Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
TGF-Β-RECEPTOR ECTODOMAIN FUSION MOLECULES AND USES THEREOF
Document Type and Number:
WIPO Patent Application WO/2018/158727
Kind Code:
A1
Abstract:
The present invention relates, in general, to polypeptides capable of binding and neutralizing transforming growth factor beta (TGF-β) ligands, and uses of these polypeptides for treating disorders related to TGF-beta expression or activation (e.g. cancer and fibrotic diseases), and methods of making such molecules.

Inventors:
LENFERINK ANNE E G (CA)
ZWAAGSTRA JOHN C (CA)
SULEA TRAIAN (CA)
O'CONNOR-MCCOURT MAUREEN D (CA)
Application Number:
PCT/IB2018/051320
Publication Date:
September 07, 2018
Filing Date:
March 01, 2018
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
NAT RES COUNCIL CANADA (CA)
International Classes:
C07K19/00; A61K38/16; A61P35/00; C07K14/71; C07K16/00; C12N5/10; C12N15/62; C12P21/02
Domestic Patent References:
WO1998048024A11998-10-29
WO2012142515A22012-10-18
Foreign References:
US8658135B22014-02-25
Other References:
ISAKA ET AL.: "Gene therapy by transforming growth factor-b receptor-IgG Fc chimera suppressed extracellular matrix accumulation in experimental glomerulonephritis", KIDNEY INTERNATIONAL, vol. 55, no. 2, 1999, pages 465 - 475, XP028806074, ISSN: 0085-2538
DE CRESCENZO ET AL.: "Real-time Monitoring of the Interactions of Transforming Growth Factor- (TGF-) Isoforms with Latency-associated Protein and the Ectodomains of the TGF- Type II and III Receptors Reveals Different Kinetic Models and Stoichiometries of Binding", THE JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 276, no. 32, 29 May 2001 (2001-05-29), pages 29632 - 29643, XP002571832, ISSN: 1083-351X
KOMESLI ET AL.: "Chimeric extracellular domain of type II transforming growth factor (TGF)-beta receptor fused to the Fc region of human immunoglobulin as a TGF-f3 antagonist", EUROPEAN JOURNAL OF BIOCHEMISTRY, vol. 254, 15 June 1998 (1998-06-15), pages 505 - 513, XP000856458, ISSN: 0014-2956
ZWAAGSTRA ET AL.: "Engineering and Therapeutic Application of Single-Chain Bivalent TGF-b Family Traps", MOLECULAR CANCER THERAPEUTICS, vol. 11, no. 7, July 2012 (2012-07-01), pages 1477 - 1487, XP055072658, ISSN: 1535-7163
See also references of EP 3589663A4
Attorney, Agent or Firm:
THOMAS, Roula et al. (CA)
Download PDF:
Claims:
A polypeptide construct useful to inhibit an effect of a Transforming Growth Factor Beta (TGF-β) isotype, the construct comprising: a first region comprising a first TGF-β receptor ectodomain (TpR-ECD), and a second region comprising the second constant domain (CH2) and/or third constant domain (CH3) of an antibody heavy chain; wherein the N-terminus of the second region is joined to the C-terminus of the first region; and wherein the construct further comprises a second TpR-ECD linked in tandem to the first TpR-ECD.

A polypeptide construct according to claim 1 , wherein said polypeptide construct inhibits TGF-β activity with at least 20-fold, 100-fold, 200-fold, 300-fold, 400-fold, 500- fold, or 600-fold greater potency than a counterpart construct having a single TpR-ECD.

A polypeptide construct according to claim 1 , wherein said polypeptide construct inhibits TGFp activity with at least 100-fold, 200-fold, 300-fold, 400-fold, 500-fold, 600- fold, 700-fold, 800-fold or 900-fold greater potency than a non-Fc fused TpR-ECD doublet.

A polypeptide construct according to claim 1 , wherein said second region comprises an Fc region of an antibody heavy chain.

A polypeptide construct according to claim 1 , wherein said TpR-ECD comprises a TGF- p Type II receptor ectodomain (TpRII-ECD).

A polypeptide construct according to claim 1 , wherein the first TpR-ECD and the second TpR-ECD bind the same TGF-p isotype.

A polypeptide construct according to claim 6, wherein the first TpR-ECD and the second TpR-ECD each bind both TGF-pi and TGF-p3.

A polypeptide construct according to claim 1 wherein the first TpR-ECD and the second TpR-ECD comprise the same amino acid sequence.

9. A polypeptide construct according to claim 1 wherein the TpR-ECD comprises the amino acid sequence of SEQ ID NO:1 .

10. A polypeptide construct according to claims 1 to 9, wherein the second region comprises the CH2 and CH3 domains of a heavy chain of an IgG antibody. 1 1 . A polypeptide construct according to claim 10, wherein the IgG antibody is an lgG1 or lgG2 antibody.

12. A polypeptide construct according to any one of claims 1 -1 1 , wherein the second region comprises a hinge region comprising cysteine residues for crosslinking of the construct with another construct. 13. A polypeptide construct according to any one of claims 1 -12 wherein the second region is selected from a group consisting of SEQ ID NO:12, SEQ ID NO:15, SEQ ID NO:18, SEQ ID NO:24, and a sequence substantially identical thereto.

14. A polypeptide construct according to claim 1 , selected from a group consisting of SEQ

ID NO:10, SEQ ID NO:14, SEQ ID NO:17, SEQ ID NO:20, SEQ ID NO:23, and a sequence substantially identical thereto.

A polypeptide construct according to any one of claims 1 -14, wherein the construct is a dimeric polypeptide comprising a first and a second polypeptide constructs linked between respective antibody constant domains by at least one disulfide bridge.

A polypeptide construct according to claim 13, wherein the dimeric polypeptide comprises: a first single chain polypeptide comprising a first and a second region; wherein a second region comprises the second constant domain (CH2) and third constant domain (CH3) of an antibody heavy chain, and a heavy chain variable region of a given antibody; wherein a first region comprises two TpRII-ECD; wherein the N-terminus of the second region is linked to the C-terminus of the first region; and

17. A nucleic acid molecule encoding any polypeptide construct of any one of claims 1 -16.

18. A vector comprising the nucleic acid molecule of claim 17.

19. A nucleic acid sequence that encodes any polypeptide of any preceding claim in a form that is secretable by a selected expression host. 20. A nucleic acid sequence according to claim 19, wherein the nucleic acid encodes a protein having the sequence SEQ ID NO:10, SEQ ID NO:14, SEQ ID NO:17, SEQ ID NO:20, SEQ ID NO:23 and a sequence substantially identical thereto.

21 . A nucleic acid sequence according to claim 20, wherein the nucleic acid is selected from the group consisting of SEQ ID NO:29, SEQ ID NO:30, SEQ ID NO:31 , SEQ ID NO:32, and SEQ ID NO:33.

22. A composition comprising a polypeptide construct of any one of claims 1 to 16 and a pharmaceutically-acceptable carrier, diluent, or excipient.

23. A transgenic cellular host comprising the nucleic acid molecule of claim 17 or a vector of claim 18. 24. The transgenic cellular host of claim 23, further comprising a second nucleic acid molecule or a second vector encoding a second polypeptide construct the same as the first polypeptide construct.

25. A method for producing a dimeric polypeptide comprising culturing the host and recovering from medium conditioned by the growth of that host a dimeric polypeptide construct according to claim 13.

26. The use of a polypeptide construct according to any one of claims 1 -16, for treatment of a medical condition, disease or disorder.

27. The use according to claim 26, wherein the medical condition, disease or disorder comprises cancer, ocular diseases, fibrotic diseases, or genetic disorders of connective tissue.

28. A polypeptide construct comprising SEQ ID NO:14.

29. The polypeptide construct according to claim 28, as a homodimer.

30. A pharmaceutical composition comprising the polypeptide of claim 28.

31 . Use of the polypeptide according to claim 28 for the treatment of cancer.

Description:
TGF-B-RECEPTOR ECTODOMAIN FUSION MOLECULES AND USES THEREOF

FIELD OF THE INVENTION

The present invention relates to TGF-β receptor ectodomain fusion molecules and uses thereof. More specifically, the present invention relates to TGF-β superfamily receptor ectodomain fusion molecules and their use in TGF-β ligand neutralization.

BACKGROUND OF THE INVENTION

TGF-β is part of a superfamily of over 30 ligands that regulate several physiological processes, including cell proliferation, migration and differentiation. Perturbation of their levels and/or signaling gives rise to significant pathological effects. For instance, TGF-β and activin ligands play critical pathogenic roles in many diseases including cancer (Hawinkels & Ten Dijke, 201 1 ; Massague et al, 2000; Rodgarkia-Dara et al, 2006). TGF-β, in particular, is considered as a critical regulator of tumor progression and is overexpressed by most tumor types. It favors tumorigenesis in part by inducing an epithelial-mesenchymal transition (EMT) in the epithelial tumor cells, leading to aggressive metastasis (Thiery et al, 2009). TGF-β also promotes tumorigenesis by acting as a powerful suppressor of the immune response in the tumor microenvironment (Li et al, 2006). In fact, TGF-β is recognized as one of the most potent immunosuppressive factors present in the tumor microenvironment. TGF-β interferes with the differentiation, proliferation and survival of many immune cell types, including dendritic cells, macrophages, NK cells, neutrophils, B-cells and T-cells; thus, it modulates both innate and adaptive immunity (Santarpia et al, 2015; Yang et al, 2010). The importance of TGF-β in the tumor microenvironment is highlighted by evidence showing that, in several tumor types (including melanoma, lung, pancreatic, colorectal, hepatic and breast), elevated levels of TGF- β ligand are correlated with disease progression and recurrence, metastasis, and mortality. Hence, significant effort has been invested in devising anti-tumor therapeutic approaches that involve TGF-β inhibition (Arteaga, 2006; Mourskaia et al, 2007; Wojtowicz-Praga, 2003). These approaches include the use of polypeptide fusions based on the TGF-β receptor ectodomain that binds or "traps" the TGF-β ligand (see WO01/83525; WO2005/028517; WO2008/1 13185; WO2008/157367; WO2010/0031 18; WO2010/099219; WO2012/071649; WO2012/142515; WO2013/000234; US5693607; US2005/0203022; US2007/0244042; US8318135; US8658135; US8815247; US2015/0225483; and US2015/0056199).

One approach to developing therapeutic agents that inhibit TGF-β function has been to use antibodies or soluble decoy receptors (also termed receptor ectodomain (ECD)-based ligand traps) to bind and sequester ligand, thereby blocking access of ligand to its cell surface receptors (Zwaagstra et al, 2012). In general, receptor ECD-based traps are a class of therapeutic agents that are able to sequester a wide range of ligands and that can be optimized using protein-engineering approaches (Economides et al, 2003; Holash et al, 2002; Jin et al, 2009).

Previously, a novel protein engineering design strategy was used to generate single-chain, bivalent TGF-β Type II receptor ectodomain (TpRII-ECD) traps that are able to potently neutralize members of the TGF-β superfamily of ligands due to avidity effects (Zwaagstra et al, 2012) [WO 2008/1 13185; WO 2010/031 168]. In this case, bivalency was achieved via covalent linkage of two TpRII ectodomains using regions of the intrinsically disordered regions (IDR) that flank the structured, ligand-binding domain of TpRII-ECD. An example of these single- chain bivalent traps, T22d35, exhibited TGF-β neutralization potencies ~100-fold higher than the monovalent non-engineered TpRII ectodomain, though the bivalent trap did not neutralize the TGF-p2 isotype and had a relatively short circulating half-life. It would be useful to provide TpRII-ECD-based traps having improved properties, such as enhanced potency.

SUMMARY OF THE INVENTION

The present invention provides a polypeptide construct with enhanced potencies in inhibiting TGFp. A polypeptide construct of the present invention comprises a first region and a second region, wherein the first region comprises a first and/or second TGFp receptor ectodomain (ECD); and wherein the second region comprises the second constant domain (C H 2) and/or third constant domain (C H 3) of an antibody heavy chain. In a preferred non-limiting embodiment, the C- terminus of the first region is linked to the N-terminus of the second region. In a preferred non- limiting embodiment, the first region of the polypeptide construct comprises a first TpRII-ECD (ECD1) and/or a second TpRII-ECD (ECD2), wherein ECD1 and ECD2 are linked in tandem.

The polypeptide construct provided, wherein the first region comprises a (TpRII-ECD)-(TpRII- ECD) doublet linked at its C-terminus with an antibody constant domain inhibits TGFp activity with at least 600-fold more potency than a counterpart construct having a single TpRII-ECD linked at its C-terminus with an antibody constant domain (i.e when a second ECD is absent, also referred to herein as a singlet). The polypeptide construct provided comprises a second, TGFp receptor ectodomain ECD that is linked in tandem to the first ECD, wherein the polypeptide construct (i.e. an ECD doublet construct) linked to an antibody constant domain exhibits TGFp neutralization (inhibits) that is at least 100, 200, 300, 400, 500, 600, 700, 800 or 900-fold greater than a counterpart construct in which the antibody constant domain is absent, (i.e. an ECD doublet construct, also referred to herein as a non-Fc fused doublet).

In connection with the TpRII-ECDs and the potency with which they inhibit TGF-β activity, it has been found that surprisingly enhanced potencies can result from careful selection of their constituents. This occurs when certain TpRII-ECDs are linked in tandem, and the C-terminus thereof is linked to the N-terminus of an antibody constant domain (Fc). When in their fused and dimeric form, comprising two such polypeptides cross-linked via cysteine bridging between the constant domain/s of each polypeptide, the resulting so-called "Fc fusions" having two TpRII-ECDs (an ECD "doublet") on each of the two "arms" can exhibit an inhibiting activity that is over 600-fold greater for TGF-pi , and over 20-fold greater for TGF-p3, as compared to "Fc fusions" having one ectodomain on each of the two "arms", as demonstrated by the inhibition of TGF-pi and -p3-induced IL-1 1 secretion by human non-small cell lung cancer (NSCLC) A549 cells, among others. The potency enhancement is evident, relative to counterparts that either lack the Fc region or that lack a second ECD (i.e. are an ECD "singlet"). The potency enhancement is at least 100, 200, 300, 400, 500, or 600-fold greater for the Fc-fused doublet over the Fc-fused singlet. The potency enhancement is at least 100, 200, 300, 400, 500, 600, 700, 800, 900-fold and approximately 1000-fold greater for the Fc-fused doublet over the non- Fc doublet. The potency enhancement is evident, relative to counterparts that either lack the Fc region (the Fc-fused doublet T22d35-Fc is 972- and 243-fold better for TGF-pi and TGF- p3, respectively than the non-Fc doublet), or that lack a second ECD (an ECD "singlet"; the Fc- fused doublet T22d35-Fc is 615- and 24-fold better for TGF-pi and TGF-p3, respectively than the non-Fc doublet). More specifically, the Fc-doublet (T22d35-Fc) exhibits a potency enhancement that is at least 970-fold greater for TGF-pi and at least 240-fold greater for TGF- p3 when compared to a non-Fc fused ECD doublet. Moreover, the Fc-doublet (T22d35-Fc) exhibits a potency enhancement that is at least 600-fold greater for TGF-pi and over 20-fold greater for TGF-p3 when compared to an Fc-singlet (T2m-Fc).

In a general aspect, there is provided a polypeptide construct comprising at least two TpRII- ECDs linked in tandem (i.e. ECD doublet), and an antibody constant domain comprising at least, the second constant domain (C H 2) and/or third constant domain (C H 3) of an antibody heavy chain, wherein the C-terminus of the ectodomains is linked to the N-terminus of the antibody constant domain. In this form, the construct is a single chain polypeptide. Thus, the antibody constant domain may comprise only the C H 2 domain, or it may comprise the C H 2 domain and the C H 3 domain.

In embodiments the polypeptides are provided as dimeric fusion polypeptides comprising two single chain polypeptides and cross-linking means coupling the chains covalently.

In other embodiments, the two ectodomains are the same, in terms of their binding targets generally and/or their target species.

In a preferred embodiment, the first region comprises two TGF-β receptor ectodomains (TGFpR-ECD or TpR-ECD'). In a preferred embodiment, the TpR-ECD is a TGF-β receptor type II ectodomain (TpRM-ECD). In a preferred embodiment, the TpR-ECD comprises SEQ ID NO:1 , and a sequence substantially identical thereto.

The second region may comprise a sequence selected from the group consisting of SEQ ID NO:12, SEQ ID NO:15, SEQ ID NO:18, SEQ ID NO:24, and a sequence substantially identical thereto.

In a preferred embodiment, the second region of a polypeptide construct of the present invention can further comprise a C H 1 . In embodiments, the constructs are monofunctional.

The polypeptide constructs of the present invention exploit an antibody heavy chain of human origin. In a preferred embodiment, the antibody heavy chain is selected from the group consisting of a human lgG1 (SEQ ID NO:15) and lgG2 (SEQ ID NO:24).

Thus, in another aspect, there is provided a polypeptide construct according to the present invention wherein the construct is a dimeric polypeptide; wherein the dimeric polypeptide comprises:

(i) a first single chain polypeptide comprising a second region comprising the second constant domain (C H 2) and third constant domain (C H 3) of an antibody heavy chain, and a first region comprising two TGF-p receptor ectodomains (TpRM-ECD), wherein the C-terminus of the first region is linked to the N-terminus of the second region, and

(ii) a second single chain polypeptide comprising a second region comprising the second constant domain (C H 2) and third constant domain (C H 3) of an antibody heavy chain, and a first region comprising two TGF-p receptor ectodomains (TpRM-ECD) linked in tandem, wherein the C-terminus of the first region is linked to the N-terminus of the second region, and the first single chain polypeptide is cross-linked with the second single chain polypeptide.

There is also provided a nucleic acid molecule encoding any polypeptide construct of the present invention. There is also provided a vector comprising the nucleic acid molecule of the present invention.

There is also provided a composition comprising one or more than one independently selected polypeptide constructs of the present invention and a pharmaceutically-acceptable carrier, diluent, or excipient.

There is also provided a transgenic cellular host comprising a nucleic acid molecule or a vector of the present invention. The transgenic cellular host can further comprise a second nucleic acid molecule or a second vector encoding a second polypeptide construct when that second polypeptide construct is the same or different from the first polypeptide construct. The second nucleic acid molecule or second vector is present necessarily when the two polypeptide constructs are different (heterodimeric), but are not necessary when the constructs are the same (homodimeric).

There is also provided the use of a polypeptide construct according to the present invention for treatment of a medical condition, disease or disorder; wherein the medical condition, disease or disorder comprises, but is not limited to, cancer, ocular diseases, fibrotic diseases, or genetic disorders of connective tissue and immune disorders. The polypeptide construct of the present invention may comprise a C H 2 and C H 3 or only a C H 2 from an antibody heavy chain that is of human origin. For example, and without wishing to be limiting, the antibody heavy chain may be selected from the group consisting of a human lgG1 and lgG2. In embodiments, the constant domain in the constructs is C H 2 per se, or C H 3 per se or C H 2-C H 3. Suitably, the antibody heavy chain component provides for disulfide crosslinking between single chain polypeptide constructs that are the same or different. Also suitably, the antibody heavy chain provides for protein A-based isolation of the dimeric polypeptide that is produced by the host cells.

In embodiments the receptor ectodomain region comprises two independently selected ectodomains that are linked in tandem, i.e., in a linear manner. In some embodiments, the ectodomains are the same in sequence, or least the same with respect to their target ligand.

The present invention also provides a nucleic acid molecule encoding the polypeptide constructs as described herein. A vector comprising the nucleic acid molecule just described is also encompassed by the invention. The invention also includes a transgenic cellular host comprising the nucleic acid molecule or a vector as described herein; the cellular host may further include a second nucleic acid molecule or a second vector encoding a second polypeptide construct different from the first polypeptide construct. Systems used to produce the present polypeptides can be secretion systems, particularly in the case where dimerization through disulfide bridges is required, and the expression polynucleotides thus encode secretion signals that are cleaved by the host upon secretion into the culturing medium.

Compositions comprising one or more than one independently selected polypeptide construct described herein and a pharmaceutically-acceptable carrier, diluent, or excipient are also encompassed by the present invention.

These and other features of the invention will now be described by way of example, with reference to the appended drawings, wherein:

BRIEF DESCRIPTION OF THE DRAWINGS FIGURE 1 A/B is a schematic drawing of the of the TGF-β Type II receptor ectodomain (TpRII- ECD; also abbreviated as T2m) and the single chain fusion of two T2m domains (also abbreviated T22d35) (Fig. 1A); Fig. 1 B provides the corresponding amino acid sequences, wherein the natural linker sequences (SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8) are underlined and the sequence of the TpRII structured domain (SEQ ID NO:4) is shown in bold.

FIGURE 2 A-G, where FIG. 2A/B/C is a schematic representation of the fusion of the T2m and T22d35 modules to the N-termini of the heavy chains of an lgG2 Fc region (2A) in order to generate fusion proteins T2m-Fc (2B) and T22d35-Fc (2C); Figure 2D provides the amino acid sequence of the T2m-Fc and T22d35-Fc fusion proteins (SEQ ID NO:9, SEQ ID NO:10). Figure 2E provides aligned sequenced of additional variants of the linker region between the Fc and ECD region in the T22d35-Fc fusions. Figure 2F and 2G provide the amino acid sequence of the T22d35-Fc linker variants using a human lgG1 Fc (Fig. 2F; SEQ ID NO: 14, SEQ ID NO:17, SEQ ID NO:20) and a human lgG2 Fc region (Fig. 2G; SEQ ID NO:23). The natural linker sequences (SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8) are underlined, the sequence of the TpRII structured domain (SEQ ID NO:4) is shown in bold and the human IgG Fc sequence variants (SEQ ID NO:12, SEQ ID NO:15, SEQ ID NO:18, SEQ ID NO:24) are shown in bold-italics.

FIGURE 3 A/B/C shows the preparative SEC elution profile for the T2m-Fc fusion protein (3A); Fractions (Fr.) 6-1 1 were pooled and concentrated. Protein integrity of the SEC purified material was then assessed by UPLC-SEC profile (3B) and SDS-PAGE assessment under non-reducing (NR) and reducing (R) conditions (3C).

FIGURE 4 A/B/C shows the preparative SEC elution profile for the T22d35-Fc fusion protein (4A); Fr. 7-10 were pooled and concentrated. Protein integrity of the SEC purified material was then assessed by UPLC-SEC profile (4B) and SDS-PAGE assessment under non-reducing (NR) and reducing (R) conditions (4C).

FIGURE 5 A/B provides SDS-PAGE analysis of the protein A purified T22d35-Fc, T22d35-Fc- lgG2-v2(CC), T22d35-Fc-lgG1-v1 (CC), T22d35-Fc-lgG1 -v2(SCC), T22d35-Fc-lgG1 -v3(GSL-CC), hlgG1 FcAK(C)-T22d35, MgG1 FcAK(CC)-T22d35 and MgG2FcAK(CC)-T22d35 variants under non-reducing (A) and reducing (B) conditions.

FIGURE 6 A/B/C provides the percentage of intact monomer (A), aggregates (B), and fragments (C) of the various fusion proteins, indicating that there are advantages to expressing the T22d35 doublet at the N-terminus of an IgG Fc portion. The table lists the numerical differences in the parameters that were analyzed.

FIGURE 7 A/B/C provides a functional evaluation of the T2m-Fc and T22d35-Fc fusion proteins compared to the non-Fc-fused single chain T22d35 trap in a A549 IL-1 1 release assay. Neutralization of TGF-βΙ (5A), -β2 (5B), -β3 (5C) was assessed and calculated as a % of the TGF-β control (Average of a triplicate experiment +/- SD). The table lists the calculated IC 50 values calculated in Graphpad Prism (4-PL algorithm ((log (inhibitor) vs. response - variable slope (four parameters)). FIGURE 8 provides a functional evaluation of the T22d35-Fc, T22d35-Fc-lgG2-v2(CC), T22d35- Fc-lgG1 -v1 (CC), T22d35-Fc-lgG1 -v2(SCC), and T22d35-Fc-lgG1-v3(GSL-CC) compared to the C-terminal Fc-fused T22d35 trap variants in an A549 IL-1 1 release assay. Neutralization of TGF- β1 , was assessed and calculated as a % of the TGF-βΙ control (Average of a triplicate experiment +/- SD). The table lists the calculated IC 50 values calculated in Graphpad Prism (4-PL algorithm ((log (inhibitor) vs. response - variable slope (four parameters)).

FIGURE 9 provides a functional evaluation of the neutralization TGF-βΙ , -β2, and β3 by the T22d35-Fc-lgG1 -v1 (CC) variant in an A549 IL-1 1 release assay. TGF-β neutralization was assessed and calculated as a % of the TGF-β control (Average of a triplicate experiment +/- SD). The table lists the calculated IC 50 values calculated in Graphpad Prism (4-PL algorithm ((log (inhibitor) vs. response - variable slope (four parameters)).

FIGURE 10 Functional in vivo evaluation of the T22d35-Fc fusion protein in a syngeneic MC-38 mouse colon carcinoma model. (A) Tumor volumes were calculated as described and plotted as average tumor values +/- SD per cohort. A two-way ANOVA was used to analyse whether statistically significant differences between the calculated average tumor volumes in the T22d35- Fc and CTL IgG treatment cohorts over the course of time. In addition, MC-38 tumor growth (calculated volume) was plotted per individual mouse for the CTL IgG (B) and T22d35-Fc treated cohorts (C).

Additional aspects and advantages of the present invention will be apparent in view of the following description. The detailed descriptions and examples, while indicating preferred embodiments of the invention, are given by way of illustration only, as various changes and modifications within the scope of the invention will become apparent to those skilled in the art DETAILED DESCRIPTION OF THE INVENTION

There are now provided polypeptide constructs that bind to and neutralize all transforming growth factor beta (TGF-βΙ , β2 and β3) isoforms. These polypeptides exploit the TGF-β receptor ectodomains to trap or sequester various TGFp species including TGF-βΙ and TGF- β3, and to some extend TGF-p2. The potency with which the present polypeptide constructs neutralize TGF-βΙ and -β3 is surprisingly far greater than related constructs, as demonstrated herein. For this reason, the present constructs are expected to be especially useful as pharmaceuticals for the treatment of medical indications such as cancer, fibrotic diseases and certain immune disorders.

The present polypeptide constructs comprise two TpR-ECDs, such as TpRII-ECDs, that are linked in tandem (C-terminus to N-terminus) and further comprise an antibody constant domain that comprises at least the second constant domain (C H 2) and/or third constant domain (C H 3) of an antibody heavy chain. The antibody constant domain (Fc) is coupled at its N-terminus to the C-terminus of the ectodomain. Having the ectodomain as a doublet, and having that doublet coupled to the N-terminus of the antibody constant domain, provides a "trap" with an enhanced the neutralization potency by a factor of 615 for TGFpi and a factor 24 for TGFp3 compared to the construct having a single ECD coupled to the N-terminus of the antibody constant domain. As used herein, the term TpRII-ECD refers to the extracellular region of the TGF-β Type II receptor that binds to the TGF-β ligand. In a preferred embodiment of the present constructs, the TGFpRII ectodomain is the ectodomain of the TGFpR species (i.e. TpRII-ECD) comprising the sequence that forms a stable three-dimensional folded structure: QLCKFCDVRFSTCDNQKSCMSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPY HD FILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNIIF (SEQ ID NO:4).

In a related form that comprises flexible natural flanking sequence, the ECD can include the underlined structures, as shown below:

IPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSCMSNCSITSICEKP QEVCVA VWRKNDENITLETVCHDPKLPYHDFILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDN IIFS EEYNTSNPD (SEQ ID NO: 1)

This sequence binds to the TGF-β ligand isotypes designated TGF-βΙ and TGF-p3. Binding affinity for TGF-p2 is less. In the present polypeptide constructs, the two TpRII-ECDs may comprise the same sequence. The two ectodomains are linked in tandem, wherein the result is a linear polypeptide in which the C-terminus of one ectodomain is linked to the N-terminus of another ectodomain.

The two ectodomains can be linked by direct fusion such that additional amino acid residues are not introduced. Alternatively, additional amino acid residues can form a linker that couples the two receptor ectodomains in tandem. In the protein construct of the present invention, the first and second regions of the polypeptide construct of the present invention are also linked. By the term "linked", it is meant that the two regions are covalently bonded. The chemical bond may be achieved by chemical reaction, or may be the product of recombinant expression of the two regions in a single polypeptide chain. In a specific, non-limiting example, the C- terminus of the first region is linked directly to the N-terminus of the second region, that is, no additional "linker" amino acids are present between the two regions. In the case where no linker is present, that is to say direct fusion of the two regions, there will be a direct link between the C-terminus of the full ectodomain and the N-terminus of the antibody constant regions C H 2-C H 3. For example, in fusing an Fc variant (SEQ ID NO:12, SEQ ID NO:15, SEQ ID NO:18, SEQ ID NO:24) to the SEQ ID NO:1 via the intrinsically disordered linker with SEQ ID NO:8, which is part of the TpRII-ECD having SEQ ID NO:1 (i.e., no additional "linker" amino acids added), one connects the aspartic acid at the last position of SEQ ID NO:1 to a glutamic acid, a threonine, a valine or a valine found at the first position of SEQ ID NO:12, SEQ ID NO:15, SEQ ID NO:18, or SEQ ID NO:24, respectively.

A common practice when producing fusion constructs is to introduce glycine or glycine-serine linkers (GSL) such as GGGGS or [G4S] n (where n is 1 , 2, 3, 4 or 5 or more, such as 10, 25 or 50) between the fused components. As taught in the above paragraph, the polypeptide fusions of the present invention can be produced by direct linkage without use of any additional amino- acid sequence except those present in the Fc region and in the receptor ectodomain region. One thus can refrain from utilizing foreign sequences as linkers, providing an advantage due to their potential for undesired immunogenicity and their added molecular weight. Entropic factors are also a potential liability for glycine and glycine-serine linkers, which are highly flexible and may become partially restricted upon target binding, hence causing a loss of entropy unfavourable to binding affinity. Therefore, only the flexible, intrinsically disordered N-terminal regions of the TGFpRII-ECD were employed as natural linkers in embodiments of the present invention. However, the particular amino acid compositions and lengths of these intrinsically disordered linkers (e.g., SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8) precluded accurate prediction of whether the resulting direct-fusion constructs will have the required geometry and favourable molecular interactions for correct binding to their intended dimeric ligands. In other embodiments, the fusion polypeptide may include a flexible artificial GSL, as exemplified in the construct in SEQ ID NO:20, where the GS linker with the SEQ ID NO:21 is introduced between the aspartic acid (D) at the last position of TpRII-ECD having SEQ ID NO:1 and the threonine (T) at the first position of the Fc region variant having SEQ ID NO:15.

The first and second regions of the polypeptide construct are, in embodiments, connected by natural intrinsically disordered polypeptide linkers selected from the group consisting of SEQ ID NO:8, 13, 16, 19, 25, and a sequence substantially identical thereto. In other embodiments, the regions of the polypeptide constructs are connected by flexible linkers selected from the group consisting of SEQ ID NO:21 and SEQ ID NO:22, and a sequence substantially identical thereto.

In this embodiment, one region of the present polypeptide constructs comprises a TpRII-ECD doublet comprising first and second receptor ectodomains linked in tandem by the natural intrinsically disordered polypeptide linker with SEQ ID NO: 6, and having amino acid sequence comprising:

IPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSCMSNCSITSICEKP QEVCVA VWRKNDENITLETVCHDPKLPYHDFILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDN IIFS EEYNTSNPDIPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSCMSNCSIT SIC EKPQEVCVAVWRKNDENITLETVCHDPKLPYHDFILEDAASPKCIMKEKKKPGETFFMCS CSS DECNDNIIFSEEYNTSNPD (SEQ ID NO:5).

The present constructs also comprise a region that comprises an antibody constant domain that comprises at least the second constant domain (C H 2) and/or third constant domain (C H 3) of an antibody heavy chain. The antibody constant domain is coupled at its N-terminus to the C-terminus of the ectodomain doublet, so that the orientation of the construct is a single chain 0f TpRII-ECD(link)TpRII-ECD(link)C H 2-C H 3.

The antibody constant domain provides for cross-linking between two of the present polypeptide constructs. This is achieved when the expressed polypeptide constructs are secreted from their expression host. Thus, production of the single chain polypeptide provides the construct in a dimeric form in which the two constructs are cross-linked via disulfide bridges that involve one or more cysteine residues within each of the antibody constant domains present in each of the constructs.

The antibody constant domain present in the construct is desirably sourced from an IgG constant region, and especially from the constant domain of either lgG1 or lgG2.

The constructs provided are monofunctional in the sense that the constant region itself may have no particular activity, other than to act as a structure through which dimers of the polypeptide constructs can form. These minimal constant regions can also be altered to provide some benefit, by incorporating the corresponding hinge regions and optionally changing the cysteine residue composition. Thus, some or all of the cysteine residues involved in bridging the two Fc fragments or naturally used to bridge between the heavy and light chains of a full-length antibody can be replaced or deleted. One advantage of minimizing the number of cysteine residues is to reduce the propensity for disulphide bond scrambling, which could promote aggregation. For example, these cysteine residues and alteration thereof are seen in the natural or non-natural linker sequences located around the junction of the first and second regions of the polypeptide constructs and which are listed below:

SEEYNTSNPDTHTCPPCPAPE (SEQ ID NO:16), SEEYNTSNPDVEPKSSDKTHTCPPCPAPE (SEQ ID NO:19), SEEYNTSNPDGGGSGGGSGGGTHTCPPCPAPE (SEQ ID NO:22) incorporating variations of human lgG1 hinge sequence; and SEEYNTSNPDERKCCVECPPCPAPP (SEQ ID NO:13) and SEEYNTSNPDVECPPCPAPP (SEQ ID NO:25) incorporating variations of human lgG2 hinge sequence; and a sequence substantially identical thereto. Not all of the naturally-occurring inter-hinge disulfide bonds need to be formed for the Fc homodimerization to occur, while noting that the stability of the Fc homodimer may depend on the number of intermolecular disulphide bridges.

In the present disclosure, an "antibody", also referred to in the art as "immunoglobulin" (Ig), refers to a protein constructed from paired heavy and light polypeptide chains. The structure of an antibody and of each of the domains is well established and familiar to those of skill in the art, though is summarized herein. When an antibody is correctly folded, each chain folds into a number of distinct globular domains joined by more linear polypeptide sequences; the immunoglobulin light chain folds into a variable (V L ) and a constant (C L ) domain, while the heavy chain folds into a variable (V H ) and three constant (C H 1 , C H 2, C H 3) domains. Once paired, interaction of the heavy and light chain variable domains (V H and V L ) and first constant domain (C L and C H 1 ) results in the formation of a Fab (Fragment, antigen-binding) containing the binding region (Fv); interaction of two heavy chains results in pairing of C H 2 and C H 3 domains, leading to the formation of a Fc (Fragment, crystallisable). Characteristics described herein for the C H 2 and C H 3 domains also apply to the Fc.

In the present invention and its specific embodiments, the polypeptide constructs that exhibit significantly enhanced potency comprise the following:

T22d35-Fc:

IPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSCMSNCSITSICEKP QEVCVA VWRKNDENITLETVCHDPKLPYHDFILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDN IIFS EEYNTSNPDIPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSCMSNCSIT SIC EKPQEVCVAVWRKNDENITLETVCHDPKLPYHDFILEDAASPKCIMKEKKKPGETFFMCS CSS DECNDNIIFSEEYNTSNPDERKCCVECPPCPAPPVAGPSVFLFPPKPKDTLMISRTPEVT CVV VDVSHEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTFRVVSVLTVVHQDWLNGKEYKCKV S NKGLPAPIEKTISKTKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDISVEWESN GQP ENNYKTTPPMLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG

(SEQ ID NO:10)

T22d35-Fc-lgG2-v2 (CC):

IPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSCMSNCSITSICEKP QEVCVA

VWRKNDENITLETVCHDPKLPYHDFILEDAASPKCIMKEKKKPGETFFMCSCSSDEC NDNIIFS

EEYNTSNPDIPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSCMSNC SITSIC

EKPQEVCVAVWRKNDENITLETVCHDPKLPYHDFILEDAASPKCIMKEKKKPGETFF MCSCSS

DECNDNIIFSEEYNTSNPDVECPPCPAPPVAGPSVFLFPPKPKDTLMISRTPEVTCV VVDVSH EDPEVQFNWYVDGVEVHNAKTKPREEQFNSTFRVVSVLTVVHQDWLNGKEYKCKVSNKGL P APIEKTISKTKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDISVEWESNGQPEN NYK TTPPMLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG (SEQ ID NO:23)

T22d35-Fc-lgG1 -v1 (CC):

IPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSCMSNCSITSICEKP QEVCVA VWRKNDENITLETVCHDPKLPYHDFILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDN IIFS EEYNTSNPDIPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSCMSNCSIT SIC EKPQEVCVAVWRKNDENITLETVCHDPKLPYHDFILEDAASPKCIMKEKKKPGETFFMCS CSS DECNDNIIFSEEYNTSNPDTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVW DVS HEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKA L PAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPE NNY KTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG (SEQ ID NO:14)

T22d35-Fc-lgG1 -v2 (SCC):

IPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSCMSNCSITSICEKP QEVCVA VWRKNDENITLETVCHDPKLPYHDFILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDN IIFS EEYNTSNPDIPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSCMSNCSIT SIC EKPQEVCVAVWRKNDENITLETVCHDPKLPYHDFILEDAASPKCIMKEKKKPGETFFMCS CSS DECNDNIIFSEEYNTSNPDVEPKSSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISR TPEV TCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEY K CKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVE WES NGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSL S PG (SEQ ID NO:17); and

T22d35-Fc-lgG1 -v3 (GSL-CC):

IPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSCMSNCSITSICEKP QEVCVA VWRKNDENITLETVCHDPKLPYHDFILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDN IIFS EEYNTSNPDIPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSCMSNCSIT SIC EKPQEVCVAVWRKNDENITLETVCHDPKLPYHDFILEDAASPKCIMKEKKKPGETFFMCS CSS DECNDNIIFSEEYNTSNPDGGGSGGGSGGGTHTCPPCPAPELLGGPSVFLFPPKPKDTLM IS RTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWL N GKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPS DIAV EWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQK SLSLSPG (SEQ ID NO:20) In a specific embodiment, the polypeptide construct comprises a polypeptide of the present invention that exhibits significantly enhanced potency, for example a polypeptide the exhibits significant enhanced potency may comprise SEQ ID NO: 10, SEQ ID N0.14, SEQ ID NO:17, SEQ ID NO: 20 or SEQ ID NO:23. In another specific embodiment, the polypeptide construct may be a homodimer comprising two polypeptides that exhibit significant enhanced potency; for example if the polypeptide construct that exhibits significant enhanced potency is SEQ ID NO: 14, the polypeptide construct is a homodimer comprising two polypeptide constructs wherein each polypeptide construct comprising SEQ ID NO.14. Likewise, if the polypeptide that exhibits enhanced potency is SEQ ID NO: 10, 14, 17, 20, or 23, a homodimer of the present invention likewise comprises two polypeptide constructs wherein each polypeptide of the homodimer is SEQ ID NO: 10, 14, 17, 20 or 23 respectively.

As noted, these single chain polypeptide constructs will dimerize when secreted from a production host, yielding a dimeric polypeptide construct comprising two single chain polypeptides linked by way of disulfide bridges that form between the constant domains of the two single chain polypeptides.

By "significantly enhanced potency" we mean that the effect or activity of a present polypeptide construct in this dimeric form is greater than a counterpart construct when measured in an assay relevant for assessing the biological activity of TGF-β. Appropriate means for making this determination are exemplified herein. For example, the N-terminal Fc-fused T22d35 doublets neutralizes TGF-β to a much better extend than the N-terminally Fc-fused T2m singlet as was illustrated by the TGF-p-induced IL-1 1 release by A549 cells (Fig. 7).

It is observed that fusion constructs of this type have advantages relative to several other versions of TpRII receptor-ectodomain based molecules, including non-Fc fused bivalent TGF- β receptor ectodomain constructs (such as the T22d35 doublet) and constructs in which a single receptor ectodomain is fused to the N-terminus of an Fc region. In particular, the presently provided Fc fusion constructs have improved manufacturability due to the presence of the Fc region (for example, purification can be accomplished using protein A chromatography). The Fc region also allows for improved circulating half-lives. Importantly, the present constructs have substantially higher TGF-β neutralization potencies compared to the singlet fusion (T2m-Fc) and non-Fc-fused doublet ectodomain (T22d35). The N-terminally fused TGF-β ECD doublet Fc constructs (T22d35-Fc) provided exhibit advantages with respect to significant improvement in TGF-β ligand neutralizing potency (as shown, for example, in the over 970-fold improvement in TGF-βΙ neutralization relative to non-Fc fused doublet, as shown in Fig. 7). Additionally, they exhibit improved manufacturability, as demonstrated by biophysical analysis showing a >99% monomeric content (i.e. the minimal presence of aggregates and the absence of fragments of the purified N-terminally fused T22d35-Fc constructs) (as illustrated in Fig. 6). Thus, an advantage of the present invention is an unexpected high potency of TGF-β ligand neutralization, including some degree of neutralization of TGF-p2, which is not observed with the T2m-Fc (Fc-singlet) or T22d35 (non Fc-fused) constructs.

In specific embodiments, the second region of the polypeptide construct of the present invention is selected from a group of sequences displaying variation in the N-terminal sequence as exemplified by SEQ ID NO:12, 15, 18, 24. These may differ in length and the number of cysteine residues retained from the hinge region as a means to modulating the degree of Fc-region dimerization and hence impacting on both efficacy and manufacturability. Thus, in embodiments, the polypeptide construct comprises a variation in the constant domain, wherein at least one cysteine residue involved in cross-linking is deleted or substituted. Suitable substitutions include serine or alanine, and preferably by serine.

A substantially identical sequence may comprise one or more conservative amino acid mutations that still provide for proper folding upon secretion into the culturing medium. It is known in the art that one or more conservative amino acid mutations to a reference sequence may yield a mutant peptide with no substantial change in physiological, chemical, physico- chemical or functional properties compared to the reference sequence; in such a case, the reference and mutant sequences would be considered "substantially identical" polypeptides. A conservative amino acid substitution is defined herein as the substitution of an amino acid residue for another amino acid residue with similar chemical properties (e.g. size, charge, or polarity). These conservative amino acid mutations may be made to the framework regions while maintaining the overall structure of the constant domains; thus the function of the Fc is maintained. In a specific, non-limiting example, the first region of the polypeptide construct of the present invention may comprise a TGF-β receptor type II, such as:

IPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSCMSNCSITSICEKP QEVCVA VWRKNDENITLETVCHDPKLPYHDFILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDN IIFS EEYNTSNPD (SEQ ID NO:1 , also referred to herein as T2m). In preferred embodiments, the polypeptide constructs comprise a TpRII-ECD "doublet", in which a TpRII-ECD is linked in tandem with another TpRII-ECD, which ectodomains can be the same or different TGF-β superfamily receptor ectodomains, such as: IPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSCMSNCSITSICEKPQEV CVA

VWRKNDENITLETVCHDPKLPYHDFILEDAASPKCIMKEKKKPGETFFMCSCSSDEC NDNIIF

-linker- QLCKFCDVRFSTCDNQKSCMSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPY HD FILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNIIFSEEYNTSNPD (SEQ ID NO:5, also referred to herein as T22d35) where in one non-limiting embodiment the linker corresponds to SEQ ID NO:6; and a sequence substantially identical thereto. "Substantially identical" is as defined above. The ectodomain doublet can incorporate the same or different ectodomains, both belonging to the TGFp superfamily receptor family. In embodiments, the ectodomains bind the same target. In other embodiments, the ectodomains are of the same receptor species. In other embodiments, the ectodomains are identical and thus are homomeric.

For example, the polypeptide constructs of the present invention may have a TGF-β neutralization potency selected from the group consisting of at least 900-fold and 200-fold, more potent than the T22d35 doublet alone for TGFpi and TGF-p3, respectively. For example, in the IL-1 1 release assay the T22d35-Fc doublet construct is approximately 972-fold more potent in neutralizing TGF-βΙ and approximately 243-fold more potent in neutralizing TGF-p3, when compared with the non-Fc-fused T22d35 doublet alone. In another example, the potency of the construct is at least 600-fold and at least 20-fold greater for neutralizing TGF-βΙ and TGF-p3, respectively, than a construct in which the antibody constant domain is coupled to a single ectodomain rather than to a doublet. The polypeptide constructs of the present invention may have an at least 615-fold and 24-fold better neutralization potency for TGF-βΙ and TGF-p3, respectively, when compared to the potency of a construct in which the antibody constant domain is coupled to a single ectodomain rather than to a doublet.

The neutralizing potency can be summarized as follows: the neutralizing potency of the Fc- fused doublet (ECD-ECD-Fc) is greater than the Fc-fused ECD monomer (ECD-Fc); i.e. ECD- ECD-Fc > ECD-Fc, whereas the ECD-Fc is more potent than the non-Fc-fused doublet (ECD- ECD) and the non-Fc fused doublet ECD is more potent than the non-Fc fused singlet ECD; i.e. ECD-ECD-Fc » ECD-Fc > ECD-ECD » ECD). In terms of manufacturability, the presence of an Fc protein allows for Protein A purification and prevents having to use cleavable tags. In addition, positioning the singlet or doublet ECD at the N-terminus of the Fc portion prevents aggregation issues due to the inappropriate pairing of the cysteine residues in the hinge region of the Fc portion. Therefore, fusion of the ECD singlet or doublet to the N- terminus of the Fc portion provides an improved manufacturability over C-terminal fusions (N- terminal fusions have a higher percentage of monomeric species, less aggregates, less fragments). In addition an unexpected significant increase is observed in TGF-β neutralization potency for all TGF-β isotypes for the N-terminal Fc fused doublet ECD compared to the N- terminal Fc fused T2m singlet ECD.

Additionally, when the polypeptide constructs of the present invention include a TpRII-ECD that binds TGF-β, the polypeptide construct may neutralize, to varying extents, all three isotypes of TGF-β (that is, TGF-βΙ , TGF-p2, and TGF-p3).

The polypeptide constructs of the present invention have, as assessed in cell-based assays, TGF-β neutralizing potencies that are significantly higher (20-fold or more) than those of bivalent comparator polypeptides, i.e. non-Fc-fused T22d35 (doublet alone) and T2m-Fc (Fc fused singlet). Within the series of polypeptide constructs of the present invention, those that contain two or more copies of the TpRII-ECD fused to the N-terminus of the Fc constant region have potencies that are higher than those constructs that contain only one copy, as assessed in cell based assays.

The polypeptide construct of the present invention is expressed as a single polypeptide chain. Once expressed, the polypeptide construct of the present invention forms a dimer wherein the C H 2 and C H 3 domains of the respective polypeptide constructs interact to form a properly assembled Fc region such as occurs when the expressed products are secreted into the culturing medium.

The polypeptide construct of the present invention may also comprise additional sequences to aid in expression, detection or purification of a recombinant antibody or fragment thereof. Any such sequences or tags known to those of skill in the art may be used. For example, and without wishing to be limiting, the antibody or fragment thereof may comprise a targeting or signal sequence (for example, but not limited to ompA), a detection/purification tag (for example, but not limited to c-Myc, His 5 , His 6 , or His 8 G), or a combination thereof. In another example, the signal peptide may be M DWTWR I LFLVAAATGTH A (SEQ ID NO:1 1 ). In a further example, the additional sequence may be a biotin recognition site such as that described in [WO/1995/04069] or in [WO/2004/076670]. As is also known to those of skill in the art, linker sequences may be used in conjunction with the additional sequences or tags, or may serve as a detection/purification tag. Suitably, the constant region comprises a protein A binding site (residing typically between about C H 2 and C H 3) that permits the single chain polypeptide to be extracted/isolated using a protein A affinity approach.

The present invention also encompasses nucleic acid sequences encoding the molecules as just described above. Given the degeneracy of the genetic code, a number of nucleotide sequences would have the effect of encoding the desired polypeptide, as would be readily understood by a skilled artisan. The nucleic acid sequence may be codon-optimized for expression in various micro-organisms. The present invention also encompasses vectors comprising the nucleic acids as just described, wherein the vectors typically comprise a promoter and signal sequence that are operably linked to the construct-encoding polynucleotide for driving expression thereof in the selected cellular production host. The vectors can be the same or different provided both result in secretion of the dimeric polypeptide construct.

Furthermore, the invention encompasses cells, also referred to herein as transgenic cellular host, comprising the nucleic acid and/or vector as described, encoding a first polypeptide construct. The host cells may comprise a second nucleic acid and/or vector encoding a second polypeptide construct different from the first polypeptide construct. The co-expression of the first and second polypeptide constructs may lead to the formation of heterodimers.

The present invention also encompasses a composition comprising one or more than one polypeptide construct as described herein. The composition may comprise a single polypeptide construct as described above, or may be a mixture of polypeptide constructs. The composition may also comprise one or more than one polypeptide construct of the present invention linked to one or more than one cargo molecule. For example, and without wishing to be limiting in any manner, the composition may comprise one or more than one polypeptide construct of the present invention linked to a cytotoxic drug in order to generate an antibody-drug conjugate (ADC) in accordance with the present invention.

The composition may also comprise a pharmaceutically acceptable diluent, excipient, or carrier. The diluent, excipient, or carrier may be any suitable diluent, excipient, or carrier known in the art, and must be compatible with other ingredients in the composition, with the method of delivery of the composition, and is not deleterious to the recipient of the composition. The composition may be in any suitable form; for example, the composition may be provided in suspension form, powder form (for example, but limited to lyophilised or encapsulated), capsule or tablet form. For example, and without wishing to be limiting, when the composition is provided in suspension form, the carrier may comprise water, saline, a suitable buffer, or additives to improve solubility and/or stability; reconstitution to produce the suspension is effected in a buffer at a suitable pH to ensure the viability of the antibody or fragment thereof. Dry powders may also include additives to improve stability and/or carriers to increase bulk/volume; for example, and without wishing to be limiting, the dry powder composition may comprise sucrose or trehalose. In a specific, non-limiting example, the composition may be so formulated as to deliver the antibody or fragment thereof to the gastrointestinal tract of the subject. Thus, the composition may comprise encapsulation, time- release, or other suitable technologies for delivery of the antibody or fragment thereof. It would be within the competency of a person of skill in the art to prepare suitable compositions comprising the present compounds.

The constructs of the present invention may be used to treat diseases or disorders associated with over-expression or over-activation of ligands of the TGF-β superfamily. The disease or disorder can be selected from, but not limited to, cancer, ocular diseases, fibrotic diseases, or genetic disorders of connective tissue. In the field of cancer therapy, it has recently been demonstrated that TGF-β is a key factor inhibiting the antitumor response elicited by immunotherapies, such as immune checkpoint inhibitors (ICI's) (Hahn & Akporiaye, 2006). Specifically, therapeutic response to ICI antibodies results primarily from the re-activation of tumor-localized T-cells. Resistance to ICI antibodies is attributed to the presence of immunosuppressive mechanisms that result in a dearth of T- cells in the tumor microenvironment. Thus, it is now recognized that in order to elicit responses in resistant patients, ICI antibodies need to be combined with agents that can activate T-cells and induce their recruitment into the tumor, i.e. reversing of the "non-T-cell-inflamed" tumor phenotype. One publication noted that overcoming the non-T-cell-inflamed tumor microenvironment is the most significant next hurdle in immuno-oncology (Gajewski, 2015).

We have shown using a proof-of-principle TGF-β trap, T22d35, that blocking of TGF-β effectively reverses the "non-T cell inflamed" tumor phenotype (Zwaagstra et al, 2012). This positions anti-TGF-β molecules as potential synergistic combinations with ICI's and other immunotherapeutics. In support of this, a 2014 study (Holtzhausen et al., ASCO poster presentation) examined effects of a TGF-β blocker when combined an anti-CTLA-4 antibody in a physiologically-relevant transgenic melanoma model. The study demonstrated that while anti-CTLA-4 antibody monotherapy failed to suppress melanoma progression, the combination of the TGF-β antagonist and anti-CTLA-4 antibody significantly and synergistically suppressed both primary melanoma tumor growth as well as melanoma metastasis. These observations correlated with significant increases in effector T-cells in melanoma tissues. We show herein that the present polypeptides having the basic structure that is T22d35-Fc significantly reduce tumor growth in a syngeneic mouse MC-38 colon cancer model. This thus positions anti-TGF-β molecules to be used in a potential synergistic combination with other immunotherapeutics. The present constructs can be useful to treat fibrotic diseases including those that affect any organ of the body, including, but not limited to kidney, lung, liver, heart, skin and eye. These diseases include, but are not limited to, chronic obstructive pulmonary disease (COPD), glomerulonephritis, liver fibrosis, post-infarction cardiac fibrosis, restenosis, systemic sclerosis, ocular surgery-induced fibrosis, and scarring. Genetic disorders of connective tissue can also be treated, and include, but are not limited to, Marfan syndrome (MFS) and Osteogenesis imperfecta (Ol).

The present invention will be further illustrated in the following examples. However, it is to be understood that these examples are for illustrative purposes only and should not be used to limit the scope of the present invention in any manner.

Materials & Methods

Production & purification

Transient CHO expression

The various TpRII-ECD fusion variants (such as T2m-Fc and T22d35-Fc) are each comprised of a heavy chain Fc region, and include the signal sequence MDWTWRILFLVAAATGTHA (SEQ ID NO:1 1) at their N-termini. The DNA coding regions for the constructs were prepared synthetically (Biobasic Inc. or Genescript USA Inc.) and were cloned into the Hind 111 (5' end) and BamH1 (3' end) sites of the pTT5 mammalian expression plasmid vector (Durocher et al, 2002). Fusion proteins were produced by transient transfection of Chinese Hamster Ovary (CHO) cells with the heavy chain T2m or T22d35 fused to the IgG heavy chain (T2m-Hc and T22d35-HC, respectively) construct. Briefly, T2m-HC or T22d35-HC plasmid DNAs were transfected into a 2.5L and 4.6L culture, respectively, of CHO-3E7 cells in Freestyle F17 medium (Invitrogen) containing 4 mM glutamine and 0.1 % Kolliphor p-188 (Sigma) and maintained at 37°C. Transfection conditions were: DNA (80% plasmid construct, 15% AKT plasmid, 5% GFP plasmid): PEIpro (ratio 1 :2.5): PEI(polyethylenimine)pro (Polyplus) (ratio = 1 :2.5). At 24 hours post-transfection, 10% Tryptone N1 feed (TekniScience Inc.) and 0.5 mM Vaporic acid (VPA, Sigma) were added and the temperature was shifted to 32°C to promote the production and secretion of the fusion proteins and then maintained for 15 days post transfection after which the cells were harvested. At final harvest the cell viability was 89.6%. Stable pool CHO expression

CHO BRI/rcTA cell pools were generated by transfecting cells with the vector expressing the target gene encoding the various Fc-fused TpRII-ECD proteins. The day after transfection, the cells were centrifuged for 5 min at 250 rpm and seeded at density of 0.5 x 10 6 cells/mL in selection medium (PowerCH02 medium supplemented with 50 μΜ of methionine sulfoximine). Selection medium was replaced every 2-3 days during 14 to 18 days with inoculation at 0.5x10 6 cells/mL. Cell number and viability were measured with the Cedex Innovatis' automated cell counter Cedex Analyzer as described above. When cell viability reached greater than 95 %, pools were inoculated at 0.2x10 6 cells/mL in 125 or 250 ml_ Erlenmeyer flasks. For the fed-batch culture, cHO BRI/rcTA cell pools were inoculated as described above. At day three post- inoculation, when cells density reached 3.5 to 4.5x10 6 cells/mL, expression of the recombinant protein was induced by adding 2 μg/mL of cumate. MSX concentration was adjusted to 125 μΜ, and F12.7 feed (Irvine Scientific) was added followed by a temperature shift to 32°C. Every 2-3 days, cultures were fed with 5% (v:v) F12.7 and samples were collected for recombinant protein (pA-HPLC) and glucose (VITROS 350, Orthoclinical Diagnostics, USA) concentration determination. Glucose was added in order to maintain a minimal concentration of 17 mM. Purification

The harvest supernatant from the CHO cells was filtered (0.2 μηι) and loaded onto a Protein A MabSelect Sure column (GE Healthcare). The column was washed with 2 column volumes of PBS and protein was eluted with 3 column volumes of 0.1 M sodium citrate pH 3.6. To maximize the yield, the flow through was reloaded onto the Protein A column and eluted as described above. Eluted fractions were neutralized with 1 M Tris, and those containing the fusion proteins were pooled and subsequently loaded onto a Hi-load Superdex S200 26/60 size exclusion chromatography (SEC) column (GE Healthcare) equilibrated in formulation buffer (DPBS without Ca 2+ , without Mg 2+ ). Protein was eluted using 1 column volume formulation buffer, collected into successive fractions and detected by UV absorbance at 280 nm. The main peak SEC fractions containing the fusion proteins were then pooled and concentrated. The integrity of the Prot-A and SEC purified fusion proteins in the pooled fractions was further analyzed by UPLC-SEC and SDS-PAGE (4-15% polyacrylamide) under reducing and non-reducing conditions (SYPRO Ruby staining). For UPLC-SEC, 2-10 μg of protein in DPBS (Hyclone, minus Ca 2+ , minus Mg 2+ ) was injected onto a Waters BEH200 SEC column (1 .7 μηι, 4.6 X 150 mm) and resolved under a flow rate of 0.4 mL/min for 8.5 min at room temperature, using the Waters Acquity UPLC H-Class Bio-System. Protein peaks were detected at 280 nm (Acquity PDA detector).

Cell lines

Human A549 non-small cell lung cancer cells were purchased from ATCC (Cat# CCL-1 85, Cedarlane, Burlington, ON). Cells were cultured in Dulbecco's Modified Eagles Medium (DMEM) supplemented with 5% Fetal Bovine Serum (FBS). MC-38 mouse colon adenocarcinoma cells were purchased from Kerafast (Cat# ENH204, Boston, MA), and cultured in Dulbecco's modified MEM supplemented with 2 mM L-glutamine and 10% fetal bovine serum. Both cell lines were maintained at 37°C, in a humidified atmosphere supplemented with 5% C0 2 .

TGF-β induced A549 cell IL-1 1 release assay

Human A549 lung cancer cells were seeded in 96-well plates (5x10 3 cells/well). The following day 10 pM TGF-β in complete media, in the absence or presence of a serial dilution of TGF-β Trap fusion protein, was incubated for 30 min at RT prior to adding to the cells. After 21 h of incubation (37°C, 5% C0 2 , humidified atmosphere) conditioned medium was harvested and added to MSD Streptavidin Gold plates (Meso Scale Diagnostics, Gaithersburg , MD) that were coated with 2 μg/mL biotinylated mouse anti-human IL- antibody (MAB618, R&D Systems, Minneapolis, MN). After 18h (4°C) plates were washed with PBS containing 0.02% Tween 20 and then 2 μg/mL SULFO-tagged goat anti-human IL- antibody (AF-218-NA, R&D Systems Minneapolis, MN) was added and plates were incubated for 1 h at RT. After a final wash , plates were read in a MESO QuickPlex SQ120 machine (Meso Scale Diagnostics, Gaithersburg , MD). IL-1 1 readouts were expressed as percent IL-1 1 release compared to control cells treated with TGF-β alone. Graphpad Prism (4-PL algorithm ((log (inhibitor) vs. response - variable slope (four parameters)) was used to calculate the IC 50 (the automatic outlier option was used when needed).

In vivo evaluation in a syngeneic mouse colon cancer MC-38 subcutaneous mouse model

Female C57BL/6-Elite mice (5-7 weeks old) were purchased from Charles River Laboratories (Wlmington , MA). Thirteen C57BL/6 mice were injected on day 0 with 3x1 0 5 MC-38 cells subcutaneously into the right flank. When tumors reached a volume of 50-100 mm 3 (day 5) animals were divided in 2 cohorts and treatment was initiated :

• Cohort 1 (7 animals): Isotype control (CTL IgG; BioxCell InVivo MAb Rat lgG2b, anti-KLH;

Clone LTF-2, Cat# BE0090); 200 in 100 fi L phosphate-buffered saline (PBS), intraperitoneal (i.p.) on day 5, 7, 9, and 1 1 . • Cohort 2 (6 animals): T22d35-Fc, 5 mg/kg in 100 μ L PBS, i.p. on day 5, 9, 12, and 16. Tumors were measured twice a week using digital calipers up to 15 days after commencing treatment. Tumor volumes were calculated from these measurements using a modified ellipsoidal formula (T vo! = TT/6x(l_ength x Width x Width)) described previously (Tomayko et al., 1986).

Results & Discussion

Fusion construct design

In order to generate TGF-β traps of interest, we fused the TpRII-ECD singlet (designated T2m) to another such singlet thereby forming the ectodomain doublet (designated T22d35) that was coupled to the N-termini of the heavy chains of a human (h)lgG2 Fc region and a human lgG1 Fc region. Figure 1 shows schematics (Figure 1A) and amino acid sequences (Figure 1 B) of the T2m and T22d35. These modules were fused to the N-termini of the heavy chains of an IgG Fc region (Figure 2A) using several linker variations (Figure 2E) in order to generate the T2m-Fc (Figure 2B) and T22d35-Fc variants (Figure 2C) fusions. The sequences of these fusions are shown in Figure 2D. We also designed variants of T22d35-Fc that explore the number of cysteine residues in the hinge region of the Fc domain, different IgG isotypes (human lgG1 versus lgG2), and sequences of varying length and nature as linkers between T22d35 and the N-terminus of the Fc domain (Figures 2E, 2F & 2G). These variations aim at exploring and eventually optimizing the functional and manufacturability attributes of the T22d35-Fc design.

Expression and purification

Purification of transient CHO material

The respective fusion protein constructs were expressed transiently in CHO-3E7 cells (see Table 1) after which the conditioned medium was harvested and purified using a protein A affinity column, followed by preparative Size Exclusion Chromatography (SEC). SEC elution profiles of the T2m-Fc (Figure 3A) and T22d35-Fc (Figure 4A) showed that these fusion proteins are relatively pure and devoid of aggregates. Fractions 6-1 1 (T2m-Fc) and 7-10 (T22d35-Fc) were pooled and concentrated to 5.6 mg/mL (T2m-Fc) and 6.03 mg/mL (T22d35-Fc). The final yields were 267 mg and 168 mg for T2m-Fc and T22d35-Fc, respectively. The final products (indicated SEC pooled fractions) were shown to be >99% pure by UPLC-SEC (Figures 3B & 4B). SDS-PAGE assessment (Figures 3C & 4C, Sypro RUBY staining) shows the T2m-Fc and T22d35-Fc bands of ~60 kDa and ~90 kDa under reducing conditions, whereas bands of approximately 90 kDa and 150 kDa can be detected, representing the fully assembled and highly pure T2m-Fc and T22d35-Fc fusion proteins, respectively, under non-reducing conditions. An overview of the production and purification details can be found in Table 1 . Together, these results demonstrate the good manufacturability of the T2m-Fc and T22d35-Fc fusion proteins.

Table 1 : Production (transient pools) and purification details of the T2m-Fc and T22d35-Fc fusion roteins.

Purification of stable CHO pool material

N- and C-terminally Fc-fused T22d35 variants were stably expressed in cHO BRI/rcTA cells in order to compare their level of expression and some of their biophysical properties. The coding region of each variant was ligated into mammalian cell expression plasmids and, after transfection, an enriched pool of cells was selected that stably expressed each of the variants. The main difference between the variants can be found in the amino acid sequence composing the linker region that separates the T22d35 doublet from the Fc domain (in the case of the N- terminal fusions), while for the C-terminal Fc-fusions, the difference between each of the variants is at the extreme amino-terminus of the protein (Table 2).

Table 2: Description of amino acid variations in the linker region of the N- and C-terminal Fc- fused T22d35 fusions (Bold: natural linker sequence; italics: artificial linker sequence). The paired cysteine residues in each of the variants are underlined.

Fusion proteins were purified by protein A affinity and using 100 mM citrate (pH3.6) as the elution buffer. Eluted fusion protein samples were neutralized with 1 M HEPES then subjected to a buffer exchange to DPBS using Zeba spin columns (Table 3), while the integrity of several of the purified fusion proteins was assessed by SDS-PAGE (Figure 5). Purification of each variant was similar. Although many of the properties were very similar between the variants, the potential for aggregation, which is indicative of improper folding, revealed some distinctions. Protein aggregation can be indicative of reduced conformational stability, and can result in decreased activity, efficacy or potency. Size-Exclusion Chromatography-High Performance Liquid Chromatography (SEC-HPLC) was used to determine the purity of each of the N- and C- terminal Fc-fused variants. This method allows for the accurate measurement of the percentage of intact monomeric species as well as the presence of impurities such as aggregates and/or degradation products. As shown in Figure 6, a striking difference can be observed between T22d35 variants expressed as N-terminal Fc fusions and those expressed as C-terminal fusions. In particular, the percentage of intact monomer (Figure 6A) was approximately 99% for all five N-terminal fusion variants (SEQ ID NO:10, SEQ ID NO:14, SEQ ID NO:17, SEQ ID NO:20, SEQ ID NO:23) whereas this was markedly lower for the three C- terminal fusions (SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28). This significant decrease in the percentage of intact monomer results from the accumulation of increased higher molecular weight aggregates observed in all of the C-terminal Fc-fusions (Figure 6B) as well as increased lower molecular weight fragments in two out of the three C-terminal Fc fusions (Figure 6C). In addition, evaluation of the titers of the individual 500 ml_ productions and the average titers of the N-terminal Fc-fused and C-terminally Fc-fused T22d35 productions shows that the N-terminal Fc-fused T22d35 variants can be produced at higher yield compared to the C-terminal fusions (Table 4). Taken together, these results indicate that there are significant and unexpected advantages to expressing the T22d35 doublet at the N-terminus of moieties, such as the Fc portion of an immunoglobulin. Taken together these data demonstrate the enhanced manufacturability of the N-terminal Fc-fused T22d35 proteins.

Table 3: Overview of protein yields of the T22d35 variants after purification of 500 ml_ of stable pool material.

Table 4: Comparison of the titers of the individual N- and C-terminal Fc-fused T22d35 fusions

Functional in vitro assessment

The A549 cell IL-1 1 release assay was used to compare TGF-β neutralization potencies of the T2m-Fc and T22d35-Fc fusion proteins to the non-Fc-fused T22d35 single chain doublet trap, as shown in Figure 7A/B/C. This data shows that for all TGF-β isotypes the potency of T22d35- Fc is superior to that of T2m-Fc and the non-Fc-fused T22d35 single chain trap, with a calculated IC 50 (Table 5) of 0.003348 and 0.003908 nM for TGF-βΙ and TGF-p3, respectively. These values demonstrate potencies that are at least 970-fold and at least 240-fold better than for T22d35 (IC 50 = 3.253 and 0.9491 nM, for TGF-βΙ and TGF-p3, respectively), and 615-fold and 24-fold better than for T2m-Fc (IC 50 = 2.059 and 0.0943 nM, for TGF-βΙ and TGF^3, respectively). In addition, T22d35-Fc neutralizes TGF^2, albeit to a much lesser extend than TGF-βΙ and -β3. In contrast, TGF-p2 neutralization is not observed for either the T2m-Fc or the T22d35 single chain trap. It should be noted that, although the neutralization potency of the T22d35-Fc trap is similar for TGF-βΙ and -β3, the T2m-Fc variant displayed a ~22-fold higher neutralization potency for TGF^3 compared to TGF-βΙ (2.059 nM and 0.0943 nM, respectively). Evaluation of the additional N-terminal Fc-fused T22d35 fusions [T22d35-Fc- lgG2-v2 (CC), T22d35-Fc-lgG1 -v1 (CC), T22d35-Fc-lgG1 -v2 (SCC), and T22d35-Fc-lgG1 -v3 (GSL-CC)] (Figure 8, Table 6) showed that all of these fusions display comparable TGF-βΙ neutralization potencies, which were very similar to the potency of T22d35-Fc. Additional evaluation of the T22d35-Fc-lgG1 -v1 (CC) variant (Fig. 9) confirms that, in line with the T22d35-Fc variant, its neutralization potency for TGF-βΙ and -β3 is very similar (IC 50 = 0.003327 nM and 0.003251 nM, respectively) whereas this potency is much lower for TGF^2 (IC 50 = 17.33 nM). Table 5: Overview of the statistical evaluation of the curves shown in Figure 5 using the 4-PL algorithm ((log (inhibitor) vs. response - variable slope (four parameters)) available in Graphpad Prism.

Table 6: Overview of the statistical evaluation of the curves shown in Figure 7 using the 4-PL algorithm ((log (inhibitor) vs. response - variable slope (four parameters)) available in Graphpad

Prism.

Functional in vivo assessment

The T22d35-Fc fusion protein (SEQ ID NO: 10) was evaluated in vivo using a syngeneic MC-38 mouse colon carcinoma model (Figure 9). The tumor growth in animals treated with the T22d35- Fc fusion was compared to the tumor growth in animals treated with a control IgG (CTL IgG). As shown in Figure 9, no significant differences in tumor growth were observed up to day 11 post- treatment, however on day 15 a significant reduction in tumor growth can be observed in the tumor volume of animals treated with T22d35-Fc, when compared to the CTL IgG (Two-Way ANOVA). This data shows that administration of T22d35-Fc caused a significant inhibition in the growth of the MC-38 tumors compared to the group treated with the CTL IgG suggesting that blockage of TGF-β in vivo can abrogate the growth of tumors in this syngeneic model of colorectal cancer.

LISTING OF SEQUENCES

SEQ Sequence Description ID NO:

1 IPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSC TpRII-ECD including the

MSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPYHDFI structure LEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNIIFSEEYNTSN domain and its

PD natural linkers

(also termed T2m)

2 IPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSC TpRII-ECD

structured

MSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPYHDFI domain with its LEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNIIF natural N- terminal linker

3 QLCKFCDVRFSTCDNQKSCMSNCSITSICEKPQEVCVAVWRKN TpRII-ECD

structured

DENITLETVCHDPKLPYHDFILEDAASPKCIMKEKKKPGETFFMC domain with its

SCSSDECNDNIIFSEEYNTSNPD with natural C- terminal linker

4 QLCKFCDVRFSTCDNQKSCMSNCSITSICEKPQEVCVAVWRKN TpRII-ECD

structured

DENITLETVCHDPKLPYHDFILEDAASPKCIMKEKKKPGETFFMC domain

SCSSDECNDNIIF

5 IPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSC TpRII-ECD- TpRII-ECD fused

MSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPYHDFI dimer including

LEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNIIFSEEYNTSN structured

PDIPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQK domains and natural linkers

SCMSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPYH (also termed DFILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNIIFSEEYN T22d35)

TSNPD

6 SEEYNTSNPDIPPHVQKSVNNDMIVTDNNGAVKF TpRII-ECD natural linker

7 IPPHVQKSVNNDMIVTDNNGAVKF TpRII-ECD _

terminal natural linker

8 SEEYNTSNPD TpRII-ECD terminal natural linker

9 IPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSC T2m-Fc fusion of

T2m with the

MSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPYHDFI hlgG2Fc(CCCC)

LEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNIIFSEEYNTSN Fc region

PDERKCCVECPPCPAPPVAGPSVFLFPPKPKDTLMISRTPEVTC

VVVDVSHEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTFRVVS

VLTVVHQDWLNGKEYKCKVSNKGLPAPIEKTISKTKGQPREPQV

YTLPPSREEMTKNQ VSL TCL VKGFYPSDIS VEWESNGQPENNYK

TTPPMLDSDGSFFL YSKLTVDKSRWQQGNVFSCSVMHEALHNH

YTQKSLSLSPG

10 IPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSC T22d35-Fc fusion of T22d35

MSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPYHDFI with the

LEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNIIFSEEYNTSN hlgG2Fc(CCCC)

PDIPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQK Fc region

SCMSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPYH DFILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNIIFSEEYN TSN PD ERKCCVECPPCPAPPVA GPSVFLFPPKPKDTLMISRTPE VTC VVVD VSHEDPEVQFNWYVDG VEVHNA KTKPREE QFNSTFR VVS VL TVVHQDWLNGKEYKCKVSNKGLPAPIEKTISKTKGQPRE PQVYTLPPSREEMTKNQVSLTCLVKGFYPSDISVEWESNGQPEN NYKTTPPMLDSDGSFFL YSKL TVDKSRWQQGNVFSCS VMHEAL HNHYTQKSLSLSPG

MDWTWRILFLVAAATGTHA signal peptide

ERKCCVECPPCPAPPVAGPSVFLFPPKPKDTLMISRTPEVTCVV hlgG2Fc(CCCC)

Fc region variant

VDVSHEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTFRVVSVL

TVVHQDWLNGKEYKCKVSNKGLPAPIEKTISKTKGQPREPQVYT

LPPSREEMTKNQ VSL TCL VKGFYPSDIS VEWESNGQPENNYKTT

PPMLDSDGSFFL YSKLTVDKSRWQQGNVFSCSVMHEALHNHYT

QKSLSLSPG

SEEYNTSNPDERKCCVECPPCPAPP T22d35-Fc natural linker with the hlgG2Fc(CCCC) hinge region

IPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSC T22d35-Fc-lgG1- v1 (CC) fusion of

MSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPYHDFI T22d35 with the

LEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNIIFSEEYNTSN hlgGIFc(CC) Fc

PDIPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQK region

SCMSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPYH DFILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNIIFSEEYN JSNPDTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCV VVDVSHEDPEVKFNWYVDG VEVHNAKTKPREEQ YNSTYRVVS V LTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVY TLPPSRDEL TKNQ VSL TCL VKGFYPSDIA VEWESNGQPENNYKT TPPVLDSDGSFFL YSKLTVDKSRWQQGNVFSCSVMHEALHNHY TQKSLSLSPG

THTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVS hlgGIFc(CC) Fc region variant

HEDPEVKFNWYVDG VEVHNAKTKPREEQ YNSTYRVVS VL TVLH

QDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPS

RDELTKNQVSLTCLVKGFYPSDIA VEWESNGQPENNYKTTPPVL

DSDGSFFL YSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSL

SLSPG

SEEYNTSNPD THTCPPCPAPE T22d35-Fc-lgG1- v1 (CO natural linker with the hlgGIFc(CC) hinge region

IPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSC T22d35-Fc-lgG1- v2(SCC) fusion

MSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPYHDFI of T22d35 with

LEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNIIFSEEYNTSN the

PDIPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQK hlgGIFc(SCC)

Fc region

SCMSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPYH DFILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNIIFSEEYN JSNPD VEPKSSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMI SRTPEVTC VVVD VSHEDPEVKFNWYVDG VEVHNAKTKPREEQ Y NSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAK GQPREPQVYTLPPSRDELTKNQVSLTCL VKGFYPSDIA VEWESN GQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSV MHEALHNHYTQKSLSLSPG

VEPKSSDKTHTCPPCPAPELLGGPS VFLFPPKPKDTLMISRTPE hlgGIFc(SCC)

Fc region variant

VTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYR

VVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPRE

PQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIA VEWESNGQPEN

NYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEAL

HNHYTQKSLSLSPG

SEEYNTSNPD VEPKSSDKTHTCPPCPAPE T22d35-Fc-lgG1- v2(SCC1 natural linker with the hlgGIFc(SCC) hinge region

IPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSC T22d35-Fc-lgG1- v3(GSL-CC)

MSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPYHDFI fusion of T22d35

LEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNIIFSEEYNTSN with the

PDIPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQK hlgGIFc(CC) Fc region and

SCMSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPYH including an DFILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNIIFSEEYN artificial GS linker

JSNPDGGGSGGGSGGGTHTCPPCPAPELLGGPSVFLFPPKPKD TLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPRE EQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTIS KA KGQPREPQ VYTLPPSRDEL TKNQ VSL TCL VKGFYPSDIA VEW ESNGQPENN YKTTPP VLDSDGSFFL YSKL TVDKSR WQQGNVFS CSVMHEALHNHYTQKSLSLSPG

GGGSGGGSGGG Artificial GS linker of the T22d35-Fc-lgG1- v3(GSL-CC) fusion

SEEYNTSNPDGGGSGGGSGGG THTCPPCPAPE T22d35-Fc-lgG1- v3(GSL-CC) linker including natural and artificial sequences and hlgGIFc(CC) hinge region

IPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSC T22d35-Fc-lgG2- v2(CC) fusion of

MSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPYHDFI T22d35 with the

LEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNIIFSEEYNTSN hlgG2Fc(CC) Fc

PDIPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQK region

SCMSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPYH

DFILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNIIFSEEYN

JSNPD VECPPCPAPPVAGPSVFLFPPKPKDTLMISRTPEVTCVV

VDVSHEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTFRVVSVL

TVVHQDWLNGKEYKCKVSNKGLPAPIEKTISKTKGQPREPQVYT

LPPSREEMTKNQ VSL TCL VKGFYPSDIS VEWESNGQPENNYKTT

PPMLDSDGSFFL YSKLTVDKSRWQQGNVFSCSVMHEALHNHYT

QKSLSLSPG

VECPPCPAPPVAGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSH hlgG2Fc(CC) Fc region variant

EDPEVQFNWYVDGVEVHNAKTKPREEQFNSTFRVVSVLTVVHQ

DWLNGKEYKCKVSNKGLPAPIEKTISKTKGQPREPQVYTLPPSR

EEMTKNQVSLTCLVKGFYPSDISVEWESNGQPENNYKTTPPML

DSDGSFFL YSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSL

SLSPG

SEEYNTSNPD VECPPCPAPP T22d35-Fc-lgG2- v2(CQ natural linker with the hlgG2Fc(CC) hinge region

PPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHED

PEVKFNWYVDG VEVHNAKTKPREEQ YNSTYRVVS VL TVLHQDW

LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDE

L TKNQVSL TCL VKGFYPSDIA VEWESNGQPENNYKTTPPVLDSD

GSFFL YSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLS hlgGI FcAK(C)-

PGIPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQK T22d35

SCMSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPYH

DFILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNIIFSEEYN

TSNPDIPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCD

NQKSCMSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKL PYHDFILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNIIFSE

EYNTSNPD

DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVV

DVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLT

VLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTL

PPSRDEL TKNQ VSL TCL VKGFYPSDIA VEWESNGQPENNYKTTP

PVLDSDGSFFL YSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQ

ACSLSLSPGIPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFS hlgG1 FcAK(CC)-

TCDNQKSCMSNCSITSICEKPQEVCVAVWRKNDENITLETVCHD T22d35

PKLPYHDFILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNII

FSEEYNTSNPDIPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDV

RFSTCDNQKSCMSNCSITSICEKPQEVCVAVWRKNDENITLETV CHDPKLPYHDFILEDAASPKCIMKEKKKPGETFFMCSCSSDECN

DNIIFSEEYNTSNPD

VECPPCPAPPVAGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSH

EDPEVQFNWYVDGVEVHNAKTKPREEQFNSTFRVVSVLTVVHQ

DWLNGKEYKCKVSNKGLPAPIEKTISKTKGQPREPQVYTLPPSR

EEMTKNQVSLTCLVKGFYPSDISVEWESNGQPENNYKTTPPML

DSDGSFFL YSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSL

SLSPGIPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCD hlgG2FcAK(CC)-

NQKSCMSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKL T22d35

PYHDFILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNIIFSE

EYNTSNPDIPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFS

TCDNQKSCMSNCSITSICEKPQEVCVAVWRKNDENITLETVCHD PKLPYHDFILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNII

FSEEYNTSNPD

ATGGATTGGACCTGGAGAATCCTCTTCCTTGTAGCAGCAGCAA

CAGGTACACATGCTATCCCTCCTCATGTTCAAAAGTCCGTTAA

CAACGACATGATCGTCACCGATAACAACGGTGCTGTCAAGTTC

CCACAACTCTGTAAGTTCTGCGATGTGCG 1 1 1 CTCCACATGTG

ATAACCAGAAGTCCTGTATGAGCAACTGCTCAATCACCTCCAT

CTGCGAAAAGCCACAAGAGGTATGCGTAGCTGTATGGCGAAA

GAACGATGAAAACATCACCCTGGAAACCGTCTGTCACGATCCA

AAGCTCCCATACCATGA 1 1 1 CATCCTGGAAGACGCAGCTTCTC

CAAAGTGTATCATGAAGGAGAAGAAGAAGCCCGGTGAAACCTT

CTTCATGTGCTCCTGTTCCTCAGATGAATGCAACGATAACATC

ATCTTCTCCGAGGAGTACAACACCTCCAACCCAGATATCCCTC

CACACGTTCAGAAGTCCGTAAACAATGACATGATTGTGACCGA

CAACAACGGGGCTGTTAAGTTCCCACAGCTCTGTAAG 1 1 1 1 GC Nucleic acid

GACGTTAGGTTCAGCACCTGTGATAATCAGAAGAGCTGCATGT seauence encodina

CCAACTGCAGCATCACCAGTA 1 1 1 GCGAGAAGCCTCAAGAAGT T22d35-Fc in

GTGTGTCGCTG 1 1 1 GGAGAAAGAACGACGAAAACATAACCCTG secretable form

GAGACCGTTTGCCACGATCCAAAACTCCCATATCACGATTTCA

TTCTGGAGGACGCCGCCAGTCCTAAATGTATAATGAAAGAGAA

GAAGAAACCAGGGGAGACCTTC 1 1 1 ATGTGCAGCTGCAGCAG

CGACGAGTGTAACGATAATATAATTTTTAGCGAGGAGTATAATA

CAAGCAATCCCGACGAGCGCAAGTGCTGCGTCGAGTGCCCTC

CATGCCCTGCCCCTCCTGTTGCCGGACCTAGTGTGTTTTTGTT

TCCTCCTAAACCTAAAGATACACTCATGATTAGCAGGACACCT

GAGGTGACATGTGTCGTCGTGGACGTGAGTCATGAAGACCCC

GAAGTGCAG 1 1 1 AATTGGTATGTCGACGGAGTCGAAGTCCATA

ATGCCAAAACTAAACCAAGGGAAGAACAGTTTAATTCAACTTTT

CGCGTGGTCTCTGTGCTGACTGTGGTGCACCAGGACTGGCTT

AATGGAAAGGAATACAAGTGTAAGGTGAGTAATAAGGGCCTGC CCGCCCCCATTGAAAAAACTATTAGTAAGACTAAAGGGCAGCC

CCGAGAGCCCCAGGTGTATAC I I I GCCCCCCTCTCGGGAGGA

GATGACTAAAAATCAGGTGAGTCTTACATGTCTTGTGAAAGGA

TTTTACCCCTCTGACATTTCAGTGGAGTGGGAGTCTAATGGCC

AGCCCGAGAATAATTACAAAACTACTCCCCCCATGTTGGACTC

TGACGGCTCATTTTTCTTGTACTCTAAACTGACAGTGGACAAAA

GTCGGTGGCAGCAGGGCAATGTG I I I I CTTGTTCAGTGATGCA

CGAGGCCCTGCATAATCACTATACACAGAAATCTCTGTCTCTG

TCACCCGGCTGATGA

ATGGACTGGACCTGGAGAATCCTGTTCCTGGTGGCTGCTGCT

ACCGGAACACACGCTATCCCCCCTCATGTGCAGAAGTCCGTG

AACAATGACATGATCGTGACAGATAACAATGGCGCCGTGAAGT

TTCCTCAGCTGTGCAAGTTCTGTGACGTGAGG I I I AGCACCTG

CGATAACCAGAAGTCCTGCATGAGCAATTGTTCTATCACATCC

ATCTGCGAGAAGCCACAGGAGGTGTGCGTGGCCGTGTGGCG

GAAGAACGACGAGAATATCACCCTGGAGACAGTGTGCCACGA

TCCTAAGCTGCCATACCATGACTTCATCCTGGAGGATGCTGCC

TCTCCCAAGTGTATCATGAAGGAGAAGAAGAAGCCTGGCGAG

ACATTCTTCATGTGCTCCTGTTCCAGCGACGAGTGCAACGATA

ATATCATCTTCAGCGAGGAGTATAACACCTCTAATCCAGATATC

CCACCCCACGTGCAGAAGTCTGTCAATAACGATATGATTGTCA

CAGATAACAATGGCGCTGTGAAG I I I CCCCAGCTGTGCAAATT

TTGTGACGTGAGA I I I I CCACCTGTGATAACCAGAAGAGCTGC

ATGTCTAATTGTTCCATCACATCTA I I I GTGAAAAACCTCAGGA

AGTGTGCGTGGCCGTGTG G AG AAAAAATG ATG AAAAC ATC AC

CCTGGAGACAGTGTGCCATGATCCCAAGCTGCCTTATCACGA Nucleic acid

CTTCATCCTGGAAGACGCTGCCAGCCCAAAATGCATTATGAAA sequence encodina

GAGAAGAAGAAGCCCGGTGAGACATTCTTCATGTGCAGCTGTT T22d35-Fc-laG1-

CTTCTGATGAATGTAACGATAATATCATC I I I I CCGAGGAGTAT v1 (CO in

AACACAAGCAATCCCGACACCCACACATGCCCTCCATGTCCAG secretable form

CTCCTGAGCTGCTGGGAGGACCTAGCGTGTTCCTG I I I CCCC

CTAAGCCAAAGGATACCCTGATGATCAGCAGGACCCCCGAGG

TGACATGCGTGGTGGTGGACGTGTCTCACGAGGACCCCGAGG

TGAAG I I I AACTGGTACGTGGACGGCGTGGAGGTGCATAATG

CCAAGACCAAGCCTAGGGAGGAGCAGTACAACTCTACCTATC

GGGTGGTGTCCGTGCTGACAGTGCTGCATCAGGATTGGCTGA

ACGGCAAGGAGTATAAGTGCAAGGTGTCCAATAAGGCTCTGC

CAGCCCCCATTGAGAAGACCATCAGCAAGGCTAAGGGCCAGC

CAAGAGAGCCCCAGGTGTACACACTGCCACCCTCTCGCGACG

AGCTGACCAAGAACCAGGTGTCCCTGACATGTCTGGTGAAGG

GCTTCTATCCTTCCGATATCGCTGTGGAGTGGGAGAGCAACG

GACAGCCAGAGAACAATTACAAGACCACACCTCCAGTGCTGG

ACTCTGATGGCTCCTTC I I I CTGTATAGCAAGCTGACCGTGGA

CAAGTCTAGGTGGCAGCAGGGCAACGTG I I I AGCTGTTCTGT

GATGCATGAGGCCCTGCACAATCATTACACACAGAAGTCCCTG

AGCCTGTCTCCTGGC

ATGGACTGGACCTGGAGAATCCTGTTCCTGGTGGCTGCTGCT

ACCGGAACACACGCTATCCCCCCTCATGTGCAGAAGTCTGTG

AACAATGACATGATCGTGACAGATAACAATGGCGCCGTGAAGT Nucleic acid seauence

TTCCCCAGCTGTGCAAGTTCTGTGACGTGAGG I I I I CCACCTG encodina

CGATAACCAGAAGTCTTGCATGTCCAATTGTAGCATCACATCT T22d35-Fc-laG1- v2(SCa in

ATCTGCGAGAAGCCTCAGGAGGTGTGCGTGGCCGTGTGGCG secretable form

GAAGAACGACGAGAATATCACCCTGGAGACAGTGTGCCACGA

TCCTAAGCTGCCATACCATGACTTCATCCTGGAGGATGCTGCC AGCCCAAAGTGTATCATGAAGGAGAAGAAGAAGCCCGGCGAG

ACATTCTTCATGTGCTCTTGTTCCAGCGACGAGTGCAACGATA ATATCATCTTCTCCGAGGAGTATAACACCAGCAATCCTGACAT CCCACCCCACGTGCAGAAGAGCGTCAATAACGATATGATTGTC ACAGATAACAATGGCGCTGTGAAG 1 1 1 CCACAGCTGTGCAAAT

TTTGTGACGTGAGATTTTCTACCTGTGATAACCAGAAGTCCTG CATGAGCAATTGTTCTATCACATCCATCTGCGAGAAGCCACAG GAAGTGTGCGTGGCCGTGTGGAGAAAAAATGATGAAAACATC ACCCTGGAGACAGTGTGCCATGATCCCAAGCTGCCTTATCAC GACTTCATCCTGGAAGACGCTGCCTCCCCTAAATGCATTATGA AAG AG AAG AAG AAG CC AG GTG AG AC ATTCTTC ATGTG C AG CT GTTCTTCTGATGAGTGCAACGATAACATCATC 1 1 1 1 CTGAGGA

GTACAACACATCCAATCCTGACGTGGAGCCAAAGAGCTCTGAT AAGACCCACACATGCCCTCCATGTCCAGCTCCTGAGCTGCTG GGAGGACCATCCGTGTTCCTG 1 1 1 CCACCTAAGCCTAAGGACA

CCCTGATGATCTCCAGGACCCCAGAGGTGACATGCGTGGTGG TGGACGTGAGCCACGAGGACCCCGAGGTGAAG 1 1 1 AACTGGT

ACGTGGATGGCGTGGAGGTGCATAATGCCAAGACCAAGCCAA GGGAGGAGCAGTACAACAGCACCTATCGGGTGGTGTCTGTGC TGACAGTGCTGCATCAGGACTGGCTGAACGGCAAGGAGTATA AGTGCAAGGTGTCTAATAAGGCTCTGCCAGCCCCCATCGAGA AGACCATCTCCAAGGCTAAGGGCCAGCCAAGAGAGCCCCAGG TGTACACACTGCCACCCAGCCGCGACGAGCTGACCAAGAACC AGGTGTCTCTGACATGTCTGGTGAAGGGCTTCTATCCCTCTGA TATCGCTGTGGAGTGGGAGTCCAACGGACAGCCTGAGAACAA TTACAAGACCACACCTCCAGTGCTGGACAGCGATGGCTC I M C

1 1 1 CTGTATTCCAAGCTGACCGTGGATAAGAGCAGGTGGCAGC

AGGGCAACGTG 1 1 1 1 CCTGTAGCGTGATGCATGAGGCCCTGC

ACAATCATTACACACAGAAGTCTCTGTCCCTGAGCCCTGGC

ATGGATTGGACCTGGAGAATCCTGTTCCTGGTGGCTGCTGCTA

CCGGAACACACGCTATCCCCCCTCATGTGCAGAAGTCTGTGA

ACAATGACATGATCGTGACAGATAACAATGGCGCCGTGAAGTT

TCCTCAGCTGTGCAAGTTCTGTGACGTGAGG 1 1 1 1 CCACCTGC

GATAACCAGAAGTCCTGCATGAGCAATTGTTCTATCACATCCA

TCTGCGAGAAGCCACAGGAGGTGTGCGTGGCCGTGTGGCGG

AAGAACGACGAGAATATCACCCTGGAGACAGTGTGCCACGAT

CCTAAGCTGCCATACCATGACTTCATCCTGGAGGATGCTGCCA

GCCCCAAGTGTATCATGAAGGAGAAGAAGAAGCCTGGCGAGA

CATTCTTCATGTGCTCTTGTTCCAGCGACGAGTGCAACGATAA

TATCATCTTCTCCGAGGAGTATAACACCAGCAATCCAGACATC Nucleic acid

CCACCCCACGTGCAGAAGAGCGTCAATAACGATATGATTGTCA seauence encodina

CAGATAACAATGGCGCTGTGAAG 1 1 1 CCCCAGCTGTGCAAATT T22d35-Fc-laG1-

TTGTGACGTGAGA 1 1 1 1 CTACCTGTGATAACCAGAAGAGCTGC v3(GSL-CC) in

ATGTCTAATTGTTCCATCACATCTA 1 1 1 GTGAAAAACCTCAGGA secretable form

AGTGTGCGTGGCCGTGTG G AG AAAAAATG ATG AAAAC ATC AC

CCTGGAGACAGTGTGCCATGATCCCAAGCTGCCTTATCACGA

CTTCATCCTGGAAGACGCTGCCTCCCCAAAATGCATTATGAAA

GAGAAGAAGAAGCCCGGTGAGACATTCTTCATGTGCAGCTGTT

CTTCTGATGAGTGCAACGATAACATCATC 1 1 1 1 CTGAGGAGTA

CAACACATCCAATCCTGACGGAGGAGGCAGCGGAGGAGGCTC

TGGAGGCGGCACCCACACATGCCCTCCATGTCCAGCTCCTGA

GCTGCTGGGAGGACCTTCCGTGTTCCTG 1 1 1 CCCCCTAAGCC

AAAGGACACCCTGATGATCTCCAGGACCCCCGAGGTGACATG

CGTGGTGGTGGACGTGAGCCACGAGGACCCCGAGGTGAAGT TTAACTGGTACGTGGATGGCGTGGAGGTGCATAATGCCAAGA

CCAAGCCAAGGGAGGAGCAGTACAACAGCACCTATCGGGTGG

TGTCTGTGCTGACAGTGCTGCATCAGGATTGGCTGAACGGCA

AGGAGTATAAGTGCAAGGTGTCTAATAAGGCTCTGCCAGCCC

CCATTGAGAAGACCATCTCCAAGGCTAAGGGCCAGCCAAGAG

AGCCCCAGGTGTACACACTGCCACCCAGCCGCGACGAGCTGA

CCAAGAACCAGGTGTCTCTGACATGTCTGGTGAAGGGCTTCTA

TCCTTCTGATATCGCTGTGGAGTGGGAGTCCAACGGACAGCC

AGAGAACAATTACAAGACCACACCTCCAGTGCTGGACTCTGAT

GGCTCCTTCTTTCTGTATTCCAAGCTGACCGTGGACAAGAGCA

GGTGGCAGCAGGGCAACGTGTTTAGCTGTTCTGTGATGCATG

AGGCCCTGCACAATCATTACACACAGAAGTCCCTGAGCCTGTC

TCCTGGC

ATGGATTGGACCTGGAGAATCCTCTTCCTTGTAGCAGCAGCAA

CAGGTACACATGCTATCCCTCCTCATGTTCAAAAGTCCGTTAA

CAACGACATGATCGTCACCGATAACAACGGTGCTGTCAAGTTC

CCACAACTCTGTAAGTTCTGCGATGTGCGTTTCTCCACATGTG

ATAACCAGAAGTCCTGTATGAGCAACTGCTCAATCACCTCCAT

CTGCGAAAAGCCACAAGAGGTATGCGTAGCTGTATGGCGAAA

GAACGATGAAAACATCACCCTGGAAACCGTCTGTCACGATCCA

AAGCTCCCATACCATGATTTCATCCTGGAAGACGCAGCTTCTC

CAAAGTGTATCATGAAGGAGAAGAAGAAGCCCGGTGAAACCTT

CTTCATGTGCTCCTGTTCCTCAGATGAATGCAACGATAACATC

ATCTTCTCCGAGGAGTACAACACCTCCAACCCAGATATCCCTC

CACACGTTCAGAAGTCCGTAAACAATGACATGATTGTGACCGA

CAACAACGGGGCTGTTAAGTTCCCACAGCTCTGTAAGTTTTGC

GACGTTAGGTTCAGCACCTGTGATAATCAGAAGAGCTGCATGT

CCAACTGCAGCATCACCAGTATTTGCGAGAAGCCTCAAGAAGT

GTGTGTCGCTGTTTGGAGAAAGAACGACGAAAACATAACCCTG

GAGACCGTTTGCCACGATCCAAAACTCCCATATCACGATTTCA Nucleic acid sequence

TTCTGGAGGACGCCGCCAGTCCTAAATGTATAATGAAAGAGAA encoding

GAAGAAACCAGGGGAGACCTTCTTTATGTGCAGCTGCAGCAG T22d35-Fc-lgG2- v2(CC) in

CGACGAGTGTAACGATAATATAATTTTTAGCGAGGAGTATAATA secretable form

CAAGCAATCCCGACGTCGAGTGCCCTCCATGCCCTGCCCCTC

CTGTTGCCGGACCTAGTGTGTTTTTGTTTCCTCCTAAACCTAAA

GATACACTCATGATTAGCAGGACACCTGAGGTGACATGTGTCG

TCGTGGACGTGAGTCATGAAGACCCCGAAGTGCAGTTTAATTG

GTATGTCGACGGAGTCGAAGTCCATAATGCCAAAACTAAACCA

AGGGAAGAACAGTTTAATTCAACTTTTCGCGTGGTCTCTGTGC

TGACTGTGGTGCACCAGGACTGGCTTAATGGAAAGGAATACAA

GTGTAAGGTGAGTAATAAGGGCCTGCCCGCCCCCATTGAAAA

AACTATTAGTAAGACTAAAGGGCAGCCCCGAGAGCCCCAGGT

GTATACTTTGCCCCCCTCTCGGGAGGAGATGACTAAAAATCAG

GTGAGTCTTACATGTCTTGTGAAAGGATTTTACCCCTCTGACAT

TTCAGTGGAGTGGGAGTCTAATGGCCAGCCCGAGAATAATTAC

AAAACTACTCCCCCCATGTTGGACTCTGACGGCTCATTTTTCTT

GTACTCTAAACTGACAGTGGACAAAAGTCGGTGGCAGCAGGG

CAATGTGTTTTCTTGTTCAGTGATGCACGAGGCCCTGCATAAT

CACTATACACAGAAATCTCTGTCTCTGTCACCCGGCTGATGA REFERENCES

All patents, patent applications and publications referred to throughout the application are listed below.

Arteaga CL (2006) Inhibition of TGFpeta signaling in cancer therapy. Curr Opin Genet Dev 16: 30-37

De Crescenzo G, Grothe S, Zwaagstra J, Tsang M, O'Connor-McCourt MD (2001 ) Real-time monitoring of the interactions of transforming growth factor-beta (TGF-beta ) isoforms with latency-associated protein and the ectodomains of the TGF-beta type II and III receptors reveals different kinetic models and stoichiometries of binding. J Biol Chem 276: 29632-29643

Durocher Y, Perret S, Kamen A (2002) High-level and high-throughput recombinant protein production by transient transfection of suspension-growing human 293-EBNA1 cells. Nucleic Acids Res 30: E9

Economides AN, Carpenter LR, Rudge JS, Wong V, Koehler-Stec EM, Hartnett C, Pyles EA, Xu X, Daly TJ, Young MR, Fandl JP, Lee F, Carver S, McNay J, Bailey K, Ramakanth S, Hutabarat R, Huang TT, Radziejewski C, Yancopoulos GD, Stahl N (2003) Cytokine traps: multi-component, high-affinity blockers of cytokine action. Nat Med 9: 47-52

Eisenberg D, Schwarz E, Komaromy M, Wall R (1984) Analysis of membrane and surface protein sequences with the hydrophobic moment plot. J Mol Biol 179: 125-142

Gajewski TF (2015) The Next Hurdle in Cancer Immunotherapy: Overcoming the Non-T-Cell- Inflamed Tumor Microenvironment. Semin Oncol 42: 663-671

Garberg P, Ball M, Borg N, Cecchelli R, Fenart L, Hurst RD, Lindmark T, Mabondzo A, Nilsson JE, Raub TJ, Stanimirovic D, Terasaki T, Oberg JO, Osterberg T (2005) In vitro models for the blood-brain barrier. Toxicol In Vitro 19: 299-334

Hahn T, Akporiaye ET (2006) Targeting transforming growth factor beta to enhance cancer immunotherapy. Curr Oncol 13: 141 -143

Haqqani AS, Caram-Salas N, Ding W, Brunette E, Delaney CE, Baumann E, Boileau E, Stanimirovic D (2013) Multiplexed evaluation of serum and CSF pharmacokinetics of brain- targeting single-domain antibodies using a NanoLC-SRM-ILIS method. Mol Pharm 10: 1542- 1556

Hawinkels LJ, Ten Dijke P (201 1) Exploring anti-TGF-beta therapies in cancer and fibrosis. Growth Factors 29 : 140- 152 Holash J, Davis S, Papadopoulos N, Croll SD, Ho L, Russell M, Boland P, Leidich R, Hylton D, Burova E, loffe E, Huang T, Radziejewski C, Bailey K, Fandl JP, Daly T, Wiegand SJ, Yancopoulos GD, Rudge JS (2002) VEGF-Trap: a VEGF blocker with potent antitumor effects. Proc Natl Acad Sci U S A 99: 1 1393-1 1398

Jin P, Zhang J, Beryt M, Turin L, Brdlik C, Feng Y, Bai X, Liu J, Jorgensen B, Shepard HM (2009) Rational optimization of a bispecific ligand trap targeting EGF receptor family ligands. Mol Med 15: 1 1 -20

Li MO, Wan YY, Sanjabi S, Robertson AK, Flavell RA (2006) Transforming growth factor-beta regulation of immune responses. Annu Rev Immunol 24: 99-146

Massague J, Blain SW, Lo RS (2000) TGFpeta signaling in growth control, cancer, and heritable disorders. Ce// 103: 295-309

Mourskaia AA, Northey JJ, Siegel PM (2007) Targeting aberrant TGF-beta signaling in preclinical models of cancer. Anticancer Agents Med Chem 7: 504-514

Rodgarkia-Dara C, Vejda S, Erlach N, Losert A, Bursch W, Berger W, Schulte-Hermann R, Grusch M (2006) The activin axis in liver biology and disease. Mutat Res 613: 123-137

Santarpia M, Gonzalez-Cao M, Viteri S, Karachaliou N, Altavilla G, Rosell R (2015)

Programmed cell death protein-1 /programmed cell death ligand-1 pathway inhibition and predictive biomarkers: understanding transforming growth factor-beta role. TransI Lung Cancer Res 4: 728-742

Thiery JP, Acloque H, Huang RY, Nieto MA (2009) Epithelial-mesenchymal transitions in development and disease. Ce// 139: 871 -890

Wojtowicz-Praga S (2003) Reversal of tumor-induced immunosuppression by TGF-beta inhibitors. Invest New Drugs 21 : 21 -32

Yang L, Pang Y, Moses HL (2010) TGF-beta and immune cells: an important regulatory axis in the tumor microenvironment and progression. Trends Immunol 31 : 220-227

Yang X, Ambrogelly A (2014) Enlarging the repertoire of therapeutic monoclonal antibodies platforms: domesticating half molecule exchange to produce stable lgG4 and lgG1 bispecific antibodies. Curr Opin Biotechnol 30: 225-229 Zheng X, Koropatnick J, Chen D, Velenosi T, Ling H, Zhang X, Jiang N, Navarro B, lchim TE, Urquhart B, Min W (2013) Silencing IDO in dendritic cells: a novel approach to enhance cancer immunotherapy in a murine breast cancer model. I 'nt J Cancer 132: 967-977 Zwaagstra JC, Sulea T, Baardsnes J, Lenferink AE, Collins C, Cantin C, Paul-Roc B, Grothe S, Hossain S, Richer LP, L'Abbe D, Tom R, Cass B, Durocher Y, O'Connor-McCourt MD (2012) Engineering and therapeutic application of single-chain bivalent TGF-beta family traps. Mol Cancer Ther 11 : 1477-1487

WO/1995/04069

WO/2004/076670

WO 2008/1 13185

WO 2010/031 168

US8815247

US62777375

US2015/0225483

WO01/83525;

WO2005/028517;

WO2008/1 13185;

WO2008/157367;

WO2010/0031 18;

WO2010/099219;

WO2012/071649;

WO2012/142515;

WO2013/000234;

US5693607;

US2005/0203022;

US2007/0244042;

US8318135;

US8658135;

US8815247;

US2015/0225483; and

US2015/0056199