C (Hofer et al., J Immunol Meth 318:75-87, 2007) at 16°C for 24 h. Subsequently, 15 μg purified pC3C- rbV K /hC K /rbVH ligated products were transformed into E. coli strain SR320 (a kind gift from Dr. Sachdev S. Sidhu, University of Toronto, Toronto, Ontario, Canada) by 30 separate electroporations (each using 0.5 μg DNAin 50 μΐ electrocompetent cells) and yielded 7.5xl0 9 independent transformants for library κ. For library λ, 4.8 x 10 9 independent transformants were obtained using the same procedure. Using VCSM13 helper phage (Stratagene; La Jolla, CA), the phagemid libraries were converted to phage libraries and stored at -80°C. Phage library κ and library λ were re-amplified using XL 1 -Blue (Stratagene) or ER2738 (Lucigen) and mixed equally before four rounds of panning against biotinylated hFc-hRORl or hRORl-AVI-6HIS. During the panning, 5μg/mL antigen was pre-incubated with streptavidin coated magnetic beads (Dynabeads MyOne Streptavidin CI ; Invitrogen) at 37 C for 30min and then binders from the phage library were captured in the presence of lmg/mL unspecific polyclonal human IgG (Thermo Scientific) when hFc-RORl was used. Starting from the third round of panning, the input phage was negatively depleted by incubation with empty beads before selection against antigen-loaded beads. Following selection, supernatants of IPTG- induced bacterial clones were analyzed by ELISA and by flow cytometry. Repeated clones were identified by DNA Γι¾ε πηίη¾ with AM, and the VL and VH sequences of unique clones were determined by DNA sequencing (Figure 1).

Example 3. Expression and purification of chimeric rabbit/human Fab and full- length IgGl antibodies

Construction, expression, and purification of chimeric rabbit/human Fab and IgGl: MAb XBRl-402 in chimeric rabbit/human Fab format was cloned into E. coli expression plasmid pC3C-His and expressed and purified as described in Kwong and Rader, Curr Protoc Protein Sci Chapter 6:Unit 6 10, 2009. For the expression of mAb XBRl-402 in chimeric rabbit/human IgGl format, the previously described vector PIGG-R11 was used (Yang et al., PloS One 6:e21018, 2011). The V H encoding sequence of Fab XBRl-402 was PCR amplified using primers XBR1 -402 VH F and XBR1-402_VH _R, and cloned via ApallSacl into PIGG-R11. Then the light chain encoding sequence of XBR1-402 was PCR amplified using primers XBR1-402_\_F and LEAD-B, and cloned via HindllllXbal into PIGG-R11 with the corresponding heavy chain encoding sequence. Note that an internal Apal site in FR4 of V H encoding sequences of Fab XBR1-402 was removed by silent mutation in primer XBR1- 402_VH_R. In addition, we changed a TAG stop codon, which was suppressed during selection in E. coli strain XLl-Blue, to CAG (glutamine) encoding the first amino acid of native V H (Figure 1) with primer XBR1-402_VH_F. The resulting PIGG-XBR1-402 plasmid was transiently transfected into HEK 293F cells (Invitrogen) using 293fectin (Invitrogen), and the protein purified with a 1-mL recombinant Protein A HiTrap column (GE Healthcare, Piscataway, NJ) as described (Yang et al., PloS One 6:e21018, 2011; and Yang and Rader, Methods Mol Biol 901:209-232, 2012). The quality and quantity of purified IgGl were analyzed by SDS-PAGE and A 280 absorbance, respectively.

All the other mAbs in chimeric rabbit/human Fab format were cloned into E. coli expression plasmid pETlla and expressed and purified as described (Yang et al., PloS One 6:e21018, 2011). For the expression of mAbs ERR1-324, ERR1-TOP43 and ERR1-TOP54 in chimeric rabbit/human IgGl format, pCEP4 (Invitrogen) was used to clone the heavy chain and light chain separately. For heavy chain, a gBlock containing a heavy-chain signal peptide encoding sequence, V H of ERR2-302 (a mAb to hROR2) and CHI (1-49) of human IgGl was synthesized by IDT (San Jose, CA) and amplified with primers KpnI/AscI-Signal and CH1- internal/overlap-R, and fused to CHI (50-88)-CH2-CH3 amplified from PIGG with primers CHl-internal/overlap-F and HC-CH3-R-XhoI by overlap extension PCR with primers KpnI/AscI-Signal and HC-CH3-R-XhoI, and then cloned into pCEP4 by Ascl/Xhol. Note that a Ehel site was introduced into CHI at Ala by synonymous mutation when the gBlock was synthesized. Consequently, this construct served as vector to clone the heavy chains of other mAbs by replacing the V H using Ascl/Ehel: V H of ERRl-324, ERR1-TOP43 and ERR1- TOP54 were amplified with forward primer ERRl-324 HC-F, ERR1-TOP43 HC-F and ERR1-TOP54 HC-F and reverse primer VH-CHl-R-Ehel separately, followed by extension PCR to add the signal peptide with primer KpnI/AscI-Signal and VH-CHl-R-Ehel. Then, each V H was inserted into the vector by Ascl/Ehel. For light chain cloning, while lambda light chains of ERR1-TOP43 and ERR1-TOP54 were amplified with primers ERR1-TOP43 LC-F and ERR1-TOP54 LC-F separately combined with LC-R-XhoI, kappa light chains of ERRl-324 was amplified with primers ERRl-324 KC-F and KC-R-XhoI. Then, a signal peptide encoding sequence was added by extension PCR with forward primer Kpnl/Ascl- Signal and reverse primer LC-R-XhoI or KC-R-XhoI. Subsequently, each light chain PCR product was cloned into pCEP4 by AscllXhol. The resulting constructs containing heavy chain or light chain for each IgG were co-transfected transiently into HEK293F cells (Invitrogen) using 293fectin (Invitrogen), and the corresponding proteins were purified with a 1-mL recombinant Protein A HiTrap column (GE Healthcare, Piscataway, NJ) as described (Yang et al., PloS One 6:e21018, 2011; and Yang and Rader, Methods Mol Biol 901 :209-232, 2012). The quality and quantity of purified IgGl was analyzed by SDS-PAGE and A 280 absorbance, respectively.

Table 1. Primer se uences for clonin antibod se uences

Example 4. Examination of antibody binding activities

ELISA: For ELISA (Figure 2), each well of a 96-well Costar 3690 plate (Corning, Corning, NY) was coated with lOOng anti-human IgGl Fc in 25μΙ, coating buffer (0.1M Na 2 C0 3 , 0.1M NaHC0 3 , pH 9.6) for lh at 37°C. After blocking with 150μΕ 3% (w/v) BSA/TBS for lh at 37°C, hFc-hRORl or hFc-mRORl was captured following incubation at 100ng/50μL for lh at 37°C. Then 100ng/50μL of Fab was applied in each well at 37°C. 2h later, 50μΕ of a 1:1000 dilution of a mouse anti-His tag mAb conjugated to horseradish peroxidase (HRP) (R&D Systems, Minneapolis, MN) in 1% (w/v) BSA/TBS was used to detect the Fab. To determine the epitopes (Figure 4), hFc fusion proteins containing different domains of hRORl were coated directly, followed by incubation with Fab before detection by mouse anti-His tag mAb conjugated to HRP. Washing with PBS was repeated and colorimetric detection was performed using 2,2'-azino-bis (3-ethylbenzthiazoline)-6-sulfonic acid (Roche) as substrate according to the manufacturer's directions. The absorbance was measured at 405nm using a SpectraMax M5 microplate reader (Molecular Devices; Sunnyvale, CA) and SoftMax Pro software (Molecular Devices).

Flow cytometry: Cells were stained using standard flow cytometry methodology. Briefly, for purified anti-RORl Fab (Figure 3), 0.1-1 x 10 6 cells were stained with 1000ng/10(^L of Fab on ice for lh. After washing twice with ice-cold flow cytometry buffer (PBS containing 1% (v/v) BSA, 0.1% sodium azide and ImM EDTA), the cells were incubated with a 1 : 1000 dilution of a mouse anti-His tag mAb conjugated to Alexa Fluor 488 (Qiagen) in ΙΟΟμΙ, flow cytometry buffer on ice for 30min.

Surface plasmon resonance: Surface plasmon resonance for the measurement of the affinities of all Fabs to hFc-hRORl and for epitope mapping studies were performed on a Biacore XI 00 instrument using Biacore reagents and software (GE Healthcare, Piscataway, NJ). Anti- Human IgG (Fc) antibody was immobilized on a CM5 sensor chip following the instruction of Human Antibody Capture Kit (GE Healthcare, Piscataway, NJ). Then, hFc-hRORl fusion proteins were captured at certain density (indicated in Figure 5). The sensor chip included an empty flow cell for instantaneous background depletion. All binding assays used lx HBS- EP+ running buffer (lOmM HEPES, 150mM NaCl, 3mM EDTA (pH 7.4), and 0.05% (v/v) Surfactant P20) and a flow rate of 30mL/min. For affinity measurements, all Fabs were injected at five different concentrations (dilution factor was 2), and the lowest concentration was tested in duplicates (the highest concentrations for each Fab are indicated in Figure 6B). The sensor chip was regenerated with MgCl 2 from the Human Antibody Capture Kit without any loss of binding capacity. Calculation of association (k on ) and dissociation (Ar 0f ) rate constants was based on a 1 :1 Langmuir binding model. The equilibrium dissociation constant (K d ) was calculated from k 0 I k on . For epitope mapping studies, each Fab was prepared at 500nM alone in lx HBS-EP+ running buffer and then injected in order as indicated in Figure 5. The results in Figure 5 show that ERRl-324 is able to bind concurrently with ERRl- Top43, i.e., they do not compete for binding. ERRl-Top43 hinders concurrent binding of ERR+-306, XBR1-402 and ERR1-Top40.

Example 5. Expression of purified, recombinant strep-tagged human ROR1 and human twin strep-tagged human ROR2

StrepII-tagged human ROR1 -extracellular domain was produced as follows: the nucleotide sequence encoding the extracellular domain of human ROR1 (NP 005003.2) was N- terminally fused to a signal sequence (MNFGLRLIFLVLTLKGVQC) and C-terminally fused with a sequence encoding a peptide comprising a strepll-tag (WSHPQFEK). The entire nucleotide sequences with flanking 5'NotI and 3'HindIII sites were produced by total gene synthesis (GenScript, Piscataway, USA), assembled in the proprietary mammalian expression vector pEvi5 by Evitria AG (Schlieren, Switzerland) and verified by DNA sequencing.

Expression of the proteins was performed in suspension-adapted CHO Kl. Supernatants from pools of transfected CHO Kl cells were harvested by centrifugation and sterile filtered (0.2μιη) before FPLC-based affinity purification using StrepTactin columns (IBA GmbH, Goettingen, Germany).

Recombinant human twin strep-tagged ROR2 (NP 004551.2; twin strep sequence WSHPQFEKGGGSGGGSGGSAWSHPQFEKGS) was expressed and purified in-house according to the following protocol: the EBNA expression vector pCB14b-ROR2-ECD- TwinStrep, directing expression of ROR2 extracellular domain (ECD), C-terminally tagged with a TwinStep tag, was transfected into HEK293T using Lipofectamine® LTX with PLUS™ Reagent (Thermo Fisher Scientific, 15388100). Following a 1-day incubation (37°C, 5% C0 2 , growth media: Dulbecco's Modified Eagle Medium (DMEM) High Glucose (4.5g/L) with L-Glutamine with 10% (v/v) Fetal Calf Serum (FCS), 100 IU/mL of Pen-Strep- Fungizone and 2mM L-glutamine (all Bioconcept)), cells were expanded under selection conditions ^g/mL of puromycin (Sigma-Aldrich, P8833-25 mg stock at 2 mg/niL)). Cells were split and further expanded (37°C, 5% C0 2 ); once confluency was reached, tissue culture dishes were coated with 20μg/ml poly-L-Lysine (Sigma-Aldrich, P1524) for 2h at 37°C and washed twice with PBS. Then, cells were trypsinized, washed with PBS and split 1:3 onto poly-L-lysine-coated plates. Again after reaching confluency, cells were washed with PBS followed by with media replacement using production media (DMEM/F-12, Gibco/Thermo Fisher Scientific, 31330-03) supplemented with ^g/mL puromycin (Sigma-Aldrich, P8833), lOOIU/mL of Pen-Strep-Fungizone (Bioconcept, 4-02F00-H), 16^g/mL of N-acetyl-L- cysteine (Sigma-Aldrich, A8199) and 10μg/mL of L-glutathione reduced (Sigma-Aldrich, G6529). Supernatant, harvested bi-weekly and filtered (0.22μπι) to remove cells, was stored at 4°C until purification. For purification, filtered supernatant was loaded onto a Streptactin® Superflow® high capacity cartridge (IBA, Gottingen, Germany, 2-1238-001) column; purification and elution was performed according to the manufacturer's protocol on an AEKTA pure (GE Healthcare). Fractions were analyzed for protein purity and integrity by SDS-PAGE. Protein-containing fractions were mixed and subjected to buffer exchange using Amicon filtration units (Millipore, Schaffhausen, Switzerland) to reach a dilution of >1 :100 in PBS, and then sterile filtered using a low retention filter (0.20μηι, Carl Roth, Karlsruhe, Germany, PA49.1).

Example 6. Expression of purified, recombinant anti-human RORl and isotype control antibodies

Expression vectors: Antibody variable region coding regions were produced by total gene synthesis (GenScript) using MNFGLRLIFLVLTLKGVQC as leader sequence, and were assembled with human IgH-γ 1 and IgL-K or IgL-λ constant regions, as applicable, in the expression vector pCB14. This vector, a derivative of the episomal mammalian expression vector pCEP4 (Invitrogen), carries the EBV replication origin, encodes the EBV nuclear antigen (EBNA-1) to permit extrachromosomal replication, and contains a puromycin selection marker in place of the original hygromycin B resistance gene.

Expression and purification of RORl antibodies: pCB14-based expression vectors were transfected into HEK293T cells using Lipofectamine® LTX Reagent with PLUS™ Reagent (Thermo Fisher Scientific, Reinach, Switzerland, 15388100); following a 1-day incubation (37°C, 5% C0 2 , growth media: Dulbecco's Modified Eagle Medium (DMEM) High Glucose (4.5g/L) with L-Glutamine with 10% (v/v) Fetal Calf Serum (FCS), lOOIU/mL of Pen-Strep- Fungizone and 2mM L-glutamine (all Bioconcept, AUschwil, Switzerland)), cells were expanded under selection conditions ^g/mL of puromycin (Sigma-Aldrich, Buchs SG, Switzerland, P8833-25mg stock at 2mg/mL)). Cells were split and further expanded (37°C, 5% C0 2 ); once confluency was reached, tissue culture dishes were coated with 20μg/ml poly- L-Lysine (Sigma-Aldrich, PI 524) for 2h at 37°C and washed twice with PBS. Then, cells were trypsinized and split 1:3 onto poly-L-lysine-coated plates. Again after reaching confluency, cells were washed with PBS followed by media replacement to production media (DMEM/F-12, Gibco/Thermo Fisher Scientific, 31330-03) supplemented with 1 μ^ηιΐ, puromycin (Sigma, P8833), 100 IU/mL of Pen-Strep-Fungizone (Bioconcept), 16^g/mL of N-acetyl-L-cysteine (Sigma- Aldrich, A8199) and 10 μg/mL of L-glutathione reduced (Sigma-Aldrich, G6529). Supernatant, harvested bi-weekly and filtered (0.22μηι) to remove cells, was stored at 4°C until purification.

For purification, filtered supernatant was loaded onto a PBS-equilibrated Protein A HiTrap column (GE Healthcare, Frankfurt am Main, Germany, 17-0405-01) or a JSR Amsphere™ Protein A column (JSR Life Sciences, Leuven, Belgium, JWT203CE) and washed with PBS; elution was performed using 0.1M glycine (pH 2.5) on an AEKTA pure (GE Healthcare). Fractions were immediately neutralized with 1M Tris-HCl buffer (pH 8.0), and analyzed for protein purity and integrity by SDS-PAGE. Protein-containing fractions were mixed and subjected to buffer exchange using Amicon filtration units (Millipore, Schaffhausen, Switzerland, UFC901008) to reach a dilution of 1 : 100 in PBS, and then sterile filtered using a low retention filter (0.20μιη, Carl Roth, Karlsruhe, Germany, PA49.1).

Isotype control antibodies were transiently expressed in CHO cells by methods known in the art and recombinant antibodies were purified by standard protein A purification from CHO cell supernatants, as known in the art. The purity and the integrity of the recombinant antibodies were analyzed by SDS-PAGE.

ERRl-324-scFv HC: C-terminal

scFv-Ig SEQ ID N0.15 4.3

(mAbl21) LPETG-Strep-GS

Knob: LPETG-Strep-

XBRl-402-scFv-324-scFv scFv-Fc- Knob: SEQ ID N0.16 GS

3.3

(mAbl l3) KIH Hole: SEQ ID N0.17 Hole: LPETG-Strep- GS

HC: LPETG-

ERR1-324-LL-R12 HC: SEQ ID NO. 18 T inStrep-GS

DvD-Ig 1.7

(mAb213) LC: SEQ ID NO. 19 LC: G 5 SLPETG- TwinStrep-GS

HC: SEQ ID NO.20 HC: LPETG-Strep

R12 (mAb067) IgG 5.7

LC: SEQ ID N0.21 LC: GjSLPETG-Strep

HC: SEQ ID N0.22 HC: LPETG-Strep

2A2 (mAb066) IgG 8.0

LC: SEQ ID N0.23 LC: G 5 SLPETG-Strep

HC: SEQ ID N0.24 HC: LPETG-Strep

Ac 10 (mAb046) IgG 7.9

LC: SEQ ID N0.25 LC: G 5 SLPETG-Strep

HC: SEQ ID N0.26 HC: LPETG-Strep

Trastuzumab (mAb042) IgG 2.7

LC: SEQ ID N0.27 LC: G 5 SLPETG-Strep

HC: LPETG- scFv-Ig 324 4-2 HC: SEQ ID N0.67

scFv-IgG TwinStrep 2.7

(mAb351) LC: SEQ ID NO.68

LC: G 5 SLPETG-Strep

Example 7. mAb ROR1 and ROR2-binding - characterization by ELISA

Each well of a 96- well plate was coated with 100 μΐ, of 2 μ^ΓηΙ, strep-tagged human ROR1 or ROR2 (from Example 5) in 0.1 M bicarbonate coating buffer (pH 9.6), and incubated for 12h at 4°C.

After blocking with 150 μΐ, of 3% (w/v) bovine serum albumin (BSA)/TBS for lh at 37°C, the following antibodies were added to a well within each plate at a concentration of 0.5 μg mL, and serially diluted (dilution factor 4) with 1% (w/v) BSA/TBS, before incubation for lh at 37°C: ERR1-301 (mAb027), XBR1-402 (mAb031), ERRl-306 (mAb033), ERRl-324 (mAb034), ERRl-403 (mAb035) and ERRl-Top43 (mAb036). HRP-conjugated F(ab')2 anti- human FC-gamma (Jackson Immunoresearch, 109-036-008) was then added at a 1 :20'000 dilution, 100 μΐ per well, and incubated for lh at 37°C prior to detection using an Spark 10M plate reader (Tecan). As shown in Figure 7, the anti-human ROR1 antibodies bind human ROR1 (panel A) and are not cross-reactive with human ROR2 (panel B).

Example 8. FACS staining of cells for hRORl expression

5xl0 5 of each cell type were added per well to 96-well plates. Plates were centrifuged (3min, 1300rpm) with re-suspension in buffer (PBS supplemented with 2% (v/v) of FCS). 2A2 (mAb066) was added to each well to reach a concentration of 2μg/mL. Plates were then incubated on ice for 30 min and washed with 200μΙ, of buffer prior to resuspension in 200μί of buffer supplemented with anti-human IgG antibody (Fc gamma-specific) PE (eBioscience 12-4998-82) at a 1:250 dilution. Following 30min incubation on ice and one washing, cells were analyzed using a FACSCalibur instrument (BD Biosciences) and data was analyzed using Flow Jo analytical software (Tree Star, Ashland, OR).

Figure 8 shows the FACS analysis data of RORl -positive human ALL cell lines 697, human triple-negative breast cancer cell lines MDA-MB-468 and HS-578T, human lung cancer cell line A549, human colon cancer cell line HT-29, and RORl -negative human breast cancer cell line T47D as a negative control.

Example 9. Conjugation of mAbs with glycine-modified toxins to form ADCs using SMAC-technology™

Sortase A. Recombinant and affinity purified Sortase A enzyme from Staphylococcus aureus was produced in E. coli as disclosed in WO2014140317A1.

Generation of glycine-modified toxins. In order to generate SMAC-technology™ conjugated ADCs with pentaglycine-modified EDA-anthracycline derivative (G5-P U) was manufactured by Concortis (Figure 9). Triglycine-modified EDA-anthracycline derivative (G3-PNU) differs from G5-PNU in that it is modified with only 3 glycine residues instead of 5. The identity and the purity of the pentaglycine-modified and triglycine-modified toxin was confirmed by mass-spectrometry and HPLC. The Gly 5 -modified and Gly 3 -modified toxin exhibited > 95% purity, as gauged by the single peak in the HPLC chromatogram.

Sortase-mediated antibody conjugation. The above-mentioned toxin was conjugated to anti- ROR1 antibodies as per Table 3 by incubating LPETG-tagged mAbs [ΙΟμΜ] with glycine modified toxin [200μΜ] and 3 μΜ Sortase A in the listed conjugation buffer for 3.5h at 25°C. The reaction was stopped by passing it through an rProtein A GraviTrap column (BioRad). Bound conjugate was eluted with 5 column volumes of elution buffer (0.1 M glycine pH 2.5, 50 nM NaCl), with 1 column volume fractions collected into tubes containing 25% v/v 1M HEPES pH 8 to neutralise the acid. Protein containing fractions were pooled and formulated in the formulation buffer of Table 3 using a ZebaSpin desalting column. ADC analytics. DAR was assessed by Reverse Phase Chromatography performed on a Polymer Labs PLRP 2.1mm x 5cm, 5μηι column run at lmL/min/80°C with a 25 minute linear gradient between 0.05 and 0.1% TFA/H 2 0 and 0.04 to 0.1% TFA/CH 3 CN. Samples were first reduced by incubation with DTT at pH 8.0 at 37°C for 15 minutes. The DAR determined by Reverse Phase Chromatography is summarized in Table 3 below.

Table 3: Analytical summary of ADCs manufactured in this study. DAR, drug-to- antibody ratio. ND, not determined.

(adc394)

From these analyses it can be concluded that the SMAC-technology conjugation has proceeded at high efficiency resulting in overall average DARs in the range of ca. 3.5 to 4.0 for IgG-format anti-RORl antibody-toxin combinations.

Example 10. In vitro cytotoxicity of single and mixtures of anti-RORl antibody-based ADCs on human 697 B cell precursor leukemia cells

Cytotoxicity of 50:50 (by weight) mixtures of ERR1-324-G5-PNU with further anti-RORl ADCs was investigated using human cell line 697, and compared to the cytotoxicity of the individual ADCs.

For this, 2.5 xlO 4 697 cells per well were plated on 96-well plates (excluding edge wells, which contained water) in 75 \xL RPMI supplemented with 10% by vol. FCS, lOOIU/ml Pen- Strep-Fungizone and 2mM L-Glutamine and were grown at 37°C in a humidified incubator at 7.5% C0 2 atmosphere. After 1-day incubation, each ADC or ADC mixture was added to respective wells in an amount of 25μΙ. of 3.5 -fold serial dilutions in growth medium (resulting in final ADC or ADC mixture concentrations from 20μg/mL to 0.88ng/ml). After 4 additional days, plates were removed from the incubator and equilibrated to room temperature. After approximately 30min, 50μΙ, was removed from each well, and then 50μΙ, of CellTiter-Glo ® 2.0 Luminescent Solution (Promega, G9423) was added to each well. After shaking the plates at 750rpm for 5min followed by 20min incubation without shaking, luminescence was measured on a Tecan Infinity F200 plate reader with an integration time of Is per well. Curves of luminescence versus ADC concentration (ng/mL) were fitted with Graphpad Prism Software. The IC 50 values were determined using the built-in "log(inhibitor) vs. response — Variable slope (four parameters)" IC 50 determination function of Prism Software.

Table 4. In vitro cell killing of 697 cells by anti-RORl ADCs or ADC mixtures (IC 5 o, n /mL , NA = Not Available

Figure 10A shows the dose-repose curves of the in vitro cell killing assays on 697 cells with the ADCs of Table 4. As per the Table and Figure, for the same total dose of ADC, the mixture of ADCs of the invention provides synergistic killing of human 697 B cell precursor leukemia cells that is superior to the individual ADCs and to the isotype control.

Table 5. In vitro cell killing of 697 cells by anti-RORl ADCs or ADC mixtures (IC 50 ,

Figure 10B shows the dose-respose curves of the in vitro cell killing assays on 697 cells with the ADCs of Table 5. For the same total dose of ADC, the mixture of ADCs of the invention provides synergistic killing of human 697 B cell precursor leukemia cells that is superior to the individual ADCs.

Table 6. In vitro cell killing of 697 cells by anti-RORl ADCs or ADC mixtures (IC 50 ,

Figure IOC shows the dose-response curves of the in vitro cell killing assays on 697 cells with the ADCs of Table 6. For the same total dose of ADC, the mixture of ADCs of the invention provides synergistic killing of human 697 B cell precursor leukemia cells that is superior to the individual ADCs.

Table 7. In vitro cell killing of 697 cells by anti-RORl ADCs or ADC mixtures (IC 50 ,

Figure 10D shows the dose-response curves of the in vitro cell killing assays on 697 cells with the ADCs of Table 7. For the same total dose of ADC, the mixture of ADCs of the invention provides synergistic killing of human 697 B cell precursor leukemia cells that is superior to the individual ADCs.

Table 8. In vitro cell killing of 697 cells by anti-RORl ADCs or ADC mixtures (IC 50 ,

Figure 10E shows the dose-response curves of the in vitro cell killing assays on 697 cells with the ADCs of Table 8. For the same total dose of ADC, the mixture of ADCs of the invention provides synergistic killing of human 697 B cell precursor leukemia cells that is superior to the individual ADCs.

Example 11. In vitro cytotoxicity of scFv-Fc format anti-RORl bi-epitope reactive ADCs (BETR-ADCs™) on human 697 B cell precursor leukemia cells

Cytotoxicity of anti-RORl scFv-Fc-based bi-epitope reactive ADCs (BETR-ADCs™) and ADC mixtures was investigated using human cell line 697. The same protocol as Example 10 was applied to the ADCs of Table 9.

Table 9. In vitro cell killing of 697 cells by scFv-Fc-based anti-RORl ADCs or ADC mixtures (IC50, ng/mL)

ADC 697 XBR1-402-G5-PNU (scFv-Fc format, adc251) 456

ERR1-324-G5-PNU (scFv-Fc format, adc252) 6Ί90

XBR1-402-G5-PNU (scFv-Fc format, adc251) / ERR1-324-G5-PNU (scFv format, 193

adc252) mixture

XBRl-402-pl-ERRl-324-p2-G5-PNU (scFv-Fc bi-epitopic reactive, adc249) 236

Figure 11 shows the dose-response curves of the in vitro cell killing assays on 697 cells with the scFv-Fc-based bi-epitope reactive anti-RORl ADCs (BETR-ADCs™) and individual anti-RORl ADCs of Table 9. As per the above Table and Figure 11, for the same total dose of ADC, the mixture of the scFv-Fc-based ADCs and the bi-epitope reactive scFv-Fc-based ADC of the invention provide cell killing of human 697 B cell precursor leukemia cells that is superior to the cell killing achieved with individual scFv-Fc-based ADCs.

Example 12. In vitro cytotoxicity of DVD-Ig-based anti-RORl bi-epitope reactive ADCs (BETR-ADCs™) on 697 cells

Cytotoxicity of DVD-Ig-based bi-epitope reactive anti-RORl ADCs (BETR-ADCs™) and individual anti-RORl ADCs was investigated using human cell line 697. The same protocol as Example 10 was applied to the ADCs of Table 10.

Table 10. In vitro cell killing of various human cancer cells by anti-RORl ADCs and

Figure 12 shows the dose-response curves of the in vitro cell killing assay on 697 cells with the ADCs of Table 10. As per the above Table and Figure 12, for the same total dose of ADC, the bi-epitope reactive anti-RORl DVD-Ig-based ADC of the invention shows cell killing of the RORl positive human cancer cells that is superior to the cell killing achieved with individual ADCs.

Example 13. In vitro cytotoxicity of single and mixtures of anti-RORl antibody-based ADCs on human colon cancer HT-29, breast cancer MDA-MB-468, lung cancer A549, and breast cancer HS 578T cells Cytotoxicity of 50:50 mixtures of anti-RORl ADCs was investigated using human cell lines: HT-29, MDA-MB-468, A549, HS 578T. For this, the following cells per well were plated on 96-well plates (excluding edge wells, which contained water) and were grown at 37°C in a humidified incubator at 7.5% C0 2 atmosphere in growth medium (DMEM supplemented with 10% by vol. FCS, lOOIU/ml Pen-Strep-Fungizone and 2mM L-Glutamine).

Table 11. Cell plating

After 1-day incubation, each ADC or ADC mixture was added to respective wells in an amount of 25 μΐ, of 3.5 -fold serial dilutions in growth medium (resulting in final ADC or ADC mixture concentrations from 20μg/mL to 0.88ng/ml). After 4 additional days, plates were removed from the incubator and equilibrated to room temperature. After approximately 30min, 50μΙ, was removed from each well, and then 50μ∑ of CellTiter-Glo ® 2.0 Luminescent Solution (Promega, G9423) was added to each well. After shaking the plates at 750rpm for 5min followed by 20min incubation without shaking, luminescence was measured on a Tecan Infinity F200 plate reader with an integration time of Is per well. Curves of luminescence versus ADC concentration (ng/mL) were fitted with Graphpad Prism Software. The IC 50 values, determined using the built-in "log(inhibitor) vs. response — Variable slope (four parameters)" ICso determination function of Prism Software, are reported in Table 12.

Table 12. In vitro cell killing of various human cancer cells by anti-RORl ADCs and ADC mixtures, as well as an isot e control IC50, n /mL

Figure 13 shows the dose-response curves of the in vitro cell killing assays on HT-29, MDA- MB-468, A549, HS 578T cells with the ADCs of Table 12, either as single ADCs (panel A) or ADC-mixture (panel B). As per the above Table and Figure 13, for the same total dose of ADC, the mixture of selected ADCs of the invention provides cell killing of human RORl positive 697 cancer cells that is superior to the cell killing of individual ADCs.

Example 14. In vitro cytotoxicity of scFv-IgG-based anti-RORl bi-epitope reactive ADCs (BETR-ADCs™) on hRORl-overexpressing EMT-6 cells

Cell line engineering for ectopic expression of hRORl in the EMT-6 murine breast cancer cell line: Murine EMT-6 breast cancer cells were cultured in DMEM complete (Dulbecco's Modified Eagle Medium (DMEM) High Glucose (4.5 g/1) with L-Glutamine with 10% (v/v) Fetal Calf Serum (FCS), 100 IU/mL of Pen-Strep-Fungizone and 2 mM L-glutamine (all Bioconcept, Allschwil, Switzerland)) at 37°C and 5% C0 2 . Cells were engineered to overexpress RORl by transposition as follows: cells were centrifuged (6 min, 1200 rpm, 4°C) and resuspended in RPMI-1640 media (5xl0 6 cells/mL). 400 μΐ, of this cell suspension was then added to 400 \iL of RPMI containing 13.3 μg of transposable vector pPB-PGK- Puro-RORl, directing co-expression of full-length RORl (NP_005003.2) and the puromycin- resistance gene, and 6.6 μg of transposase-containing vector pcDNA3.1_hy_mPB. The DNA/EMT-6 cell mixture was transferred to electroporation cuvettes (0.4 cm-gap, 165-2088, BioRad, Cressier, Switzerland) and electroporated using the Biorad Gene Pulser II with capacitance extender at 300V and 950 μΡ. Then, cells were incubated for 5-10 min at room- temperature. Following the incubation, cells were centrifuged at 1200 rpm for 6 min, washed once and subsequently resuspended in DMEM complete prior to incubation at 37°C in a humidified incubator at 5% C0 2 atmosphere. One day after electroporation, cell pools stably expressing human RORl were selected by adding 3 μg/mL pui mycin (Sigma-Aldrich, P8833).

RORl expression on selected EMT-6-ROR1 cells was confirmed by flow cytometry (not shown). To isolate RORl -expressing EMT-6 cell clones, following trypsinization, 10 6 cells were centrifuged in FACS tubes; obtained pellets were resuspended in buffer (PBS with 2% (v/v) FCS). Cells were then incubated with 2A2 (mAb066, Baskar et al., 2012); 30 min, 4°C, final concentration 2 μg/mL), followed by centrifugation and washing. Cells were then resuspended as previously and incubated with anti-human IgG antibody (Fc gamma-specific) PE (eBioscience, Vienna, Austria, 12-4998-82) with a 1 :250 dilution in the dark (30 min, 4°C), washed once in buffer and kept on ice until FACS sorting.

Using a FACS Aria II, cells were single cell sorted into a 96-well flat-bottom plate containing 200 μΐ, of DMEM complete per well. This plate was incubated at 37°C and clones were expanded to 6-well plates before analysis of RORl -expression by flow cytometry as outlined above, using a FACSCalibur instrument (BD Biosciences) and FlowJo analytical software (Tree Star, Ashland, OR) for analysis. Figure 17A shows the FACS analysis data of clone 14 detected with anti-RORl antibody 2A2 (mAb066).

Cytotoxicity. Cytotoxicity of an scFv-Ig-based bi-epitope reactive anti-RORl ADC (BETR- ADC™) and individual anti-RORl ADCs was investigated using the above engineered EMT- 6 cells (clone 14). The same protocol as in Example 10 was applied to the ADCs of Table 13, plating 1000 EMT-6 cells per well.

Table 13. In vitro cell killing of human cancer cells by anti-RORl ADCs and ADC mixtures, as well as an isoty e control IC 5 Q, DM)

Figure 17B shows the dose-response curves of the in vitro cell killing assay on hRORl- overexpressing EMT-6 cells with the ADCs of Table 13. As per the above Table and Figure 17B, for the same total dose of ADC, the bi-epitope reactive anti-RORl scFv-Ig-based ADC of the invention shows cell killing of the RORl positive human cancer cells that is superior to the cell killing achieved with individual ADCs.

REFERENCES

WO2016102679

WO2014140317

Baskar et al, Int J Med Sci. 2012;9(3):193-9

Skerra and Pluckthun, Science 2401038-41, 1988. Balakrishnan et al. (2016) Clin Cancer Res. doi: 10.1158/1078-0432

Rebagay et al. (2012) Frontiers Oncol. 2(34):l-8; doi 10.3389

Shatz et al., mAbs 5:6, 872-881 (2013).

Lefranc MP et al., 2015 Nucleic Acids Res. Jan;43: D413-22. doi: 10.1093/nar/gkul056

Ward et al., Nature 341 :544-546, 1989

Bird et al., Science 242: 423-426, 1988

Huston et al., Proc. Natl. Acad. Sci. USA 85:5879-5883, 1988

Reiter et al., Int. J. Cancer 67:113-23, 1996

Cai and Garen, Proc. Natl. Acad. Sci. USA 93:6280-85, 1996.

Dumoulin et al., Nat. Struct. Biol. 11 :500-515, 2002

Ghahroudi et al., FEBS Letters 414:521-526, 1997

Bond et al J. Mol. Biol. 332643-55, 2003.

Chen et al, PNAS 2011; 108(28); 11399-11404.

Dorr BM et al., PA¾S 2014; 111, 13343-8.

Beerli et al. (2015) Plos One 10, el 31177

Quintieri et al., Clin. Cancer Res 11:1608-1617 (2005)

Cai and Garen, Proc. Natl. Acad. Sci. USA 93:6280-85, 1996

Harlow & Lane, Using Antibodies, A Laboratory Manual, Cold Spring Harbor Laboratory

Press, Cold Spring Harbor, New York, 1998.

Shinkai et al. (1992) Cell 68:855-67

Popkov et al, J. Mol. Biol. 325:325-335, 2003.

Yang et al Plos One 6:e21018, 2011.

McCartney-Francis et al., Proc. Natl. Acad. Sci. U S A 81 :1794-1798,

Hofer et al., J Immunol Meth 318:75-87, 2007

Rader, Methods Mol Biol 525:101-128, xiv, 2009.

Yang and Rader, Methods Mol Biol 901 :209-232, 2012

Garen, Proc. Natl. Acad. Sci. USA 93:6280-85, 1996

Kwong and Rader, Curr Protoc Protein Sci Chapter 6:Unit 6 10, 2009

SEQUENCES The following sequences form part of the disclosure of the present application. A WIPO ST 25 compatible electronic sequence listing is provided with this application, too. For the avoidance of doubt, if discrepancies exist between the sequences in the following table and the electronic sequence listing, the sequences in this table shall be deemed to be the correct ones.

ER 1-306HC QEQLKESGGGLVQPGGSLKLSCKASGFDLSNYGVSWVRQAPGKGLEWIGYID amino acid PTFDYTYYASWVNGRFSISRENTQNTVSLQINSLTPADTATYFCARWVYGVD sequence DYGDGNWLDLWGQGTLVTISSASTKGPSVFPLAPSSKSTSGGTAALGCLVKD

YFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSWTVPSSSLGTQTYIC

NVNHKPSNTKVD KVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLM

ISRTPEVTCVWDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRWS

VLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRD

ELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYS

KLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK

ERR1-306LC QFVLTQSPSVSAALGASAKLTCTLSSAHKTYTIDWYQQQQGEAPRYLMELKS amino acid DGSYTKGTGVPDRFSGSSSGADRYLIIPSVQADDEADYYCGTDYSGGYVFGG sequence GTQLTVTGQPKAAPSVTLFPPSSEELQANKATLVCLISDFYPGAVTVAWiCAD

SSPVKAGVETTTPSKQSNNKYAASSYLSLTPEQWKSHKSYSCQVTHEGSTVE

KTVAPTECS

XBR1-402 HC QEQQKESGGGLFKPTDTLTLTCTASGFDISSYYMSWVRQAPGNGLEWIGAIG amino acid ISGNAYYASWAKSRSTITRNTNLNTVTLKMTSLTAADTATYFCARDHPTYGM sequence DLWGPGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTV

SWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSN

TKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVT

CVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQD

WLNG EYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVS

LTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSR

WQQGNVFSCSV HEALHNHYTQKSLSLSPGK

XBR1-402 LC SYELTQLPSVSVSLGQTARITCEGNNIGSKAVHWYQQKPGLAPGLLIYDDDE amino acid RPSGVPDRFSGSNSGDTATLTISGAQAGDEADYYCQVWDSSAYVFGGGTQLT sequence VTGQPKAAPSVTLFPPSSEELQANKATLVCLISDFYPGAVTVAWKADSSPVK

AGVETTTPSKQSNNKYAASSYLSLTPEQWKSHKSYSCQVTHEGSTVEKTVAP

TECS

ERR1-403HC QEQLKESGRGLVQPGGSLKLSCKASGFDFSGWY TWVRQAPGKGLE IGTIG amino acid TTKGRTYYASWVNGRFTISSDNAQNTVDLQMNSLTAADRA YFCVRGSDYFD sequence LWGPGTLVTISSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVS

WNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNT

KVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPP PKDTLMISRTPEVTC

VVVDVSHEDPEVKFNWYVDGVEVHNAKT PREEQYNSTYRVVSVLTVLHQDW

LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSL

TCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRW

QQGNVFSCSVMHEALHNHYTQKSLSLSPGK

ERR1-403LC SYELTQLPSVSVSLGQTARITCGGNSIGSKAVNWYQQKPGLAPGLLIYDDDE amino acid RPSGVPARFSGSNSGDTATLTISGAQAGDEADYYCQLWDSSAGAYVFGGGTQ sequence LTVTGQPKAAPSVTLFPPSSEELQANKATLVCLISDFYPGAVTVAWKADSSP

VKAGVETTTPSKQSNNKYAASSYLSLTPEQWKSHKSYSCQVTHEGSTVEKTV

APTECS

Top43HC amino QSLEESGGRLVTPGTPLTLTCTVSGFSLSSYWMSWVRQAPG GLEWIGAIYG acid sequence SGNTYYASWAKGRFTISKTSTTVDLKITSPTTEDTA YFCARDVHSTATDLW

GPGTLVTISSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWN SGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKV

DKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVV

VDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLN

GKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTC

LVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQ

G FSCSVMHEALHNHYTQKSLSLSPGK Top43LC amino SYELTQLPSVSVSLGQTARITCGGNNIGSKAVNWYQQKPGLAPGLLIYNDDE acid sequence RPSGVPDRFSGSNSGDTATLTISGAQAGDEADYYCQLWDSSAGAYVFGGGTQ

LTVTGQPKAAPSVTLFPPSSEELQANKATLVCLISDFYPGAVTVAWKADSSP VKAGVETTTPSKQSNNKYAASSYLSLTPEQWKSH SYSCQVTHEGSTVEKTV

APTECS

XBR1-402 scFv SYELTQLPSVSVSLGQTARITCEGNNIGSKAVHWYQQKPGLAPGLLIYDDDE HC & LC amino RPSGVPDRFSGSNSGDTATLTISGAQAGDEADYYCQVWDSSAYVFGGGTQLT acid sequence VTGGGGSGGGGSGGGGSQEQQKESGGGLFKPTDTLTLTCTASGFDISSYYMS

WVRQAPGNGLEWIGAIGISGNAYYASWAKSRSTITRNTNLNTVTLKMTSLTA ADTATYFCARDHPTYGMDLWGPGTLVTVSSEPKSSD THTCPPCPAPELLGG PSVFLFPPKPKDTL ISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKT

KPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKG

QPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNY T

TPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSP

GK

ERRl-324-scFv ELVLTQTPSPVSAAVGGTVTINCQASQSVYGNNELAWYQQKPGQPPKLLIYR HC & LC amino ASILTSGVPSRFKGSGSGTQFTLTISNVQREDAATYYCLGGYVSQSYRAAFG acid sequence GGTELEILGGGGSGGGGSGGGGSQSLEESGGGLVQPGESLTLTCTVSGFSLS

RNGMTWVRQAPGKGLEWIGIITSSGDKYYATWAKGRFTISKTSSTTVDLKMT SLTTEDTATYFCARGTVSSDIWGPGTLVTISSEPKSSDKTHTCPPCPAPELL GGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNA

KTKPREEQYNS YRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKA

KGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNY

KTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSL

SPGK

scFv-402-Fc-KIH- SYELTQLPSVSVSLGQTARITCEGNNIGSKAVHWYQQKPGLAPGLLIYDDDE pl (Knob) RPSGVPDRFSGSNSGDTATLTISGAQAGDEADYYCQVWDSSAYVFGGGTQLT

VTGGGGSGGGGSGGGGSQEQQKESGGGLFKPTDTLTLTCTASGFDISSYY S WVRQAPGNGLEWIGAIGISGNAYYASWAKSRSTITRNTNLNTVTLKMTSLTA ADTATYFCARDHPTYGMDLWGPGTLVTVSSEPKSSDKTHTCPPCPAPELLGG PSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKT

KPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKG

QPREPQVYTLPPSREEMTKNQVSLSCAVKGFYPSDIAVEWESNGQPENNYKT

TPPVLDSDGSFFLVSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSP

GK

scFv-ERRl-324- ELVLTQTPSPVSAAVGGTVTINCQASQSVYGNNELAWYQQKPGQPPKLLIYR Fc-KIH-p2 (Hole) ASILTSGVPSRFKGSGSGTQFTLTISNVQREDAATYYCLGGYVSQSYRAAFG

GGTELEILGGGGSGGGGSGGGGSQSLEESGGGLVQPGESLTLTCTVSGFSLS RNGMTWVRQAPGKGLEWIG11TSSGDKYYATWAKGRFTISKTSSTTVDLK T SLTTEDTATYFCARGTVSSDIWGPGTLVTISSEPKSSDKTHTCPPCPAPELL GGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNA

KTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKA

KGQPREPQVYTLPPSREEMTKNQVSLWCLVKGFYPSDIAVEWESNGQPENNY

KTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSL

SPGK 18 324-LL-R12 HC QSLEESGGGLVQPGESLTLTCTVSGFSLSRNGMTWVRQAPGKGLEWIGIITS amino acid SGDKYYATWAKGRFTISKTSSTTVDLKMTSLTTEDTATYFCARGTVSSDIWG sequence PGTLVTISSASTKGPSVFPLAPQEQLVESGGRLVTPGGSLTLSCKASGFDFS

AYYMSWVRQAPGKGLEWI TIYPSSGKTYYAT VNGRFTISSDNAQNTVDLQ MNSLTAADRATYFCARDSYADDGALFNIWGPGTLVTISSASTKGPSVFPLAP SSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSL SSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPEL LGGPSVFLFPPKPKDTL ISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHN AKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISK AKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENN Y TTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLS LSPGK

19 324-LL-R12 LC ELVLTQTPSPVSAAVGGTVTINCQASQSVYGNNELAWYQQKPGQPPKLLIYR amino acid ASILTSGVPSRFKGSGSGTQFTLTISNVQREDAATYYCLGGYVSQSYRAAFG sequence GGTELEILRTVAAPSVFIFPPELVLTQSPSVSAALGSPAKITCTLSSAHKTD

TIDWYQQLQGEAPRYLMQVQSDGSYTKRPGVPDRFSGSSSGADRYLIIPSVQ ADDEADYYCGADYIGGYVFGGGTQLTVTGQPKAAPSVTLFPPSSEELQANKA TLVCLISDFYPGAVTVAWKADSSPV AGVETTTPSKQSNNKYAASSYLSLTP EQWKSHKSYSCQVTHEGSTVEKTVAPTECS

20 R12 HC amino QEQLVESGGRLVTPGGSLTLSCKASGFDFSAYYMS VRQAPGKGLEWIATIY acid sequence PSSGKTYYATWVNGRFTISSDNAQNTVDLQMNSLTAADRATYFCARDSYADD

GALFNIWGPGTLVTISSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPE PVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNH KPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRT PEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTV LHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTK NQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTV DKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK

1 R12 LC amino ELVLTQSPSVSAALGSPAKITCTLSSAHKTDTIDWYQQLQGEAPRYLMQVQS acid sequence DGSYTKRPGVPDRFSGSSSGADRYLIIPSVQADDEADYYCGADYIGGYVFGG

GTQLTVTGQPKAAPSVTLFPPSSEELQANKATLVCLISDFYPGAVTVAWKAD SSPVKAGVETTTPSKQSNNKYAASSYLSLTPEQWKSHKSYSCQVTHEGSTVE KTVAPTECS

2 2A2 HC amino QVQLQQSGAELVRPGASVTLSCKASGY FSDYEMHWVIQTPVHGLEWIGAID acid sequence PETGGTAYNQKFKGKAILTADKSSSTAYMELRSLTSEDSAVYYCTGYYDYDS

FTYWGQGTLVTVSAASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVT VSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPS NTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEV TCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQ DWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQV SLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKS RWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK

3 2A2 LC amino DIVMTQSQKIMSTTVGDRVSITCKASQNVDAAVAWYQQKPGQSPKLLIYSAS acid sequence NRYTGVPDRFTGSGSGTDFTLTISNMQSEDLADYFCQQYDIYPYTFGGGTKL

EIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQS GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKS FNRGEC AclO HC amino QIQLQQSGPEVVKPGASVKISCKASGYTFTDYYITWVKQKPGQGLEWIGWIY acid sequence PGSGNTKYNEKFKGKATLTVDTSSSTAF QLSSLTSEDTAVYFCANYGNYWF

AYWGQGTQVTVSAASTKGPSVFPLAPSS STSGGTAALGCLVKDYFPEPVTV SWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSN

TKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTL ISRTPEVT

CVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQD

WLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVS

LTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSR

WQQGNVFSCSVMHEALHNHYTQKSLSLSPGK

AclO LC amino DIVLTQSPASLAVSLGQRATISCKASQSVDFDGDSY N YQQKPGQPPKVLI acid sequence YAASNLESGIPARFSGSGSGTDFTLNIHPVEEEDAATYYCQQSNEDP TFGG

GTKLEI RTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDN ALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSP

VTKSFNRGEC pCEP4-h 0 l-F ATCCTGTTTCTCGTAGCTGCTGCAACTGGAGCACACTCCGCCCGGGGCGCCG

CCGCCCAG

pCEP4-hR0Rl- CCACTCGATCTTCTGGGCCTCGAAGATGTCGTTCAGGCCCTCCATCTTGTTC Avi-tag-R TTCTCCTT

pCEP4-signal-F- GCTGGGTACCGGCGCGCCACCATGGACTGGACTTGGAGAATCCTGTTTCTCG Kpnl TAGCTGCT

PCEP4-6HIS-R- GCCGGCCTCGAGTCAGTGATGGTGATGGTGGTGCTCGTGCCACTCGATCTTC Xhol TGGGCCTC

hRORl-His_R CGGCCTCGAGTCAGTGATGGTGATGGTGGTGCTCCATCTTGTTCTTCTCCTT

SP-hROR2_F GCTGGGTACCGGCGCGCCACCATGGACTGGACTTGGAGAATCCTGTTTCTCG

TAGCTGCTGCAACTGGAGCACACTCCGAAGTGGAGGTTCTGGATCCG

hROR2-His_R CGGCCTCGAGTCAGTGATGGTGATGGTGGTGCCCCATCTTGCTGCTGTCTCG

XBR1-402_VH_F GAGGAGGAGCTCACTCTCAGGAGCAGCAGAAGGAGTCCGGG

XBR1-402_VH_R CGATGGGCCCTTGGTGGAGGCTGAAGAGACGGTGACGAGGGTCCCTGGCCCC

CAGAGGTC

XBR1-402_ _F GAGAAGCTTGTTGCTCTGGATCTCTGGTGCCTACGGGTCCTATGAGCTGACA

CAGCTGCC

LEAD-B GGCCATGGCTGGTTGGGCAGC

Kpnl/Ascl-Signal GGTACCGGCGCGCCACCATGGACTGGACTTGGAGAATCCTGTTTCTCGTAGC

TGCTGCAA

CHl-internal/overlap-R GCCGCTGGTCAGGGCTCCTG

CHl-internal/overlap-F CAGGAGCCCTGACCAGCGGC

HC-CH3-R-Xhol GGCCTCGAGTCATT ACCCGGAG CAGGGA

ERR1-324 HC-F TTTCTCGTAGCTGCTGCAACTGGAGCACACTCC

CAGTCGCTGGAGGAGTCCGGG

ERR1-TOP43 HC- TTTCTCGTAGCTGCTGCAACTGGAGCACACTCC

F CAGTCGTTGGAGGAGTCCGGG

ERR1-TOP54 HC- TTTCTCGTAGCTGCTGCAACTGGAGCACACTCC

F CAGTCGTTGGAGGAGTCCGGG

VH-CHl-R-Ehel GGAGGGCGCCAGGGGGAAGACCGATGGGCCCTTGGT ERR1-TOP43 LC- TTTCTCGTAGCTGCTGCAACTGGAGCACACTCC

F TCCTATGAGCTGACACAGCTG

ERR1-TOP54 LC- TTTCTCGTAGCTGCTGCAACTGGAGCACACTCC

F TCCTATGAGCTGACACAGCTG

ERR1-324 KC-F TTTCTCGTAGCTGCTGCAACTGGAGCACACTCC

GAGCTCGTGCTGACCCAGACT

LC-R-Xhol GGCCTCGAGTTATGAACATTCTGTAGGGGC

KC-R-Xhol GGCCTCGAGTTAACACTCTCCCCTGTTGAA

SP-hRORl_F GCTGGGTACCGGCGCGCCACCATGGACTGGACTTGGAGAATCCTGTTTCTCG

TAGCTGCTGCAACTGGAGCACACTCCGCCCGGGGCGCCGCCGCCCAG

Trastuzumab HC EVQLVESGGGLVQPGGSLRLSCAASGFNIKDTYIHWVRQAPGKGLEWVARIY amino acid PTNGYTRYADSVKGRFTISADTSKNTAYLQ NSLRAEDTAVYYCSRWGGDGF sequence YAMDYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEP

VTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHK

PSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTP

EVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVL

HQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKN

QVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVD

KSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK

Trastuzumab LC DIQMTQSPSSLSASVGDRVTITCRASQDVNTAVAWYQQKPGKAPKLLIYSAS amino acid FLYSGVPSRFSGSRSGTDFTLTISSLQPEDFATYYCQQHYTTPPTFGQGTKV sequence EIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQS

GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKS

FNRGEC

Staphylococcus aureus sortase A recognition sequence, with X -LPXTG

being any amino acid

Staphylococcus aureus sortase A recognition sequence, with X -LPXAG

being any amino acid

recognition sequence for Staphylococcus aureus sortase A or -LPXSG

engineered sortase A 4S-9 from Staphylococcus aureus, with X

being any amino acid

recognition sequence for engineered sortase A 2A-9 from -LAXTG

Staphylococcus aureus, with X being any amino acid

Streptococcus pyogenes sortase A recognition sequence, with X -LPXTA

being any amino acid

Staphylococcus aureus sortase recognition sequence -NPQTN

Linker derived from Staphylococcus aureus sortase A recognition -LPXT (Gn) - sequence, with X being any amino acid and n > 1 and < 21

Linker derived from Staphylococcus aureus sortase A recognition -LPXA(Gn) - sequence, with X being any amino acid and n > 1 and < 21

Linker derived from recognition sequence for Staphylococcus -LPXS (Gn) - aureus sortase A or engineered sortase A 4S-9 from

Staphylococcus aureus, with X being any amino acid and n > 1 and

< 21

Linker derived from recognition sequence for engineered sortase -LAXT (Gn) - A 2A-9 from Staphylococcus aureus, with X being any amino acid

and n > l and < 21 Linker derived from Streptococcus pyogenes sortase A -LPXT (Gn) - or recognition sequence, with X being any amino acid and n > 1 and -LPXT (An) - < 21

Linker derived from Staphylococcus aureus sortase recognition -NPQT (Gn) - sequence, with n > 1 and < 21

signal sequence MNFGLRLIFLVLTLKGVQC strep ll-tag WSHPQFEK

scFv-lgG_324_4-2 HC sequence QSLEESGGGLVQPGESLTLTCT

VSGFSLSR GMTWVRQAPGKGL EWIGIITSSGDKYYATWAKGRF TISKTSSTTVDLKMTSLTTEDT ATYFCARGTVSSDIWGPGTLVT ISSGGGGSGGGGSGGGGSGGGG SELVLTQTPSPVSAAVGGTVTI NCQASQSVYGNNELAWYQQKPG QPPKLLIYRASILTSGVPSRFK GSGSGTQFTLTISNVQREDAAT YYCLGGYVSQSYRAAFGGGTEL EILGGGSGGGSGGGSGGGSGGG SQEQQKESGGGLFKPTDTLTLT CTASGFDISSYYMSWVRQAPGN GLEWIGAIGISGNAYYASWAKS RSTITRNTNLNTVTLKMTSLTA ADTATYFCARDHPTYGMDLWGP GTLVTVSSASTKGPSVFPLAPS SKSTSGGTAALGCLVKDYFPEP VTVSWNSGALTSGVHTFPAVLQ SSGLYSLSSVVTVPSSSLGTQT YICNVNHKPSNTKVDKKVEPKS CDKTHTCPPCPAPELLGGPSVF LFPPKPKDTLMISRTPEVTCVV VDVSHEDPEVKFNWYVDGVEVH NAKTKPREEQYNSTYRVVSVLT VLHQDWLNGKEYKCKVSNKALP APIEKTISKAKGQPREPQVYTL PPSRDELTKNQVSLTCLVKGFY PSDIAVEWESNGQPENNYKTTP PVLDSDGSFFLYSKLTVDKSRW QQGNVFSCSVMHEALHNHYTQK SLSLSPGR

scFv-lgG_324_4-2 LC sequence SYELTQLPSVSVSLGQTARITC

EGNNIGSKAVHWYQQKPGLAPG LLIYDDDERPSGVPDRFSGSNS GDTATLTISGAQAGDEADYYCQ VWDSSAYVFGGGTQLTVTGQPK AAPSVTLFPPSSEELQANKATL VCLISDFYPGAVTVAWKADSSP VKAGVETTTPSKQSNNKYAASS YLSLTPEQWKSHKSYSCQVTHE GSTVEKTVAPTECS

E 1-324 VL CDR1 QASQSVYGNNELA

ERR1-324 VL CDR2 RASILTS 71 ERR1-324 VL CDR3 LGGYVSQSYRAA

72 ERR1-324 VH CDR1 RNGMT

73 ERR1-324 VH CDR2 11TSSGDKYYATWAKG

74 ERR1-324 VH CDR3 GTVSSDI

75 2A2 VH CDR1 GYTFSDYEMH

76 2A2 VH CDR2 AIDPETGGTAYNQKFKG

77 2A2 VH CDR3 YYDYDSFTY

78 2A2 VL CDR1 KASQ VDAAVA

79 2A2 VL CDR2 SAS RYT

80 2A2 VL CDR3 QQYDIYPYT

81 XBR1-402 VH CDR1 SYYMS

82 XBR1-402 VH CDR2 AIGISGNAYYASWAKS

83 XBR1-402 VH CDR3 DHPTYGMDL

84 XBR1-402 VL CDR1 EGNNIGSKAVH

85 XBR1-402 VL CDR2 DDDERPS

86 XBR1-402 VL CDR3 QVWDSSAYV

87 R12 VH CDR1 AYYMS

88 R12 VH CDR2 IYPSSGKTYYATWVNG

89 R12 VH CDR3 DSYADDGALFNI

90 R12 VL CDR1 TLSSAHKTDTID

91 R12 VL CDR2 GSYTKRP

92 R12 VL CDR3 GADYIGGYV

93 TOP43 VH CDR1 SYWMS

94 TOP43 VH CDR2 Al YGSG NTYYASWAKG

95 TOP43 VH CDR3 DVHSTATDL

96 TOP43 VL CDR1 GGNNIGSKAVN

97 TOP43 VL CDR2 NDDERPS

98 TOP43 VL CDR3 QLWDSSAGAYV