Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
DOSING REGIMENS FOR TARGETED TGF-B INHIBITION FOR USE IN TREATING CANCER IN TREATMENT NAIVE SUBJECTS
Document Type and Number:
WIPO Patent Application WO/2019/222252
Kind Code:
A1
Abstract:
This disclosure relates to dosage regimens for targeted TGF-β inhibition with a bi-functional fusion protein for use in a method of treating cancer or inhibiting tumor growth in treatment naive patients.

Inventors:
OJALVO LAUREEN (US)
EL BAWAB SAMER (DE)
DUSSAULT ISABELLE (US)
VUGMEYSTER YULIA (US)
KHANDELWAL AKASH (DE)
CHRISTENSEN OLAF (US)
Application Number:
PCT/US2019/032271
Publication Date:
November 21, 2019
Filing Date:
May 14, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
MERCK PATENT GMBH (DE)
OJALVO LAUREEN (US)
International Classes:
A61K38/17; A61K39/00; A61K39/395
Foreign References:
US20180002436A12018-01-04
US20160009807A12016-01-14
Other References:
See also references of EP 3813868A4
Attorney, Agent or Firm:
ASHRAF, Shovon et al. (US)
Download PDF:
Claims:
CLAIMS

WHAT IS CLAIMED IS: 1. A method of treating advanced non-small cell lung cancer (NSCLC) or inhibiting tumor growth in a treatment naïve patient in need thereof, the method comprising administering to the patient a dose of at least 500 mg of a protein comprising a first polypeptide and a second polypeptide, wherein the first polypeptide comprises: (a) at least a variable region of a heavy chain of an antibody that binds to human protein Programmed Death Ligand 1 (PD-L1); and (b) human Transforming Growth Factor Receptor II (TGF RII), or a fragment thereof, capable of binding Transforming Growth Factor (TGF ), wherein the second polypeptide comprises at least a variable region of a light chain of an antibody that binds PD-L1, and wherein the heavy chain of the first polypeptide and the light chain of the second polypeptide, when combined, form an antigen binding site that binds PD-L1. 2. The method of claim 1, wherein the first polypeptide comprises the amino acid sequence of SEQ ID NO: 3, and the second polypeptide comprises the amino acid sequence of SEQ ID NO: 1. 3. The method of claim 1 or 2, wherein the dose is 500 mg to 2400 mg. 4. The method of any one of claims 1-3, wherein the dose is 1200 mg to 3000 mg. 5. The method of any one of claims 1-4, wherein the dose is 1200 mg. 6. The method of any one of claims 1-4, wherein the dose is 2400 mg. 7. The method of any one of claims 1-4, wherein the dose is administered once every two weeks or once every three weeks. . The method of claim 7, wherein the dose is 1200 mg, administered once every two weeks.

9. The method of claim 7, wherein the dose is 2100 mg, administered once every three weeks. 10. The method of claim 7, wherein the dose is 2400 mg, administered once every three weeks. 11. The method of any one of claims 1-10, wherein the advanced NSCLC exhibits squamous or non-squamous histology. 12. The method of any one of claims 1-11, wherein the cancer exhibits high PD-L1 expression. 13. The method of any one of claims 1-12, wherein the patient does not have a mutation selected from the group consisting of EGFR sensitizing mutation, ALK translocation, a ROS1 mutation, and BRAF V600E mutation. 14. The method of any one of claims 1-13, wherein the treatment results in a disease response or improved survival of the patient. 15. The method of claim 14, wherein the disease response is a complete response, a partial response, or a stable disease. 16. The method of claim 14, wherein the survival is progression-free survival (PFS). 17. The method of any one of claims 1-16, wherein the protein is administered by intravenous administration. 18. The method of claim 17, wherein the intravenous administration is performed with a prefilled bag, a prefilled pen, or a prefilled syringe comprising a formulation comprising the protein. 19. The method of claim 18, wherein the bag is connected to a channel comprising a tube and/or a needle. 20. An intravenous drug delivery formulation for use in a method of treating advanced non-small cell lung cancer (NSCLC) or inhibiting tumor growth in a treatment naïve cancer patient in need thereof, the formulation comprising 500 mg– 3000 mg of a protein comprising a first polypeptide and a second polypeptide; wherein the first polypeptide comprises: (a) at least a variable region of a heavy chain of an antibody that binds to human protein Programmed Death Ligand 1 (PD-L1); and (b) human Transforming Growth Factor Receptor II (TGF RII), or a fragment thereof, capable of binding Transforming Growth Factor (TGF ), wherein the second polypeptide comprises at least a variable region of a light chain of an antibody that binds PD-L1, and wherein the heavy chain of the first polypeptide and the light chain of the second polypeptide, when combined, form an antigen binding site that binds PD-L1. 21. The intravenous drug delivery formulation for use of claim 20, wherein the first polypeptide comprises the amino acid sequence of SEQ ID NO: 3, and the second polypeptide comprises the amino acid sequence of SEQ ID NO: 1. 22. The intravenous drug delivery formulation for use of claim 20 or 21 comprising 1200 mg to 2400 mg of the protein. 23. The intravenous drug delivery formulation for use of claim 20 or 21 comprising 1200 mg of the protein. 24. The intravenous drug delivery formulation for use of claim 20 or 21 comprising 2400 mg of the protein. 25. The intravenous drug delivery formulation for use of any one of claims 20-22, wherein the formulation is administered to the patient once every two weeks or once every three weeks. 26. The intravenous drug delivery formulation for use of claim 25, wherein a formulation comprising 1200 mg of the protein is administered once every two weeks. 27. The intravenous drug delivery formulation for use of claim 25, wherein a formulation comprising 2400 mg of the protein is administered once every three weeks. 28. The intravenous drug delivery formulation for use of any one of claims 20-27, wherein the formulation is contained in a bag, a pen, or a syringe.

29. The intravenous drug delivery formulation for use of claim 28, wherein the bag is connected to a channel comprising a tube and/or a needle. 30. The intravenous drug delivery formulation for use of any one of claims 20-29, wherein the formulation is a lyophilized formulation or a liquid formulation. 31. A drug delivery device for use in a method of treating advanced non-small cell lung cancer (NSCLC) or inhibiting tumor growth in a treatment naïve cancer patient in need thereof, the device comprising a formulation comprising 500 mg– 3000 mg of a protein comprising a first polypeptide and a second polypeptide; wherein the first polypeptide comprises: (a) at least a variable region of a heavy chain of an antibody that binds to human protein Programmed Death Ligand 1 (PD-L1); and (b) human Transforming Growth Factor Receptor II (TGF RII), or a fragment thereof, capable of binding Transforming Growth Factor (TGF ), wherein the second polypeptide comprises at least a variable region of a light chain of an antibody that binds PD-L1, and wherein the heavy chain of the first polypeptide and the light chain of the second polypeptide, when combined, form an antigen binding site that binds PD-L1. 32. The drug delivery device for use of claim 31, wherein the first polypeptide comprises the amino acid sequence of SEQ ID NO: 3, and the second polypeptide comprises the amino acid sequence of SEQ ID NO: 1. 33. The drug delivery device for use of claim 31 or 32 comprising 1200 mg to 2400 mg of the protein. 34. The drug delivery device for use of claim 31 or 32 comprising 1200 mg of the protein. 35. The drug delivery device for use of claim 31 or 32 comprising 2400 mg of the protein. 36. The drug delivery device for use of any one of claims 31-33, wherein the formulation is administered to the patient once every two weeks or once every three weeks.

37. The drug delivery device for use of claim 36, wherein a formulation comprising 1200 mg of the protein is administered once every two weeks. 38. The drug delivery device for use of claim 36, wherein a formulation comprising 2400 mg of the protein is administered once every three weeks. 39. The drug delivery device for use of any one of claims 31-38, wherein the device is a bag, a pen, or a syringe. 40. The drug delivery device for use of claim 39, wherein the bag is connected to a channel comprising a tube and/or a needle. 41. The intravenous drug delivery formulation for use of any one of claims 20-30, or the drug delivery device for use of any one of claims 31-40, wherein the advanced NSCLC exhibits squamous or non-squamous histology. 42. The intravenous drug delivery formulation for use of claim 41, or the drug delivery device for use of claim 41, wherein the cancer exhibits high PD-L1 expression. 43. The intravenous drug delivery formulation for use of any one of claims 41-42, or the drug delivery device for use of any one of claims 41-42, wherein the patient does not have a mutation selected from the group consisting of EGFR sensitizing mutation, ALK translocation, ROS1 mutation, and BRAF V600E mutation. 44. The intravenous drug delivery formulation for use of any one of claims 41-43, or the drug delivery device for use of any one of claims 41-43, wherein the treatment results in a disease response or improved survival of the patient. 45. The intravenous drug delivery formulation for use of claim 44, or the drug delivery device for use of claim 44, wherein the disease response is a complete response, a partial response, or a stable disease. 46. The intravenous drug delivery formulation for use of claim 44, or the drug delivery device for use of claim 44, wherein the survival is progression-free survival (PFS). 47. An anti-PD-L1/TGF Trap protein comprising a first polypeptide and a second polypeptide for use in a method of treating advanced non-small cell lung cancer (NSCLC) or inhibiting tumor growth in a treatment naïve cancer patient in need thereof, the method comprising administering 500 mg– 3000 mg of the protein to the patient; wherein the first polypeptide comprises: (a) at least a variable region of a heavy chain of an antibody that binds to human protein Programmed Death Ligand 1 (PD-L1); and (b) human Transforming Growth Factor Receptor II (TGF RII), or a fragment thereof, capable of binding Transforming Growth Factor (TGF ), wherein the second polypeptide comprises at least a variable region of a light chain of an antibody that binds PD-L1, and wherein the heavy chain of the first polypeptide and the light chain of the second polypeptide, when combined, form an antigen binding site that binds PD-L1. 48. The anti-PD-L1/TGF Trap protein for use of claim 47, wherein the first polypeptide comprises the amino acid sequence of SEQ ID NO: 3, and the second polypeptide comprises the amino acid sequence of SEQ ID NO: 1. 49. The anti-PD-L1/TGF Trap protein for use of claim 47 or 48, wherein 1200 mg to 2400 mg of the protein is administered to the pateint. 50. The anti-PD-L1/TGF Trap protein for use of claim 47 or 48, wherein 1200 mg of the protein is administered to the pateint. 51. The anti-PD-L1/TGF Trap protein for use of claim 47 or 48, wherein 2400 mg of the protein is administered to the pateint. 52. The anti-PD-L1/TGF Trap protein for use of any one of claims 47-49, wherein the protein is administered to the patient once every two weeks or once every three weeks. 53. The anti-PD-L1/TGF Trap protein for use of claim 50, wherein 1200 mg of the protein is administered once every two weeks. 54. The anti-PD-L1/TGF Trap protein for use of claim 51, wherein 2400 mg of the protein is administered once every three weeks. 55. The anti-PD-L1/TGF Trap protein for use of any one of claims 47-54, wherein the protein is contained in a bag, a pen, or a syringe.

56. The anti-PD-L1/TGF Trap protein for use of claim 55, wherein the bag is connected to a channel comprising a tube and/or a needle. 57. The anti-PD-L1/TGF Trap protein for use of any one of claims 47-56, wherein the protein is comprised in a lyophilized formulation or a liquid formulation.

Description:
DOSING REGIMENS FOR TARGETED TGF- INHIBITION FOR USE IN TREATING CANCER IN TREATMENT NAÏVE SUBJECTS CROSS-REFERENCE TO RELATED APPLICATIONS

[0001] This application claims the benefit of and priority to U.S. Provisional Patent Application No.62/671,963, filed May 15, 2018; and to U.S. Provisional Patent Application No.62/804,931, filed February 13, 2019, the entire disclosures of which are incorporated by reference herein. SEQUENCE LISTING

[0002] The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on May 3, 2019, is named EMD-007WO_SL_ST25.txt and is 75,847 bytes in size. FIELD OF THE DISCLOSURE

[0003] The present disclosure relates generally to dosage regimens for targeted TGF- inhibition with a bi-functional fusion protein for use in a method of treating cancer or inhibiting tumor growth in treatment naïve patients. BACKGROUND

[0004] The programmed death 1 (PD-1)/PD-L1 axis is an important mechanism for tumor immune evasion. Effector T cells chronically sensing antigen take on an exhausted phenotype marked by PD-1 expression, a state under which tumor cells engage by upregulating PD-L1. Additionally, in the tumor microenvironment, myeloid cells, macrophages, parenchymal cells and T cells upregulate PD-L1. Blocking the axis restores the effector function in these T cells. [0005] US patent application publication number US 20150225483 A1, incorporated herein by reference, describes a bi-functional fusion protein that combines an anti-programmed death ligand 1 (PD-L1) antibody with the soluble extracellular domain of tumor growth factor beta receptor type II (TGF RII) as a TGF neutralizing“Trap,” into a single molecule. Specifically, the protein is a heterotetramer, consisting of the two immunoglobulin light chains of anti-PD- L1, and two heavy chains comprising the heavy chain of anti-PD-L1 genetically fused via a flexible glycine-serine linker to the extracellular domain of the human TGF RII (see Fig.1). This anti-PD-L1/ TGF Trap molecule is designed to target two major mechanisms of immunosuppression in the tumor microenvironment. US patent application publication number US 20150225483 A1 describes administration of the Trap molecule at doses based on the patient’s weight. [0006] Lung cancer is the leading cause of cancer death in the USA and results in more cancer deaths than breast cancer, prostate cancer, and colorectal cancer combined. Non-small cell lung cancer accounts for approximately 80% of all cases of lung cancer. It is estimated in 2018 there would be 234,030 new cases of lung and bronchus cancer and 154,050 people would die from their lung cancers in the USA alone (Siegel, 2018). In the EU, 275,700 deaths due to lung cancer were predicted in 2017 (Malvezzi, 2017). Worldwide, an estimated 1.8 million new cases of lung cancer were diagnosed in 2012, approximately 13% of the total of all new cancers diagnosed (Ferlay, 2012). Immune checkpoint inhibitors have shown improved treatment outcome in patients with non-small cell lung cancer (NSCLC); however, there is room to further improve benefits. SUMMARY OF THE DISCLOSURE

[0007] The present disclosure provides improved dosing regimens for administration of bifunctional proteins targeting PD-L1 and TGF . Specifically, body weight independent (BW- independent) dosing regimens and related dosage forms involving administration of at least 500 mg (e.g., 1200 mg, 2400 mg) of the bifunctional protein administered at various dosing frequencies can be used as an anti-tumor and anti-cancer therapeutic. The BW-independent dosing regimen ensures that all patients, irrespective of their body weight, will have adequate drug exposure at the tumor site. [0008] The bifunctional protein of the present disclosure (anti-PD-L1/TGF Trap molecule) includes a first and a second polypeptide. The first polypeptide includes: (a) at least a variable region of a heavy chain of an antibody that binds to human protein Programmed Death Ligand 1 (PD-L1); and (b) human Transforming Growth Factor Receptor II (TGF RII), or a fragment thereof, capable of binding Transforming Growth Factor (TGF ) (e.g., a soluble fragment). The second polypeptide includes at least a variable region of a light chain of an antibody that binds PD-L1, in which the heavy chain of the first polypeptide and the light chain of the second polypeptide, when combined, form an antigen binding site that binds PD-L1 (e.g., any of the antibodies or antibody fragments described herein). Because the bifunctional protein of the present disclosure binds to two targets, (1) PD-L1, which is largely membrane bound, and (2) TGF , which is soluble in blood and interstitium, the BW-independent dosing regimen requires a dose that is effective not only to inhibit PD-L1 at the tumor site but also sufficient to inhibit TGF . [0009] In one aspect, the disclosure provides a method of treating cancer or inhibition of a tumor, e.g., non-small cell lung cancer (e.g., advanced non-small cell lung cancer), melanoma, pancreatic cancer, colorectal cancer (e.g., pretreated colorectal cancer (CRC)), ovarian cancer, glioblastoma, gastric cancer (e.g., pretreated recurrent or refractory unresectable Stage IV gastric cancer), biliary tract cancer, esophageal cancer (squamous cell carcinoma or adenocarcinoma), adenoma of the head or the neck, and squamous carcinoma of the head or the neck in a treatment naïve patient. In one aspect, the present invention provides a method of treating NSCLC with high PD-L1 expression, by administering an anti-PD-L1/TGF Trap molecule described in the present disclosure to a patient in need thereof. In certain

embodiments, the present invention provides a method of treating advanced non-small cell lung cancer (NSCLC) (e.g., NSCLC with high PD-L1 expression) or inhibiting tumor growth in a treatment naïve patient in need thereof by administering 1200 mg of an anti-PD-L1/TGF Trap molecule of the present disclosure once every two weeks to the patient. In certain other embodiments, the present invention provides a method of treating advanced non-small cell lung cancer (NSCLC) (e.g., NSCLC with high PD-L1 expression) or inhibiting tumor growth in a treatment naïve patient in need thereof by administering 2400 mg of an anti-PD-L1/TGF Trap molecule of the present disclosure once every three weeks to the patient. [0010] The disclosure also features a method of promoting local depletion of TGF . The method includes administering a protein described above, where the protein binds TGF in solution, binds PD-L1 on a cell surface, and carries the bound TGF into the cell (e.g., a cancer cell). [0011] The disclosure also features a method of inhibiting SMAD3 phosphorylation in a cell (e.g., a cancer cell or an immune cell), the method including exposing the cell in the tumor microenvironment to a protein described above. [0012] Other embodiments and details of the disclosure are presented herein below. BRIEF DESCRIPTION OF THE DRAWINGS

[0013] FIG.1 is a schematic drawing of an anti-PD-L1/TGF Trap molecule including one anti-PD-L1 antibody fused to two extracellular domain (ECD) of TGF Receptor II via a (Gly 4 Ser) 4 Gly (SEQ ID NO: 11) linker. [0014] FIG.2 shows a graph of a two-step ELISA demonstrating that anti-PD-L1/TGF Trap simultaneously binds to both PD-L1 and TGF . [0015] FIG.3 is a graph showing anti-PD-L1/TGF Trap induces a dramatic increase in IL- 2 levels. [0016] FIG.4A is a graph showing in vivo depletion of TGF 1 in response to the anti-PD- L1/TGF Trap. Line graphs represent naïve, isotype control, and three different doses, as indicated in the legend. FIG.4B is a graph showing in vivo depletion of TGF 2 in response to the anti-PD-L1/TGF Trap. Line graphs represent naïve, isotype control, and three different doses, as indicated in the legend. FIG. 4C is a graph showing in vivo depletion of TGF 3 in response to the anti-PD-L1/TGF Trap. Line graphs represent naïve, isotype control, and three different doses, as indicated in the legend. FIG. 4D is a graph showing that occupancy of PD- L1 by the anti-PD-L1/TGF Trap supports a receptor binding model in the EMT-6 tumor system. [0017] FIG.5 is a graph showing anti-tumor efficacy of anti-PD-L1/TGF Trap control (anti-PD-L1(mut)/TGF ) in Detroit 562 xenograft model. [0018] FIG.6A is a box-plot of C avg distribution for an entire population for a fixed (1200 mg) versus mg/kg based dosing (17.65 mg/kg) in a simulated population of 68 kg median body weight. FIG.6B is a box-plot of exposure AUC distribution for an entire population for a fixed (1200 mg) versus mg/kg based dosing (17.65 mg/kg) in a simulated population of 68 kg median body weight. FIG. 6C is a box-plot of C trough distribution for an entire population for a fixed (1200 mg) versus mg/kg based dosing (17.65 mg/kg) in a simulated population of 68 kg median body weight. FIG. 6D is a box-plot of C max distribution for an entire population for a fixed (1200 mg) versus mg/kg based dosing (17.65 mg/kg) in a simulated population of 68 kg median body weight. [0019] FIG.6E is a box-plot of C avg distribution for an entire population for a fixed (500 mg) versus mg/kg based dosing (7.35 mg/kg) in a simulated population of 68 kg median body weight. FIG.6F is a box-plot of exposure AUC distribution for an entire population for a fixed (500 mg) versus mg/kg based dosing (7.35 mg/kg) in a simulated population of 68 kg median body weight. FIG. 6G is a box-plot of C trough distribution for an entire population for a fixed (500 mg) versus mg/kg based dosing (7.35 mg/kg) in a simulated population of 68 kg median body weight. FIG. 6H is a box-plot of C max distribution for an entire population for a fixed (500 mg) versus mg/kg based dosing (7.35 mg/kg) in a simulated population of 68 kg median body weight. [0020] FIGs.7A–7C are graphs showing the predicted PK and PD-L1 receptor occupancy (“RO”) of ant-PD-L1/TGF Trap molecules at doses and schedules associated with tumor stasis in mice. FIG.7A is a graph showing the predicted plasma concentration vs. time. FIG. 7B is a graph showing the predicted PD-L1 RO vs. time in PBMC. FIG.7C is a graph showing the predicted PD-L1 RO vs. time in tumor. [0021] FIG.8 is a schematic diagram of the therapeutic regimen. Abbreviations used in the figure: NSCLC = non-small cell lung cancer; Q2W= every 2 weeks; Q3W= every 3 weeks; BOR = best overall response; DOR = duration of response; OS = overall survival; and R* = randomization. PD-L1 high represents 80% PD-L1 positive tumor cells as determined by the PD-L1 IHC 73-10 assay (Dako), or TPS 50% as determined by the Dako IHC 22C3

PharmDx assay; both assays select a similar patient population at their respective cutoffs. DETAILED DESCRIPTION [0022] By“TGF RII” or“TGF Receptor II” is meant a polypeptide having the wild-type human TGF Receptor Type 2 Isoform A sequence (e.g., the amino acid sequence of NCBI Reference Sequence (RefSeq) Accession No. NP_001020018 (SEQ ID NO: 8)), or a polypeptide having the wild-type human TGF Receptor Type 2 Isoform B sequence (e.g., the amino acid sequence of NCBI RefSeq Accession No. NP_003233 (SEQ ID NO: 9)) or having a sequence substantially identical to the amino acid sequence of SEQ ID NO: 8 or of SEQ ID NO: 9. The TGF RII may retain at least 0.1%, 0.5%, 1%, 5%, 10%, 25%, 35%, 50%, 75%, 90%, 95%, or 99% of the TGF -binding activity of the wild-type sequence. The polypeptide of expressed TGF RII lacks the signal sequence. [0023] By a“fragment of TGF RII capable of binding TGF” is meant any portion of NCBI RefSeq Accession No. NP_001020018 (SEQ ID NO: 8) or of NCBI RefSeq Accession No. NP_003233 (SEQ ID NO: 9), or a sequence substantially identical to SEQ ID NO: 8 or SEQ ID NO: 9 that is at least 20 (e.g., at least 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 175, or 200) amino acids in length that retains at least some of the TGF -binding activity (e.g., at least 0.1%, 0.5%, 1%, 5%, 10%, 25%, 35%, 50%, 75%, 90%, 95%, or 99%) of the wild-type receptor or of the corresponding wild-type fragment. Typically, such fragment is a soluble fragment. An exemplary such fragment is a TGF RII extra-cellular domain having the sequence of SEQ ID NO: 10. Certain other exemplary fragments of human TGF RII capable of binding TGF are represented by the sequence of SEQ ID NOs: 50, 51, 52, 53, or 54. [0024] “Treatment naïve” refers to patients who have not received prior systemic treatment for their advanced/Stage IV NSCLC. In various embodiments of the present disclosure, treatment naïve patients have not received prior therapy with an anti-PD-1, anti-PD-L1, anti- PD-L2, anti-CD137, or anti-Cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) antibody (including ipilimumab), or any other antibody or drug specifically targeting T-cell co- stimulation or checkpoint pathways. In various embodiments of the present disclosure, treatment naïve patients are selected for first-line (1L) treatment of the present invention. [0025] “PD-L1 positive” or“PD-L1+” indicates 1% PD-L1 positive tumor cells as determined, for example, by the Dako IHC 22C3 PharmDx assay, or by the VENTANA PD-L1 (SP263) assay. [0026] “PD-L1 high” or“high PD-L1” refers to 80% PD-L1 positive tumor cells as determined by the PD-L1 IHC 73-10 assay (Dako), or tumor proportion score (TPS) 50% as determined by the Dako IHC 22C3 PharmDx assay (TPS is a term of art related to the IHC 22C3 PharmDx assay, which describes the percentage of viable tumor cells with partial or complete membrane staining (e.g., staining for PD-L1)). Both IHC 73-10 and IHC 22C3 assays select a similar patient population at their respective cutoffs. In certain embodiments,

VENTANA PD-L1 (SP263) assay, which has high concordance with 22C3 PharmDx assay (see Sughayer et al., Appl. Immunohistochem. Mol. Morphol., (2018)), can also be used for determining PD-L1 high expression level. [0027] By“substantially identical” is meant a polypeptide exhibiting at least 50%, desirably 60%, 70%, 75%, or 80%, more desirably 85%, 90%, or 95%, and most desirably 99% amino acid sequence identity to a reference amino acid sequence. The length of comparison sequences will generally be at least 10 amino acids, desirably at least 15 contiguous amino acids, more desirably at least 20, 25, 50, 75, 90, 100, 150, 200, 250, 300, or 350 contiguous amino acids, and most desirably the full-length amino acid sequence. [0028] By“patient” is meant either a human or non-human animal (e.g., a mammal).

“Patient,”“subject,”“patient in need thereof,” and“subject in need thereof” are used interchangeably in this disclosure, and refer to a living organism suffering from or prone to a disease or condition that can be treated by administration using the methods and compositions provided in this disclosure. [0029] The terms“treat,”“treating,” or“treatment,” and other grammatical equivalents as used in this disclosure, include alleviating, abating, ameliorating, or preventing a disease, condition or symptoms, preventing additional symptoms, ameliorating or preventing the underlying metabolic causes of symptoms, inhibiting the disease or condition, e.g., arresting the development of the disease or condition, relieving the disease or condition, causing regression of the disease or condition, relieving a condition caused by the disease or condition, or stopping the symptoms of the disease or condition, and are intended to include prophylaxis. The terms further include achieving a therapeutic benefit and/or a prophylactic benefit. By therapeutic benefit is meant eradication or amelioration of the underlying disorder being treated. Also, a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the patient, notwithstanding that the patient may still be afflicted with the underlying disorder. [0030] The term“cancer” is meant advanced/Stage IV non-small cell lung cancer (NSCLC). Advanced/Stage IV NSCLC is used according to its plain and ordinary meaning, and refers to stages IVA or IVB of NSCLC, characterized by, for example, metastasis to one or more sites. Thus, in various embodiments, the cancer is a metastatic NSCLC. [0031] Throughout the description and claims of this specification the word“comprise” and other forms of the word, such as“comprising” and“comprises,” means including but not limited to, and is not intended to exclude, for example, other components. [0032] By“co-administer” it is meant that a composition described herein is administered at the same time, just prior to, or just after the administration of additional therapies. The protein and the composition of the present disclosure can be administered alone or can be co- administered with a second, third, or fourth therapeutic agent(s) to a patient. Co-administration is meant to include simultaneous or sequential administration of the protein or composition individually or in combination (more than one therapeutic agent). [0033] The term“a” is not meant to limit as a singular. In certain embodiments, the term “a” may refer to a plural form. As used throughout this disclosure, the singular forms“a,”“an,” and“the” include plural reference unless the context clearly dictates otherwise. Thus, for example, a reference to“a composition” includes a plurality of such compositions, as well as a single composition. [0034] A“reconstituted” formulation is one which has been prepared by dissolving a lyophilized formulation in an aqueous carrier such that the bifunctional molecule is dissolved in the reconstituted formulation. The reconstituted formulation is suitable for intravenous administration (IV) to a patient in need thereof. [0035] The term“about” refers to any minimal alteration in the concentration or amount of an agent that does not change the efficacy of the agent in preparation of a formulation and in treatment of a disease or disorder. In embodiments, the term“about” may include ±15% of a specified numerical value or data point. [0036] Ranges can be expressed in this disclosure as from“about” one particular value, and/or to“about” another particular value. When such a range is expressed, another aspect includes from the one particular value and/or to the other particular value. Similarly, when values are expressed as approximations, by use of the antecedent“about,” it is understood that the particular value forms another aspect. It is further understood that the endpoints of each of the ranges are significant both in relation to the other endpoint, and independently of the other endpoint. It is also understood that there are a number of values disclosed in this disclosure, and that each value is also disclosed as“about” that particular value in addition to the value itself. It is also understood that throughout the application, data are provided in a number of different formats and that this data represent endpoints and starting points and ranges for any combination of the data points. For example, if a particular data point“10” and a particular data point“15” are disclosed, it is understood that greater than, greater than or equal to, less than, less than or equal to, and equal to 10 and 15 are considered disclosed as well as between 10 and 15. It is also understood that each unit between two particular units are also disclosed. For example, if 10 and 15 are disclosed, then 11, 12, 13, and 14 are also disclosed. [0037] An“isotonic” formulation is one which has essentially the same osmotic pressure as human blood. Isotonic formulations will generally have an osmotic pressure from about 250 to 350 mOsmol/KgH 2 O. The term "hypertonic" is used to describe a formulation with an osmotic pressure above that of human blood. Isotonicity can be measured using a vapor pressure or ice- freezing type osmometer, for example. [0038] The term“buffering agent” refers to one or more components that when added to an aqueous solution is able to protect the solution against variations in pH when adding acid or alkali, or upon dilution with a solvent. In addition to phosphate buffers, there can be used glycinate, carbonate, citrate buffers and the like, in which case, sodium, potassium or ammonium ions can serve as counterion. [0039] An“acid” is a substance that yields hydrogen ions in aqueous solution. A “pharmaceutically acceptable acid” includes inorganic and organic acids which are nontoxic at the concentration and manner in which they are formulated. [0040] A“base” is a substance that yields hydroxyl ions in aqueous solution.

“Pharmaceutically acceptable bases” include inorganic and organic bases which are non-toxic at the concentration and manner in which they are formulated. [0041] A“lyoprotectant” is a molecule which, when combined with a protein of interest, prevents or reduces chemical and/or physical instability of the protein upon lyophilization and subsequent storage. [0042] A“preservative” is an agent that reduces bacterial action and may be optionally added to the formulations herein. The addition of a preservative may, for example, facilitate the production of a multi-use (multiple-dose) formulation. Examples of potential preservatives include octadecyldimethylbenzyl ammonium chloride, hexamethonium chloride, benzalkonium chloride (a mixture of alkylbenzyldimethylammonium chlorides in which the alkyl groups are long-chain compounds), and benzethonium chloride. Other types of preservatives include aromatic alcohols such as phenol, butyl and benzyl alcohol, alkyl parabens such as methyl or propyl paraben, catechol, resorcinol, cyclohexanol, 3pentanol, and m-cresol. [0043] A“surfactant” is a surface active molecule containing both a hydrophobic portion (e.g., alkyl chain) and a hydrophilic portion (e.g., carboxyl and carboxylate groups). Surfactant may be added to the formulations of the invention. Surfactants suitable for use in the formulations of the present invention include, but are not limited to, polysorbates (e.g.

polysorbates 20 or 80); poloxamers (e.g. poloxamer 188); sorbitan esters and derivatives; Triton; sodium laurel sulfate; sodium octyl glycoside; lauryl-, myristyl-, linoleyl-, or stearyl- sulfobetadine; lauryl-, myristyl-, linoleyl- or stearyl-sarcosine; linoleyl-, myristyl-, or cetyl- betaine; lauramidopropyl-cocamidopropyl-, linoleamidopropyl-, myristamidopropyl-, palmidopropyl-, or isostearamidopropylbetaine (e.g., lauroamidopropyl); myristamidopropyl-, palmidopropyl-, or isostearamidopropyl-dimethylamine; sodium methyl cocoyl-, or disodium methyl oleyl-taurate; and the MONAQUAT TM series (Mona Industries, Inc., Paterson, N.J.), polyethylene glycol, polypropyl glycol, and copolymers of ethylene and propylene glycol (e.g., Pluronics, PF68 etc.). Body Weight-Independent Dosing Regimen [0044] Body weight-independent dosing regimens involving the administration to treatment naïve patients of at least 500 mg of the bifunctional anti-PD-L1/TGF Trap molecules described herein have been developed, informed by the results of a variety of pre-clinical and clinical assessments of the molecules. Two studies investigated the safety, tolerability, and pharmacokinetics of the molecules, and included assessments of PD-L1 target occupancy on peripheral blood mononuclear cells obtained from the blood of treated patients and measurements of the concentrations of TGF 1, TGF 2, and TGF 3. These assessments were based on data from a total of 350 subjects (dose escalation cohorts of 1, 3, 10 and 20 mg/kg in solid tumors, and expansion cohorts of 3 mg/kg, 10 mg/kg, 500 mg, and 1200 mg in selected tumor types). PK/Efficacy Model (Mouse Model) [0045] Experiments were also conducted to determine the efficacy of the anti-PD-L1/TGF Trap molecule in a tumor model. Efficacy results from EMT-6 xenografts were used to establish the PK/Efficacy model. The established PK model in mice was used to simulate anti- PD-L1/TGF Trap plasma exposure for the efficacy experiment settings. The estimated parameters are reported in Table 1. The estimated KC50 value was 55.3 µg/mL. This value represents the average plasma concentrations for which 50% of the maximal anti-tumor activity of the anti-PD-L1/TGF Trap molecule could be achieved. [0046] Basic diagnostics plots of the model revealed no model misspecification. The model predictions are able to capture the tumor volume distributions. Conditional weighted residuals are normally distributed with a 0 mean and 1 variance without a trend. The PK/Efficacy model was then used to simulate tumor growth inhibition (TGI) using the human predicted concentration-time profiles at different doses. [0047] Table 1: Mouse PK/Efficacy model parameters for anti-PD-L1/TGF Trap molecule in EMT-6 xenograft mice

Response Analysis Based on PD-L1 Occupancy (in a Mouse Model) [0048] Using the efficacy experiments, responses in mice have been analyzed and sorted by either tumor regression or tumor stasis, and PK and PD-L1 receptor occupancy (RO) have been predicted based on the integrated PK/RO model. The approach demonstrated that an anti-PD- L1/TGF Trap molecule plasma concentration between 40 and 100 µg/mL associated with a PD-L1 RO above 95% in tumor is required to reach tumor regression. The plasma

concentration of anti-PD-L1/TGF Trap molecule between 10 and 40 µg/mL associated with a PD-L1 RO above 95% in periphery is required to reach tumor stasis. [0049] Response analysis and predicted PK/RO in mice lead to FIGs. 7A-7C, which summarize the PK/RO/Efficacy for the anti-PD-L1/TGF Trap molecule in mice.95% of PD- L1 RO is achieved at a plasma concentration of 40 µg/mL with an expected/estimate TGI of only about 65%. Increasing the concentration above 40 µg/mL results in an additional increase in tumor growth inhibition. 95% of tumor growth inhibition is achieved at average plasma concentration of about 100 µg/mL. [0050] Based on the population PK model described below, a flat dose of at least 500 mg administered once every two weeks is required to maintain an average concentration of about 100 µg/mL, while a flat dose of about 1200 mg administered once every two weeks is required to maintain a C trough of about 100 µg/mL. In certain embodiments about 1200 mg to about 3000 mg (e.g., about 1200, about 1300, about 1400, about 1500, about 1600, about 1700, about 1800, about 1900, about 2000, about 2100, about 2200, about 2300, about 2400, etc.) of a protein product of the present disclosure (e.g., anti-PD-L1/TGF Trap) is administered to a subject. In certain embodiments, about 1200 mg of anti-PD-L1/TGF Trap molecule is administered to a subject once every two weeks. In certain embodiments, about 2400 mg of anti-PD-L1/TGF Trap molecule is administered to a subject once every three weeks. [0051] In embodiments, about 1200 mg to about 3000 mg (e.g., about 1200 mg, about 1300 mg, about 1400 mg, about 1500 mg, about 1600 mg, about 1700 mg, about 1800 mg, about 1900 mg, about 2000 mg, about 2100 mg, about 2200 mg, about 2300 mg, about 2400 mg, etc.) of the protein product with a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3, and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1 is administered to a subject. In certain embodiments, about 1200 mg to about 3000 mg (e.g., about 1200 mg, about 1300 mg, about 1400 mg, about 1500 mg, about 1600 mg, about 1700 mg, about 1800 mg, about 1900 mg, about 2000 mg, about 2100 mg, about 2200 mg, about 2300 mg, about 2400 mg, etc.) of the protein product with a first polypeptide that includes a first polypeptide comprising the amino acid sequences of SEQ ID NOs: 35, 36, and 37, and a second polypeptide comprising the amino acid sequences of SEQ ID NOs: 38, 39, and 40 is administered to a subject. [0052] In certain embodiments, about 1200 mg of the protein product with a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3, and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1 is administered to a subject once every two weeks. In certain embodiments, about 2400 mg of the protein product with a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3, and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1 is administered to a subject once every three weeks. In certain embodiments, about 1200 mg of the protein product that includes a first polypeptide comprising the amino acid sequences of SEQ ID NOs: 35, 36, and 37, and a second polypeptide comprising the amino acid sequences of SEQ ID NOs: 38, 39, and 40 is administered to a subject once every two weeks. In certain embodiments, about 2400 mg of the protein product that includes a first polypeptide comprising the amino acid sequences of SEQ ID NOs: 35, 36, and 37, and a second polypeptide comprising the amino acid sequences of SEQ ID NOs: 38, 39, and 40 is administered to a subject once every three weeks. Establishing Body Weight-Independent Dosing Regimen [0053] Informed by the clinical and pre-clinical data, a new, body weight-independent dosing regimen for the administration of anti-PD-L1/TGF Trap molecules has been created to achieve less variability in exposure, reduce dosing errors, reduce the time necessary for dose preparation, and reduce drug wastage compared to the mg/kg dosing, thus facilitating favorable treatment outcomes. According to one embodiment, a flat dose of at least 500 mg can be administered, regardless of the patient’s body weight. According to another embodiment, a flat dose of at least 1200 mg can be administered, regardless of the patient’s body weight.

According to another embodiment, a flat dose of 2400 mg can be administered, regardless of the patient’s body weight. Typically, such doses would be administered repeatedly, such as once every two weeks or once every 3 weeks, for example. For example, a flat dose of 1200 mg can be administered once every two weeks, or a flat dose of 2400 mg can be administered once every three weeks. Pharmacokinetic (PK) Analysis Sampling in Humans [0054] An example of pharmacokinetic analysis to determine the optimal flat dose of the anti-PD-L1/TGF Trap is provided by the experiments described below. [0055] Serum samples for pharmacokinetic (PK) data analysis were collected before the start of the first dose and at the following time points after the first dose: on Day 1 immediately after the infusion and 4 hours after the start of the infusion; on Day 2 at least 24 hours after the Day 1 end of infusion; and on Days 8 and 15. At selected subsequent dosing occasions pre- dose, end-of-infusion and 2 to 8 hours after the end of infusion samples were collected on days 15, 29, 43. For later time points on days 57, 71 and 85, pre-dose samples were or were to be collected followed by once every 6 weeks PK sampling until 12 weeks, then once every 12 weeks PK sampling. In the expansion phase sparse PK sampling was conducted. [0056] The PK data described above were used to produce a population PK model and to perform simulations of possible dosing regimens. Modeling method, known as the full approach model, described in Gastonguay, M., Full Covariate Models as an Alternative to Methods Relying on Statistical Significance for Inferences about Covariate Effects: A Review of Methodology and 42 Case Studies, (2011) p. 20, Abstract 2229, was applied to the population model data obtained from the simulations to obtain parameters having the following features: 2-compartment PK model with linear elimination, IIV on CL, V1, and V2, combined additive and proportional residual error, full covariate model on CL and V1. The following baseline covariates were included in the final model: age, weight, sex, race, albumin, CRP, platelet count, eGFR, hepatic impairment, ECOG score, tumor size, tumor type, and previous treatment with biologics. The following estimates of typical parameter estimates of pharmacokinetics of the protein of the present disclosure (e.g., anti-PD-L1/TGF Trap) were obtained: clearance (CL) 0.0177 L/h (6.2%), central volume of distribution (V1) 3.64 L (8.81%), peripheral volume of distribution (V2) 0.513 L (25.1%), and intercompartmental clearance (Q) 0.00219 L/h (17.8%). The inter-patient variability was 22% for CL, 20% for V1, and 135% for V2. Body weight was a relevant covariate on both CL and V1. To support the flat dosing approach, the impact of the dosing strategy on the exposure variability of the protein of the present disclosure (e.g., anti-PD-L1/TGF Trap) was explored. Specifically, simulations were performed to compare the exposure distribution using a flat dosing approach of 1200 mg once every two weeks versus a BW-adjusted dosing approach of either 17.65 mg/kg once every two weeks (corresponding to 1200 mg once every two weeks for a 68 kg subject or 15 mg/kg once every two weeks (corresponding to 1200 mg for a 80 kg subject). Further simulations were performed to compare the exposure distribution using a flat dosing approach of 500 mg once every two weeks versus a BW-adjusted dosing approach of 7.35 mg/kg once every two weeks (corresponding to 500 mg once every two weeks for a 68 kg subject). In addition, simulations were performed to assess the following flat doses at once every three weeks: 1200 mg, 1400, mg, 1600 mg, 1800 mg, 2000 mg, 2200 mg, 2400 mg, 2600 mg, 2800 mg, 3000 mg. [0057] The following methodology for simualtions was used: N=200 sets of parameter estimates were drawn from multivariate normal distribution of parameter estimates, using the final PK model variance-covariance matrix. For each parameter estimate, 200 IIV estimates were drawn from $OMEGA multivariate normal distribution, resulting in total 40000 (200 x 200) subjects. The original dataset (N=380) was resampled with replacement to generate 40000 sets of matched covariates and steady-state exposure metrics (AUC, C avg , C trough and C max ) were generated for each dosing regimen. [0058] Simulations showed that across a wide BW spectrum, variability in exposure is slightly higher for BW-based dosing in comparison with fixed dosing. An example of exposure distribution at 17.65 mg/kg and 1200 mg flat dose, or 7.35 mg/kg and 500 mg flat dose for a median body weight of 68 kg is shown in FIGs.6A and 6E, respectively. Simulations also showed the opposite trend in exposure distributions across weight quartiles across the patient population: low-weight patients have higher exposure with fixed dosing, whereas high-weight patients have higher exposure with BW-adjusted dosing. Establishing Efficacious Dose/Dosing Regimen in Humans: preliminary dose-response in 2 nd Line Non Small Cell Lung Cancer (2L NSCLC) following once every 2 weeks (q2w) dosing of anti-PD-L1/TGF Trap [0059] An example of the therapeutic efficacy of the anti-PD-L1/TGF Trap is established by the clinical study described below. [0060] Patients with advanced NSCLC unselected for PD-L1 who progressed following 1 st line standard treatment (no prior immunotherapy) were randomized to receive the anti-PD- L1/TGF Trap of the present disclosure at 500 mg or 1200 mg (n=40 per cohort) once every two weeks (q2w), until disease progression, unacceptable toxicity, or trial withdrawal. The primary objective was to assess best overall response (BOR) per Response Evaluation Criteria in Solid Tumors version 1.1 (RECIST v1.1). Other objectives included dose exploration and safety/tolerability assessment. Tumor cell PD-L1 expression levels (Ab clone 73-10 (Dako) [>80% = >50% with 22C3]) were characterized as PD-L1 <1%, 1% (PD-L1+), or 80% (PD- L1–high). Tumor cell PD-L1 expression was evaluable in 75 patients. [0061] As of data cutoff at the time of analysis, 80 patients received anti-PD-L1/TGF Trap for a median of 11.9 weeks (range, 2-66.1), with a median follow-up of 51.1 weeks. Ten patients remain on treatment. Investigator-assessed confirmed overall response rate (ORR) was 23.8% (500 mg ORR, 20.0%; 1200 mg ORR, 27.5%), with 18 partial responses (PR) seen across both dose levels, and 1 complete response (CR) seen at 1200 mg. As shown in Table 2, clinical activity was observed across PD-L1 expression levels: ORR was 37.0% in PD-L1+ and 85.7% in PD-L1–high patients at 1200 mg. The most common treatment-related adverse events (TRAEs) were pruritus (20.0%), maculopapular rash (18.8%), and decreased appetite (12.5%). Grade 3 TRAEs occurred in 23 patients (28.8%), anad Grade 4 TRAEs occurred in 2 patients. Eight patients (500 mg, n=2; 1200 mg, n=6) discontinued treatment due to TRAEs. No treatment-related deaths occurred. [0062] Table 2: Observed response rate in 2L NSCLC patients treated with either 500 mg or 1200 mg of anti-PD-L1/TGF Trap once every 2 weeks

[0063] These results demonstrate that anti-PD-L1/TGF Trap monotherapy was well tolerated and showed efficacy across PD-L1 subgroups, with an ORR at 1200 mg of 37.0% and 85.7% in PD-L1+ and PD-L1–high patients, respectively. Given the response rates significantly improved at higher PD-L1 tumor cell expression (e.g., patients treated at 1200 mg), this promising activity of anti-PD-L1/TGF Trap observed as a 2L treatment is expected to translate or increase as a first line (1L) therapy in treatment naïve PD-L1–high advanced NSCLC patients. Establishing Dosing Regimen with Various Dosing Frequencies [0064] Data regimens with various dosing frequencies have been created to allow less frequent administration and/or to allow coordination of dosing schedules with concomicant medications. Specifically, the preliminary population PK modeling and simulation

methodology described above has been used to simulate exposures for various dosing regimens and to compare regimens based on exposure. [0065] Based on these simulations, a flat dose of at least 500 mg administered once every two weeks is required to maintain an average concentration of about 100 µg/mL for a typical subject, while a flat dose of about 1200 mg administered once every two weeks is required to maintain a C trough of about 100 µg/mL. [0066] Based on simulations for C avg , 1200 mg once every two weeks is equivalent to 1800 mg once every three weeks, while for C trough , 1200 mg once every two weeks is equivalent to 2800 mg once every three weeks. And for C avg , 500 mg once every two weeks is equivalent to 750 mg once every three weeks; for C trough 500 mg once every two weeks is equivalent to 1,167 mg once every three weeks. TGF as a Cancer Target [0067] The current disclosure permits localized reduction in TGF in a tumor

microenvironment by capturing the TGF using a soluble cytokine receptor (TGF RII) tethered to an antibody moiety targeting a cellular immune checkpoint receptor found on the exterior surface of certain tumor cells or immune cells. An example of an antibody moiety of the disclosure to an immune checkpoint protein is anti-PD-L1. This bifunctional molecule, sometimes referred to in this document as an“antibody-cytokine Trap,” is effective precisely because the anti-receptor antibody and cytokine Trap are physically linked. The resulting advantage (over, for example, administration of the antibody and the receptor as separate molecules) is partly because cytokines function predominantly in the local environment through autocrine and paracrine functions. The antibody moiety directs the cytokine Trap to the tumor microenvironment where it can be most effective, by neutralizing the local immunosuppressive autocrine or paracrine effects. Furthermore, in cases where the target of the antibody is internalized upon antibody binding, an effective mechanism for clearance of the cytokine/cytokine receptor complex is provided. Antibody-mediated target internalization was shown for PD-L1, and anti-PD-L1/TGF Trap was shown to have a similar internalization rate as anti-PD-L1. This is a distinct advantage over using an anti-TGF antibody because first, an anti-TGF antibody might not be completely neutralizing; and second, the antibody can act as a carrier extending the half-life of the cytokine. [0068] Indeed, as described below, treatment with the anti-PD-L1/TGF Trap elicits a synergistic anti-tumor effect due to the simultaneous blockade of the interaction between PD- L1 on tumor cells and PD-1 on immune cells, and the neutralization of TGF in the tumor microenvironment. Without being bound by theory, this presumably is due to a synergistic effect obtained from simultaneous blocking the two major immune escape mechanisms, and in addition, the depletion of the TGF in the tumor microenvironment by a single molecular entity. This depletion is achieved by (1) anti-PD-L1 targeting of tumor cells; (2) binding of the TGF autocrine/paracrine in the tumor microenvironment by the TGF Trap; and (3) destruction of the bound TGF through the PD-L1 receptor-mediated endocytosis.

Furthermore, the TGF RII fused to the C-terminus of Fc (fragment of crystallization of IgG) was several-fold more potent than the TGF RII-Fc that places the TGF RII at the N-terminus of Fc. [0069] TGF had been a somewhat questionable target in cancer immunotherapy because of its paradoxical roles as the molecular Jekyll and Hyde of cancer (Bierie et al., Nat. Rev.

Cancer, 2006; 6:506-20). Like some other cytokines, TGF activity is developmental stage and context dependent. Indeed TGF can act as either a tumor promoter or a tumor suppressor, affecting tumor initiation, progression and metastasis. The mechanisms underlying this dual role of TGF remain unclear (Yang et al., Trends Immunol.2010; 31:220–227). Although it has been postulated that Smad-dependent signaling mediates the growth inhibition of TGF signaling, while the Smad independent pathways contribute to its tumor-promoting effect, there are also data showing that the Smad-dependent pathways are involved in tumor progression (Yang et al., Cancer Res.2008; 68:9107-11). [0070] Both the TGF ligand and the receptor have been studied intensively as therapeutic targets. There are three ligand isoforms, TGF 1, 2 and 3, all of which exist as homodimers. There are also three TGF receptors (TGF R), which are called TGF R type I, II and III (López-Casillas et al., J Cell Biol.1994; 124:557-68). TGF RI is the signaling chain and cannot bind ligand. TGF RII binds the ligand TGF 1 and 3, but not TGF 2, with high affinity. The TGF RII/TGF complex recruits TGF RI to form the signaling complex (Won et al., Cancer Res. 1999; 59:1273-7). TGF RIII is a positive regulator of TGF binding to its signaling receptors and binds all 3 TGF isoforms with high affinity. On the cell surface, the TGF /TGF RIII complex binds TGF RII and then recruits TGF RI, which displaces

TGF RIII to form the signaling complex. [0071] Although the three different TGF isoforms all signal through the same receptor, they are known to have differential expression patterns and non-overlapping functions in vivo. The three different TGF- isoform knockout mice have distinct phenotypes, indicating numerous non-compensated functions (Bujak et al., Cardiovasc Res. 2007; 74:184-95). While TGF 1 null mice have hematopoiesis and vasculogenesis defects and TGF 3 null mice display pulmonary development and defective palatogenesis, TGF 2 null mice show various developmental abnormalities, the most prominent being multiple cardiac deformities (Bartram et al., Circulation. 2001; 103:2745-52; Yamagishi et al., Anat Rec. 2012; 295:257-67).

Furthermore, TGF is implicated to play a major role in the repair of myocardial damage after ischemia and reperfusion injury. In an adult heart, cardiomyocytes secrete TGF , which acts as an autocrine to maintain the spontaneous beating rate. Importantly, 70-85% of the TGF secreted by cardiomyocytes is TGF 2 (Roberts et al., J Clin Invest.1992; 90:2056-62). Despite cardiotoxicity concerns raised by treatment with TGF RI kinase inhibitors, present applicants have observed a lack of toxicity, including cardiotoxicity, for anti-PD-L1/TGF Trap in monkeys. [0072] Therapeutic approaches to neutralize TGF include using the extracellular domains of TGF receptors as soluble receptor Traps and neutralizing antibodies. Of the receptor Trap approach, soluble TGF RIII may seem the obvious choice since it binds all the three TGF ligands. However, TGF RIII, which occurs naturally as a 280-330 kD glucosaminoglycan (GAG)-glycoprotein, with extracellular domain of 762 amino acid residues, is a very complex protein for biotherapeutic development. The soluble TGF RIII devoid of GAG could be produced in insect cells and shown to be a potent TGF neutralizing agent (Vilchis-Landeros et al, Biochem J 355:215, 2001). The two separate binding domains (the endoglin-related and the uromodulin-related) of TGF RIII could be independently expressed, but they were shown to have affinities 20 to 100 times lower than that of the soluble TGF RIII, and much diminished neutralizing activity (Mendoza et al., Biochemistry. 2009; 48:11755-65). On the other hand, the extracellular domain of TGF RII is only 136 amino acid residues in length and can be produced as a glycosylated protein of 25-35 kD. The recombinant soluble TGF RII was further shown to bind TGF 1 with a K D of 200 pM, which is fairly similar to the K D of 50 pM for the full length TGF RII on cells (Lin et al., J Biol Chem.1995; 270:2747-54). Soluble TGF RII-Fc was tested as an anti-cancer agent and was shown to inhibit established murine malignant mesothelioma growth in a tumor model (Suzuki et al., Clin. Cancer Res., 2004; 10:5907-18). Because TGF RII does not bind TGF 2, and TGF RIII binds TGF 1 and 3 with lower affinity than TGF RII, a fusion protein of the endoglin domain of TGF RIII and extracellular domain of TGF RII was produced in bacteria and was shown to inhibit the signaling of TGF 1 and 2 in cell based assays more effectively than either TGF RII or RIII (Verona et al., Protein Eng Des Sel.2008; 21:463-73). [0073] Still another approach to neutralize all three isoforms of the TGF ligands is to screen for a pan-neutralizing anti-TGF antibody, or an anti-receptor antibody that blocks the receptor from binding to TGF 1, 2 and 3. GC1008, a human antibody specific for all isoforms of TGF , was in a Phase I/II study in patients with advanced malignant melanoma or renal cell carcinoma (Morris et al., J Clin Oncol 2008; 26:9028 (Meeting abstract)). Although the treatment was found to be safe and well tolerated, only limited clinical efficacy was observed, and hence it was difficult to interpret the importance of anti-TGF therapy without further characterization of the immunological effects (Flavell et al., Nat Rev Immunol.2010; 10:554- 67). There were also TGF -isoform-specific antibodies tested in the clinic. Metelimumab, an antibody specific for TGF 1 was tested in Phase 2 clinical trial as a treatment to prevent excessive post-operative scarring for glaucoma surgery; and Lerdelimumab, an antibody specific for TGF 2, was found to be safe but ineffective at improving scarring after eye surgery in a Phase 3 study (Khaw et al., Ophthalmology 2007; 114:1822–1830). Anti-TGF RII antibodies that block the receptor from binding to all the three TGF isoforms, such as the anti– human TGF RII antibody TR1 and anti-mouse TGF RII antibody MT1, have also shown some therapeutic efficacy against primary tumor growth and metastasis in mouse models (Zhong et al., Clin Cancer Res.2010; 16:1191-205). However, in a recent Phase I study of antibody TR1 (LY3022859), dose escalation beyond 25 mg (flat dose) was considered unsafe due to uncontrolled cytokine release, despite prophylactic treatment (Tolcher et al., Cancer Chemother Pharmacol 2017; 79:673-680). To date, the vast majority of the studies on TGF targeted anticancer treatment, including small molecule inhibitors of TGF signaling that often are quite toxic, are mostly in the preclinical stage and the anti-tumor efficacy obtained has been limited (Calone et al., Exp Oncol.2012; 34:9-16; Connolly et al., Int J Biol Sci.2012; 8:964-78). [0074] The antibody-TGF Trap of the disclosure is a bifunctional protein containing at least portion of a human TGF Receptor II (TGF RII) that is capable of binding TGF . In certain embodiments, the TGF Trap polypeptide is a soluble portion of the human TGF Receptor Type 2 Isoform A (SEQ ID NO: 8) that is capable of binding TGF . In certain embodiments, TGF Trap polypeptide contains at least amino acids 73-184 of SEQ ID NO: 8. In certain embodiments, the TGF Trap polypeptide contains amino acids 24-184 of SEQ ID NO: 8. In certain embodiments, the TGF Trap polypeptide is a soluble portion of the human TGF Receptor Type 2 Isoform B (SEQ ID NO: 9) that is capable of binding TGF . In certain embodiments, the TGF Trap polypeptide contains at least amino acids 48-159 of SEQ ID NO: 9. In certain embodiments, the TGF Trap polypeptide contains amino acids 24-159 of SEQ ID NO: 9. In certain embodiments, the TGF Trap polypeptide contains amino acids 24-105 of SEQ ID NO: 9. In certain exemplary embodiments, the TGF Trap polypeptide contains the sequence of SEQ ID NOs: 10, 50, 51, 52, 53, or 54. [0075] In another embodiment, the antibody-TGF Trap of the disclosure is one of the fusion proteins disclosed in WO 2018/205985. In some embodiments, the fusion protein is one of the constructs listed in Table 2 of this publication, such as construct 9 or 15 thereof. In other embodiments, the antibody having the heavy chain sequence of SEQ ID NO: 11 and the light chain sequence of SEQ ID NO: 12 of this publication [corresponding to SEQ ID NO: 61 and 62, respectively, of the present disclosure] is fused via a linking sequence (G 4 S) x G, wherein x is 4-5, to the TGF RII extracellular domain sequence of SEQ ID NO: 14 or SEQ ID NO: 15 of said publication [corresponding to SEQ ID NO: 50 and 51, respectively, of the present disclosure]. Mechanisms of Action [0076] The approach of targeting T cell inhibition checkpoints for dis-inhibition with therapeutic antibodies is an area of intense investigation (for a review, see Pardoll, Nat Rev Cancer.2012; 12:253-264). In one approach, the antibody moiety or antigen binding fragment thereof targets T cell inhibition checkpoint receptor proteins on the T cell, such as, for example: CTLA-4, PD-1, BTLA, LAG-3, TIM-3, or LAIR1. In another approach, the antibody moiety targets the counter-receptors on antigen presenting cells and tumor cells (which co-opt some of these counter-receptors for their own immune evasion), such as for example: PD-L1 (B7-H1), B7-DC, HVEM, TIM-4, B7-H3, or B7-H4. [0077] The disclosure contemplates antibody TGF Traps that target, through their antibody moiety or antigen binding fragment thereof, T cell inhibition checkpoints for dis-inhibition. To that end the applicants have tested the anti-tumor efficacy of combining a TGF Trap with antibodies targeting various T cell inhibition checkpoint receptor proteins, such as anti-PD-1, anti-PD-L1, anti-TIM-3 and anti-LAG3. [0078] The programmed death 1 (PD-1)/PD-L1 axis is an important mechanism for tumor immune evasion. Effector T cells chronically sensing antigen take on an exhausted phenotype marked by PD-1 expression, a state under which tumor cells engage by upregulating PD-L1. Additionally, in the tumor microenvironment, myeloid cells, macrophages, parenchymal cells and T cells upregulate PD-L1. Blocking the axis restores the effector function in these T cells. Anti-PD-L1/TGF Trap also binds TGF (1, 2, and 3 isoforms), which is an inhibitory cytokine produced in the tumor microenvironment by cells including apoptotic neutrophils, myeloid- derived suppressor cells, T cells and tumor. Inhibition of TGF by soluble TGF RII reduced malignant mesothelioma in a manner that was associated with increases in CD8+ T cell anti- tumor effects. The absence of TGF 1 produced by activated CD4+ T cells and Treg cells has been shown to inhibit tumor growth, and protect mice from spontaneous cancer. Thus, TGF appears to be important for tumor immune evasion. [0079] TGF has growth inhibitory effects on normal epithelial cells, functioning as a regulator of epithelial cell homeostasis, and it acts as a tumor suppressor during early carcinogenesis. As tumors progress toward malignancy, the growth inhibitory effects of TGF on the tumor are lost via mutation in one or more TGF pathway signaling components or through oncogenic reprogramming. Upon loss of sensitivity to TGF inhibition, the tumor continues to produce high levels of TGF , which then serve to promote tumor growth. The TGF cytokine is overexpressed in various cancer types with correlation to tumor stage. Many types of cells in the tumor microenvironment produce TGF including the tumor cells themselves, immature myeloid cells, regulatory T cells, and stromal fibroblasts; these cells collectively generate a large reservoir of TGF in the extracellular matrix. TGF signaling contributes to tumor progression by promoting metastasis, stimulating angiogenesis, and suppressing innate and adaptive anti-tumor immunity. As a broadly immunosuppressive factor, TGF directly down-regulates the effector function of activated cytotoxic T cells and NK cells and potently induces the differentiation of naïve CD4+ T cells to the immunosuppressive regulatory T cells (Treg) phenotype. In addition, TGF polarizes macrophages and neutrophils to a wound-healing phenotype that is associated with production of immunosuppressive cytokines. As a therapeutic strategy, neutralization of TGF activity has the potential to control tumor growth by restoring effective anti-tumor immunity, blocking metastasis, and inhibiting angiogenesis. [0080] Combining these pathways, PD-1 or PD-L1, and TGF , is attractive as an antitumor approach. Concomitant PD-1 and TGF blockade can restore pro-inflammatory cytokines. Anti-PD-L1/TGF Trap includes, for example, an extracellular domain of the human TGF receptor TGF RII covalently joined via a glycine/serine linker to the C terminus of each heavy chain of the fully human IgG1 anti-PD-L1 antibody. Given the emerging picture for PD-1/PD- L1 class, in which responses are apparent but with room for increase in effect size, it is assumed that co-targeting a complementary immune modulation step will improve tumor response. A similar TGF-targeting agent, fresolimumab, which is a monoclonal antibody targeting TGF 1, 2 and 3, showed initial evidence of tumor response in a Phase I trial in subjects with melanoma. [0081] In certain embodiments, the present disclosure provides experiments, which demonstrated that the TGF RII portion of anti-PD-L1/TGF Trap (the Trap control“anti-PDL- 1(mut)/ TGF Trap”) elicited antitumor activity. For example, following subcutaneous implantation in a Detroit 562 human pharyngeal carcinoma model, anti-PDL-1(mut)/ TGF Trap elicited a dose-dependent reduction in tumor volume when administered at 25 µg, 76 µg, or 228 µg (FIG. 5). [0082] In certain embodiments, the present disclosure provides experiments, which demonstrated that the protein of the present disclosure simultaneously bound to both PD-L1 and TGF (FIG. 2). [0083] In certain embodiments, the present disclosure provides experiments, which demonstrated that the protein of the present disclosure (e.g. anti-PD-L1/TGF Trap) inhibited PD-L1 and TGF dependent signaling in vitro. In certain embodiments, the present disclosure provides experiments, which demonstrated that the protein of the present disclosure enhanced T cell effector function in vitro via blockade of PD-L1-mediated immune inhibition as measured by an IL-2 induction assay following superantigen stimulation (FIG.3). At approximately 100 ng/ml, the protein of the present disclosure induced a dramatic increase in IL-2 levels in vitro (FIG. 3). [0084] In certain embodiments, the present disclosure provides experiments, which demonstrated that the protein of the present disclosure (e.g. anti-PD-L1/TGF Trap) caused depletion of TGF from blood in vivo. Treatment of orthotopically implanted EMT-6 breast cancer cells in JH mice with 55 µg, or 164 µg, or 492 µg of the protein of the present disclosure resulted in efficient and specific depletion of TGF 1 (FIG.4A), TGF 2 (FIG.4B), and TGF 3 (FIG. 4C). Furthermore, the present disclosure provides experiments, which demonstrated that the protein of the present disclosure occupied the PD-L1 target, supporting the notion that that the protein of the present disclosure fit to a receptor binding model in the EMT-6 tumor system (FIG. 4D). [0085] In certain embodiments, the present disclosure provides experiments, which demonstrated that the protein of the present disclosure efficiently, specifically, and

simultaneously bound to PD-L1 and TGF , possessed potent antitumor activity in a variety of mouse models, suppressed tumor growth and metastasis, as well as extended survival and conferred long-term protective antitumor immunity. [0086] An example of determining the mechanism of action of the anti-PD-L1/TGF Trap molecule in vivo is described below: [0087] In a first-in-human phase I dose escalation study, in addition to monitoring the pharmacokinetics of the anti-PD-L1/TGF Trap molecule, the mechanism of action, particularly against the TGFb cytokines, was investigated. [0088] Patients were treated with anti-PD-L1/TGF Trap molecule intravenously administered at 5 dose levels of about 0.3, about 1, about 3, about 10, or about 20 mg/kg once every two weeks, PK analyses were performed from samples for up to day 85. PD-L1 target occupancy was measured in CD3+ PBMCs by flow cytometry from patient blood collected at pre-dose, Day 2 (D2), D15, and D43. Further, the blood levels of TGFb1-3 and pro- inflammatory cytokines were measured at these time points with an additional time point at D8 using analytically validated Luminex bead- and ECLIA-based multiplex immunoassays. In one aspect, patients can be treated with anti-PD-L1/TGF Trap molecule intravenously administered at 6 dose levels, including the ones described above, at a dose of about 30 mg/kg or about 40 mg/kg every two weeks. PK analyses of patients treated at 6 dose levels may be performed from samples for up to after the 6 th dose. PD-L1 target occupancy may also be measured in CD3+ PBMCs by flow cytometry from patient blood collected at pre-dose, Day 2 (D2), D15, D43, and up to D85. Further, the blood levels of TGFb1-3 and pro-inflammatory cytokines may be measured at these time points with an additional time point, e.g., at D8, using analytically validated Luminex bead- and ECLIA-based multiplex immunoassays. [0089] Results indicated that the anti-PD-L1/TGF Trap molecule PK exposure during the first cycle increased in an approximately dose-proportional manner between 3 to 20 mg/kg, without significant accumulation within the first 85 days of treatment. There was about 80% PD-L1 target occupancy at 3mg/kg– 20 mg/kg which was maintained throughout the dosing interval. There was further a small (1.7 fold on D2) but significant induction of IFNg at 0.3–20 mg/kg (p=0.001, n=19). Levels of TGF 1, TGF 2, and TGF 3 in blood were reduced by a minimum of 99%, 92%, and 91%, respectively, at all-time points for dose levels 1–20 mg/kg. At the lower dose of 0.3 mg/kg, TGF 1–3 levels were depleted at D2 and D8, but not at D15. Moreover, there was further a strong correlation between the drug PK levels and the TGF trapping. Thus, complete TGF 1–3 trapping was achieved at a drug dose level of 1 mg/kg or above. Anti-PD-L1 Antibodies [0090] The disclosure can include any anti-PD-L1 antibody, or antigen-binding fragment thereof, described in the art. Anti-PD-L1 antibodies are commercially available, for example, the 29E2A3 antibody (Biolegend, Cat. No. 329701). Antibodies can be monoclonal, chimeric, humanized, or human. Antibody fragments include Fab, F(ab’)2, scFv and Fv fragments, which are described in further detail below. [0091] Exemplary antibodies are described in PCT Publication WO 2013/079174. These antibodies can include a heavy chain variable region polypeptide including an HVR-H1, HVR- H2, and HVR-H3 sequence, where: (a) the HVR-H1 sequence is X 1 YX 2 MX 3 (SEQ ID NO: 21); (b) the HVR-H2 sequence is SIYPSGGX 4 TFYADX 5 VKG (SEQ ID NO: 22); (c) the HVR-H3 sequence is IKLGTVTTVX 6 Y (SEQ ID NO: 23); further where: X 1 is K, R, T, Q, G, A, W, M, I, or S; X 2 is V, R, K, L, M, or I; X 3 is H, T, N, Q, A, V, Y, W, F, or M; X 4 is F or I; X 5 is S or T; X 6 is E or D. [0092] In a one embodiment, X 1 is M, I, or S; X 2 is R, K, L, M, or I; X 3 is F or M; X 4 is F or I; X 5 is S or T; X 6 is E or D. [0093] In another embodiment X 1 is M, I, or S; X 2 is L, M, or I; X 3 is F or M; X 4 is I; X 5 is S or T; X 6 is D. [0094] In still another embodiment, X 1 is S; X 2 is I; X 3 is M; X 4 is I; X 5 is T; X 6 is D. [0095] In another aspect, the polypeptide further includes variable region heavy chain framework sequences juxtaposed between the HVRs according to the formula: (HC-FR1)- (HVR-H1)-(HC-FR2)-(HVR-H2)-(HC-FR3)-(HVR-H3)-(HC-FR4). [0096] In yet another aspect, the framework sequences are derived from human consensus framework sequences or human germline framework sequences. [0097] In a still further aspect, at least one of the framework sequences is the following: HC-FR1 is EVQLLESGGGLVQPGGSLRLSCAASGFTFS (SEQ ID NO: 24); HC-FR2 is WVRQAPGKGLEWVS (SEQ ID NO: 25); HC-FR3 is RFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR (SEQ ID NO: 26); HC-FR4 is WGQGTLVTVSS (SEQ ID NO: 27). [0098] In a still further aspect, the heavy chain polypeptide is further combined with a variable region light chain including an HVR-L1, HVR-L2, and HVR-L3, where: (a) the HVR-L1 sequence is TGTX 7 X 8 DVGX 9 YNYVS (SEQ ID NO: 28); (b) the HVR-L2 sequence is X 10 VX 11 X 12 RPS (SEQ ID NO: 29); (c) the HVR-L3 sequence is SSX13TX14X15X16X17RV (SEQ ID NO: 30); further where: X 7 is N or S; X 8 is T, R, or S; X 9 is A or G; X 10 is E or D; X 11 is I, N or S; X 12 is D, H or N; X 13 is F or Y; X 14 is N or S; X 15 is R, T or S; X 16 is G or S; X 17 is I or T. [0099] In another embodiment, X 7 is N or S; X 8 is T, R, or S; X 9 is A or G; X 10 is E or D; X 11 is N or S; X 12 is N; X 13 is F or Y; X 14 is S; X 15 is S; X 16 is G or S; X 17 is T. [0100] In still another embodiment, X 7 is S; X 8 is S; X 9 is G; X 10 is D; X 11 is S; X 12 is N; X 13 is Y; X 14 is S; X 15 is S; X 16 is S; X 17 is T. [0101] In a still further aspect, the light chain further includes variable region light chain framework sequences juxtaposed between the HVRs according to the formula: (LC- FR1MHVR-L1)-(LC-FR2)-(HVR-L2)-(LC-FR3)-(HVR-L3)-(LC-FR4). [0102] In a still further aspect, the light chain framework sequences are derived from human consensus framework sequences or human germline framework sequences. [0103] In a still further aspect, the light chain framework sequences are lambda light chain sequences. [0104] In a still further aspect, at least one of the framework sequence is the following: LC-FR1 is QSALTQPASVSGSPGQSITISC (SEQ ID NO: 31); LC-FR2 is WYQQHPGKAPKLMIY (SEQ ID NO: 32); LC-FR3 is GVSNRFSGSKSGNTASLTISGLQAEDEADYYC (SEQ ID NO: 33); LC-FR4 is FGTGTKVTVL (SEQ ID NO: 34). [0105] In another embodiment, the disclosure provides an anti-PD-L1 antibody or antigen binding fragment including a heavy chain and a light chain variable region sequence, where: (a) the heavy chain includes an HVR-H1, HVR-H2, and HVR-H3, wherein further: (i) the HVR-H1 sequence is X 1 YX 2 MX 3 (SEQ ID NO: 21); (ii) the HVR-H2 sequence is

SIYPSGGX 4 TFYADX 5 VKG (SEQ ID NO: 22); (iii) the HVR-H3 sequence is

IKLGTVTTVX 6 Y (SEQ ID NO: 23), and; (b) the light chain includes an HVR-L1, HVR-L2, and HVR-L3, wherein further: (iv) the HVR-L1 sequence is TGTX 7 X 8 DVGX 9 YNYVS (SEQ ID NO: 28); (v) the HVR-L2 sequence is X 10 VX 11 X 12 RPS (SEQ ID NO: 29); (vi) the HVR-L3 sequence is

SSX 13 TX 14 X 15 X 16 X 17 RV (SEQ ID NO: 30); wherein: X 1 is K, R, T, Q, G, A, W, M, I, or S; X 2 is V, R, K, L, M, or I; X 3 is H, T, N, Q, A, V, Y, W, F, or M; X 4 is F or I; X 5 is S or T; X 6 is E or D; X 7 is N or S; X 8 is T, R, or S; X 9 is A or G; X 10 is E or D; X 11 is I, N, or S; X 12 is D, H, or N; X 13 is F or Y; X 14 is N or S; X 15 is R, T, or S; X 16 is G or S; X 17 is I or T. [0106] In one embodiment, X 1 is M, I, or S; X 2 is R, K, L, M, or I; X 3 is F or M; X 4 is F or I; X 5 is S or T; X 6 is E or D; X 7 is N or S; X 8 is T, R, or S; X 9 is A or G; X 10 is E or D; X 11 is N or S; X 12 is N; X 13 is F or Y; X 14 is S; X 15 is S; X 16 is G or S; X 17 is T. [0107] In another embodiment, X 1 is M, I, or S; X 2 is L, M, or I; X 3 is F or M; X 4 is I; X 5 is S or T; X 6 is D; X 7 is N or S; X 8 is T, R, or S; X 9 is A or G; X 10 is E or D; X 11 is N or S; X 12 is N; X 13 is F or Y; X 14 is S; X 15 is S; X 16 is G or S; X 17 is T. [0108] In still another embodiment, X 1 is S; X 2 is I; X 3 is M; X 4 is I; X 5 is T; X 6 is D; X 7 is S; X 8 is S; X 9 is G; X 10 is D; X 11 is S; X 12 is N; X 13 is Y; X 14 is S; X 15 is S; X 16 is S; X 17 is T. [0109] In a further aspect, the heavy chain variable region includes one or more framework sequences juxtaposed between the HVRs as: (HC-FR1)-(HVR-H1)-(HC-FR2)-(HVR-H2)-(HC- FR3)-(HVR-H3)-(HC-FR4), and the light chain variable regions include one or more framework sequences juxtaposed between the HVRs as: (LC-FR1 MHVR-L1 )-(LC-FR2)- (HVR-L2)-(LC-FR3)-(HVR-L3)-(LC-FR4). [0110] In a still further aspect, the framework sequences are derived from human consensus framework sequences or human germline sequences.

[0111] In a still further aspect, one or more of the heavy chain framework sequences is the following:

HC-FR1 is EVQLLESGGGLVQPGGSLRLSCAASGFTFS (SEQ ID NO: 24);

HC-FR2 is WVRQAPGKGLEWVS (SEQ ID NO: 25);

HC-FR3 is RFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR (SEQ ID NO: 26); HC-FR4 is WGQGTLVTVSS (SEQ ID NO: 27).

[0112] In a still further aspect, the light chain framework sequences are lambda light chain sequences.

[0113] In a still further aspect, one or more of the light chain framework sequences is the following:

LC-FR1 is QSALTQPASVSGSPGQSITISC (SEQ ID NO: 31);

LC-FR2 is WYQQHPGKAPKLMIY (SEQ ID NO: 32);

LC-FR3 is GVSNRFSGSKSGNTASLTISGLQAEDEADYYC (SEQ ID NO: 33); LC-FR4 is FGTGTKVTVL (SEQ ID NO: 34).

[0114] In a still further aspect, the heavy chain variable region polypeptide, antibody, or antibody fragment further includes at least a C H 1 domain.

[0115] In a more specific aspect, the heavy chain variable region polypeptide, antibody, or antibody fragment further includes a C H 1, a C H 2, and a C H 3 domain.

[0116] In a still further aspect, the variable region light chain, antibody, or antibody fragment further includes a C L domain.

[0117] In a still further aspect, the antibody further includes a C H 1, a C H 2, a C H 3, and a C L domain. [0118] In a still further specific aspect, the antibody further includes a human or murine constant region. [0119] In a still further aspect, the human constant region is selected from the group consisting of IgG1, IgG2, IgG2, IgG3, IgG4. [0120] In a still further specific aspect, the human or murine constant region is lgG1. [0121] In yet another embodiment, the disclosure features an anti-PD-L1 antibody including a heavy chain and a light chain variable region sequence, where: (a) the heavy chain includes an HVR-H1, an HVR-H2, and an HVR-H3, having at least 80% overall sequence identity to SYIMM (SEQ ID NO: 35), SIYPSGGITFYADTVKG (SEQ ID NO: 36), and IKLGTVTTVDY (SEQ ID NO: 37), respectively, and (b) the light chain includes an HVR-L1, an HVR-L2, and an HVR-L3, having at least 80% overall sequence identity to TGTSSDVGGYNYVS (SEQ ID NO: 38), DVSNRPS (SEQ ID NO: 39), and SSYTSSSTRV (SEQ ID NO: 40), respectively. [0122] In a specific aspect, the sequence identity is 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%. [0123] In yet another embodiment, the disclosure features an anti-PD-L1 antibody including a heavy chain and a light chain variable region sequence, where: (a) the heavy chain includes an HVR-H1, an HVR-H2, and an HVR-H3, having at least 80% overall sequence identity to MYMMM (SEQ ID NO: 41), SIYPSGGITFYADSVKG (SEQ ID NO: 42), and IKLGTVTTVDY (SEQ ID NO: 37), respectively, and (b) the light chain includes an HVR-L1, an HVR-L2, and an HVR-L3, having at least 80% overall sequence identity to TGTSSDVGAYNYVS (SEQ ID NO: 43), DVSNRPS (SEQ ID NO: 39), and SSYTSSSTRV (SEQ ID NO: 40), respectively. [0124] In a specific aspect, the sequence identity is 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%. [0125] In a still further aspect, in the antibody or antibody fragment according to the disclosure, as compared to the sequences of HVR-H1, HVR-H2, and HVR-H3, at least those amino acids remain unchanged that are highlighted by underlining as follows: (a) in HVR-H1 SYIMM (SEQ ID NO: 35), (b) in HVR-H2 SIYPSGGITFYADTVKG (SEQ ID NO: 36), (c) in HVR-H3 IKLGTVTTVDY (SEQ ID NO: 37); and further where, as compared to the sequences of HVR-L1, HVR-L2, and HVR-L3 at least those amino acids remain unchanged that are highlighted by underlining as follows: (a) HVR-L1 TGTSSDVGGYNYVS (SEQ ID NO: 38) (b) HVR-L2 DVSNRPS (SEQ ID NO: 39) (c) HVR-L3 SSYTSSSTRV (SEQ ID NO: 40). [0126] In another aspect, the heavy chain variable region includes one or more framework sequences juxtaposed between the HVRs as: (HC-FR1)-(HVR-H1)-(HC-FR2)-(HVR-H2)-(HC- FR3)-(HVR-H3)-(HC-FR4), and the light chain variable regions include one or more framework sequences juxtaposed between the HVRs as: (LC-FR1)-(HVR-L1)-(LC-FR2)- (HVR-L2)-(LC-FR3)-(HVR-L3)-(LC-FR4). [0127] In yet another aspect, the framework sequences are derived from human germline sequences. [0128] In a still further aspect, one or more of the heavy chain framework sequences is the following: HC-FR1 is EVQLLESGGGLVQPGGSLRLSCAASGFTFS (SEQ ID NO: 24); HC-FR2 is WVRQAPGKGLEWVS (SEQ ID NO: 25); HC-FR3 is RFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR (SEQ ID NO: 26); HC-FR4 is WGQGTLVTVSS (SEQ ID NO: 27). [0129] In a still further aspect, the light chain framework sequences are derived from a lambda light chain sequence. [0130] In a still further aspect, one or more of the light chain framework sequences is the following: LC-FR1 is QSALTQPASVSGSPGQSITISC (SEQ ID NO: 31); LC-FR2 is WYQQHPGKAPKLMIY (SEQ ID NO: 32); LC-FR3 is GVSNRFSGSKSGNTASLTISGLQAEDEADYYC (SEQ ID NO: 33); LC-FR4 is FGTGTKVTVL (SEQ ID NO: 34). [0131] In a still further specific aspect, the antibody further includes a human or murine constant region. [0132] In a still further aspect, the human constant region is selected from the group consisting of IgG1, IgG2, IgG2, IgG3, IgG4. [0133] In certain embodiments, the disclosure features an anti-PD-L1 antibody including a heavy chain and a light chain variable region sequence, where: (a) the heavy chain sequence has at least 85% sequence identity to the heavy chain sequence: EVQLLESGGGLVQPGGSLRLSCAASGFTFSSYIMMVWRQAPGKGLEWVSSIYPSGGITF YADWKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARIKLGTVTTVDYWGQGTLV TVSS (SEQ ID NO: 44), and (b) the light chain sequence has at least 85% sequence identity to the light chain sequence: QSALTQPASVSGSPGQSITISCTGTSSDVGGYNYVSWYQQHPGKAPKLMIYDVSN RPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSYTSSSTRVFGTGTKVTVL (SEQ ID NO: 45). [0134] In various embodiments, the heavy chain sequence has at least 86% sequence identity to SEQ ID NO: 44 and the light chain sequence has at least 86% sequence identity to SEQ ID NO: 45; the heavy chain sequence has at least 87% sequence identity to SEQ ID NO: 44 and the light chain sequence has at least 87% sequence identity to SEQ ID NO: 45; the heavy chain sequence has at least 88% sequence identity to SEQ ID NO: 44 and the light chain sequence has at least 88% sequence identity to SEQ ID NO: 45; the heavy chain sequence has at least 89% sequence identity to SEQ ID NO: 44 and the light chain sequence has at least 89% sequence identity to SEQ ID NO: 45; the heavy chain sequence has at least, 90% sequence identity to SEQ ID NO: 44 and the light chain sequence has at least 90% sequence identity to SEQ ID NO:45; the heavy chain sequence has at least 91% sequence identity to SEQ ID NO: 44 and the light chain sequence has at least 91% sequence identity to SEQ ID NO: 45; the heavy chain sequence has at least 92% sequence identity to SEQ ID NO: 44 and the light chain sequence has at least 92% sequence identity to SEQ ID NO: 45; the heavy chain sequence has at least 93% sequence identity to SEQ ID NO: 44 and the light chain sequence has at least 93% sequence identity to SEQ ID NO: 45; the heavy chain sequence has at least 94% sequence identity to SEQ ID NO: 44 and the light chain sequence has at least 94% sequence identity to SEQ ID NO: 45; the heavy chain sequence has at least 95% sequence identity to SEQ ID NO: 44 and the light chain sequence has at least 95% sequence identity to SEQ ID NO: 45; the heavy chain sequence has at least 96% sequence identity to SEQ ID NO: 44 and the light chain sequence has at least 96% sequence identity to SEQ ID NO: 45; the heavy chain sequence has at least 97% sequence identity to SEQ ID NO: 44 and the light chain sequence has at least 97% sequence identity to SEQ ID NO: 45; the heavy chain sequence has at least 98% sequence identity to SEQ ID NO: 44 and the light chain sequence has at least 98% sequence identity to SEQ ID NO: 45; the heavy chain sequence has at least 99% sequence identity to SEQ ID NO: 44 and the light chain sequence has at least 99% sequence identity to SEQ ID NO: 45; or the heavy chain sequence comprises SEQ ID NO: 44 and the light chain sequence comprises SEQ ID NO: 45. [0135] In certain embodiments, the disclosure provides for an anti-PD-L1 antibody including a heavy chain and a light chain variable region sequence, where: (a) the heavy chain sequence has at least 85% sequence identity to the heavy chain sequence: EVQLLESGGGLVQPGGSLRLSCAASGFTFSMYMMMWVRQAPGKGLEVWSSIYPSGGI TFYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAIYYCARIKLGTVTTVDYWG QGTLVTVSS (SEQ ID NO: 46), and (b) the light chain sequence has at least 85% sequence identity to the light chain sequence: QSALTQPASVSGSPGQSITISCTGTSSDVGAYNYVSWYQQHPGKAPKLMIYDVSNR PSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSYTSSSTRVFGTGTKVTVL (SEQ ID NO: 47). [0136] In various embodiments, the heavy chain sequence has at least 86% sequence identity to SEQ ID NO: 46 and the light chain sequence has at least 86% sequence identity to SEQ ID NO: 47; the heavy chain sequence has at least 87% sequence identity to SEQ ID NO: 46 and the light chain sequence has at least 87% sequence identity to SEQ ID NO: 47; the heavy chain sequence has at least 88% sequence identity to SEQ ID NO: 46 and the light chain sequence has at least 88% sequence identity to SEQ ID NO: 47; the heavy chain sequence has at least 89% sequence identity to SEQ ID NO: 46 and the light chain sequence has at least 89% sequence identity to SEQ ID NO: 47; the heavy chain sequence has at least, 90% sequence identity to SEQ ID NO: 46 and the light chain sequence has at least 90% sequence identity to SEQ ID NO:47; the heavy chain sequence has at least 91% sequence identity to SEQ ID NO: 46 and the light chain sequence has at least 91% sequence identity to SEQ ID NO: 47; the heavy chain sequence has at least 92% sequence identity to SEQ ID NO: 46 and the light chain sequence has at least 92% sequence identity to SEQ ID NO: 47; the heavy chain sequence has at least 93% sequence identity to SEQ ID NO: 46 and the light chain sequence has at least 93% sequence identity to SEQ ID NO: 47; the heavy chain sequence has at least 94% sequence identity to SEQ ID NO: 46 and the light chain sequence has at least 94% sequence identity to SEQ ID NO: 47; the heavy chain sequence has at least 95% sequence identity to SEQ ID NO: 46 and the light chain sequence has at least 95% sequence identity to SEQ ID NO: 47; the heavy chain sequence has at least 96% sequence identity to SEQ ID NO: 46 and the light chain sequence has at least 96% sequence identity to SEQ ID NO: 47; the heavy chain sequence has at least 97% sequence identity to SEQ ID NO: 46 and the light chain sequence has at least 97% sequence identity to SEQ ID NO: 47; the heavy chain sequence has at least 98% sequence identity to SEQ ID NO: 46 and the light chain sequence has at least 98% sequence identity to SEQ ID NO: 47; the heavy chain sequence has at least 99% sequence identity to SEQ ID NO: 46 and the light chain sequence has at least 99% sequence identity to SEQ ID NO: 47; or the heavy chain sequence comprises SEQ ID NO: 46 and the light chain sequence comprises SEQ ID NO: 47. [0137] In another embodiment the antibody binds to human, mouse, or cynomolgus monkey PD-L1. In a specific aspect the antibody is capable of blocking the interaction between human, mice, or cynomolgus monkey PD-L1 and the respective human, mouse, or cynomolgus monkey PD-1 receptors. [0138] In another embodiment, the antibody binds to human PD-L1 with a KD of 5x10 -9 M or less, preferably with a KD of 2x10 -9 M or less, and even more preferred with a KD of 1x10 -9 M or less. [0139] In yet another embodiment, the disclosure relates to an anti-PD-L1 antibody or antigen binding fragment thereof which binds to a functional epitope including residues Y56 and D61 of human PD-L1. [0140] In a specific aspect, the functional epitope further includes E58, E60, Q66, R113, and M115 of human PD-L1. [0141] In a more specific aspect, the antibody binds to a conformational epitope, including residues 54-66 and 112-122 of human PD-L1. [0142] In certain embodiments, the disclosure is related to an anti-PD-L1 antibody, or antigen binding fragment thereof, which cross-competes for binding to PD-L1 with an antibody according to the disclosure as described herein. [0143] In certain embodiments, the disclosure features proteins and polypeptides including any of the above described anti-PD-L1 antibodies in combination with at least one

pharmaceutically acceptable carrier. [0144] In certain embodiments, the disclosure features an isolated nucleic acid encoding a polypeptide, or light chain or a heavy chain variable region sequence of an anti-PD-L1 antibody, or antigen binding fragment thereof, as described herein. In certain embodiments, the disclosure provides for an isolated nucleic acid encoding a light chain or a heavy chain variable region sequence of an anti-PD-L1 antibody, wherein: (a) the heavy chain includes an HVR-H1, an HVR-H2, and an HVR-H3 sequence having at least 80% sequence identity to SYIMM (SEQ ID NO: 35), SIYPSGGITFYADTVKG (SEQ ID NO: 36), and IKLGTVTTVDY (SEQ ID NO: 37), respectively, or (b) the light chain includes an HVR-L1, an HVR-L2, and an HVR-L3 sequence having at least 80% sequence identity to TGTSSDVGGYNYVS (SEQ ID NO: 38), DVSNRPS (SEQ ID NO: 39), and SSYTSSSTRV (SEQ ID NO: 40), respectively. [0145] In a specific aspect, the sequence identity is 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%. [0146] In a further aspect, the nucleic acid sequence for the heavy chain is:

(SEQ ID NO: 48)

and the nucleic acid sequence for the light chain is:

(SEQ ID NO: 49). [0147] Further exemplary anti-PD-L1 antibodies that can be used in an anti-PD-L1/TGF Trap are described in US patent application publication US 2010/0203056. In one embodiment of the disclosure, the antibody moiety is YW243.55S70. In another embodiment of the disclosure, the antibody moiety is MPDL3289A. [0148] In certain embodiments, the disclosure features an anti-PD-L1 antibody moiety including a heavy chain and a light chain variable region sequence, where: (a) the heavy chain sequence has at least 85% sequence identity to the heavy chain sequence:

(b) the light chain sequence has at least 85% sequence identity to the light chain sequence:

[0149] In various embodiments, the heavy chain sequence has at least 86% sequence identity to SEQ ID NO: 12 and the light chain sequence has at least 86% sequence identity to SEQ ID NO: 13; the heavy chain sequence has at least 87% sequence identity to SEQ ID NO: 12 and the light chain sequence has at least87% sequence identity to SEQ ID NO: 13; the heavy chain sequence has at least 88% sequence identity to SEQ ID NO: 12 and the light chain sequence has at least 88% sequence identity to SEQ ID NO: 13; the heavy chain sequence has at least89% sequence identity to SEQ ID NO: 12 and the light chain sequence has at least 89% sequence identity to SEQ ID NO: 13; the heavy chain sequence has at least, 90% sequence identity to SEQ ID NO: 12 and the light chain sequence has at least 90% sequence identity to SEQ ID NO: 13; the heavy chain sequence has at least 91% sequence identity to SEQ ID NO: 12 and the light chain sequence has at least 91% sequence identity to SEQ ID NO: 13; the heavy chain sequence has at least 92% sequence identity to SEQ ID NO: 12 and the light chain sequence has at least 92% sequence identity to SEQ ID NO: 13; the heavy chain sequence has at least 93% sequence identity to SEQ ID NO: 12 and the light chain sequence has at least 93% sequence identity to SEQ ID NO: 13; the heavy chain sequence has at least 94% sequence identity to SEQ ID NO: 12 and the light chain sequence has at least 94% sequence identity to SEQ ID NO: 13; the heavy chain sequence has at least 95% sequence identity to SEQ ID NO: 12 and the light chain sequence has at least 95% sequence identity to SEQ ID NO: 13; the heavy chain sequence has at least 96% sequence identity to SEQ ID NO: 12 and the light chain sequence has at least 96% sequence identity to SEQ ID NO: 13; the heavy chain sequence has at least 97% sequence identity to SEQ ID NO: 12 and the light chain sequence has at least 97% sequence identity to SEQ ID NO: 13; the heavy chain sequence has at least 98% sequence identity to SEQ ID NO: 12 and the light chain sequence has at least 98% sequence identity to SEQ ID NO: 13; the heavy chain sequence has at least 99% sequence identity to SEQ ID NO: 12 and the light chain sequence has at least 99% sequence identity to SEQ ID NO: 13; or the heavy chain sequence comprises SEQ ID NO: 12 and the light chain sequence comprises SEQ ID NO: 13. [0150] In certain embodiments, the disclosure features an anti-PD-L1 antibody moiety including a heavy chain and a light chain variable region sequence, where: (a) the heavy chain sequence has at least 85% sequence identity to the heavy chain sequence:

(b) the light chain sequence has at least 85% sequence identity to the light chain sequence:

[0151] In various embodiments, the heavy chain sequence has at least 86% sequence identity to SEQ ID NO: 14 and the light chain sequence has at least 86% sequence identity to SEQ ID NO: 13; the heavy chain sequence has at least 87% sequence identity to SEQ ID NO: 14 and the light chain sequence has at least 87% sequence identity to SEQ ID NO: 13; the heavy chain sequence has at least 88% sequence identity to SEQ ID NO: 14 and the light chain sequence has at least 88% sequence identity to SEQ ID NO: 13; the heavy chain sequence has at least 89% sequence identity to SEQ ID NO: 14 and the light chain sequence has at least 89% sequence identity to SEQ ID NO: 13; the heavy chain sequence has at least, 90% sequence identity to SEQ ID NO: 14 and the light chain sequence has at least 90% sequence identity to SEQ ID NO: 13; the heavy chain sequence has at least 91% sequence identity to SEQ ID NO: 14 and the light chain sequence has at least 91% sequence identity to SEQ ID NO: 13; the heavy chain sequence has at least 92% sequence identity to SEQ ID NO: 14 and the light chain sequence has at least 92% sequence identity to SEQ ID NO: 13; the heavy chain sequence has at least 93% sequence identity to SEQ ID NO: 14 and the light chain sequence has at least 93% sequence identity to SEQ ID NO: 13; the heavy chain sequence has at least 94% sequence identity to SEQ ID NO: 14 and the light chain sequence has at least 94% sequence identity to SEQ ID NO: 13; the heavy chain sequence has at least 95% sequence identity to SEQ ID NO: 14 and the light chain sequence has at least 95% sequence identity to SEQ ID NO: 13; the heavy chain sequence has at least 96% sequence identity to SEQ ID NO: 14 and the light chain sequence has at least 96% sequence identity to SEQ ID NO: 13; the heavy chain sequence has at least 97% sequence identity to SEQ ID NO: 14 and the light chain sequence has at least 97% sequence identity to SEQ ID NO: 13; the heavy chain sequence has at least 98% sequence identity to SEQ ID NO: 14 and the light chain sequence has at least 98% sequence identity to SEQ ID NO: 13; the heavy chain sequence has at least 99% sequence identity to SEQ ID NO: 14 and the light chain sequence has at least 99% sequence identity to SEQ ID NO: 13; or the heavy chain sequence comprises SEQ ID NO: 14 and the light chain sequence comprises SEQ ID NO: 13. [0152] Further exemplary anti-PD-L1 antibodies that can be used in an anti-PD-L1/TGF Trap are described in US patent application publication US 2018/0334504. [0153] In certain embodiments, the disclosure features an anti-PD-L1 antibody moiety including a heavy chain and a light chain variable region sequence, where (a) the heavy chain sequence has at least 85% sequence identity to the heavy chain sequence:

(b) the light chain sequence has at least 85% sequence identity to the light chain sequence:

[0154] In various embodiments, the heavy chain sequence has at least 86% sequence identity to SEQ ID NO: 55 and the light chain sequence has at least 86% sequence identity to SEQ ID NO: 56; the heavy chain sequence has at least 87% sequence identity to SEQ ID NO: 55 and the light chain sequence has at least 87% sequence identity to SEQ ID NO: 56; the heavy chain sequence has at least 88% sequence identity to SEQ ID NO: 55 and the light chain sequence has at least 88% sequence identity to SEQ ID NO: 56; the heavy chain sequence has at least 89% sequence identity to SEQ ID NO: 55 and the light chain sequence has at least 89% sequence identity to SEQ ID NO: 56; the heavy chain sequence has at least, 90% sequence identity to SEQ ID NO: 55 and the light chain sequence has at least 90% sequence identity to SEQ ID NO: 56; the heavy chain sequence has at least 91% sequence identity to SEQ ID NO: 55 and the light chain sequence has at least 91% sequence identity to SEQ ID NO: 56; the heavy chain sequence has at least 92% sequence identity to SEQ ID NO: 55 and the light chain sequence has at least 92% sequence identity to SEQ ID NO: 56; the heavy chain sequence has at least 93% sequence identity to SEQ ID NO: 55 and the light chain sequence has at least 93% sequence identity to SEQ ID NO: 56; the heavy chain sequence has at least 94% sequence identity to SEQ ID NO: 55 and the light chain sequence has at least 94% sequence identity to SEQ ID NO: 56; the heavy chain sequence has at least 95% sequence identity to SEQ ID NO: 55 and the light chain sequence has at least 95% sequence identity to SEQ ID NO: 56; the heavy chain sequence has at least 96% sequence identity to SEQ ID NO: 55 and the light chain sequence has at least 96% sequence identity to SEQ ID NO: 56; the heavy chain sequence has at least 97% sequence identity to SEQ ID NO: 55 and the light chain sequence has at least 97% sequence identity to SEQ ID NO: 56; the heavy chain sequence has at least 98% sequence identity to SEQ ID NO: 55 and the light chain sequence has at least 98% sequence identity to SEQ ID NO: 56; the heavy chain sequence has at least 99% sequence identity to SEQ ID NO: 55 and the light chain sequence has at least 99% sequence identity to SEQ ID NO: 56; or the heavy chain sequence comprises SEQ ID NO: 55 and the light chain sequence comprises SEQ ID NO: 56. [0155] In certain embodiments, the disclosure features an anti-PD-L1 antibody moiety including a heavy chain and a light chain variable region sequence, where (a) the heavy chain sequence has at least 85% sequence identity to the heavy chain sequence:

(b) the light chain sequence has at least 85% sequence identity to the light chain sequence:

[0156] In various embodiments, the heavy chain sequence has at least 86% sequence identity to SEQ ID NO: 57 and the light chain sequence has at least 86% sequence identity to SEQ ID NO: 58; the heavy chain sequence has at least 87% sequence identity to SEQ ID NO: 57 and the light chain sequence has at least 87% sequence identity to SEQ ID NO: 58; the heavy chain sequence has at least 88% sequence identity to SEQ ID NO: 57 and the light chain sequence has at least 88% sequence identity to SEQ ID NO: 58; the heavy chain sequence has at least 89% sequence identity to SEQ ID NO: 57 and the light chain sequence has at least 89% sequence identity to SEQ ID NO: 58; the heavy chain sequence has at least, 90% sequence identity to SEQ ID NO: 57 and the light chain sequence has at least 90% sequence identity to SEQ ID NO: 58; the heavy chain sequence has at least 91% sequence identity to SEQ ID NO: 57 and the light chain sequence has at least 91% sequence identity to SEQ ID NO: 58; the heavy chain sequence has at least 92% sequence identity to SEQ ID NO: 57 and the light chain sequence has at least 92% sequence identity to SEQ ID NO: 58; the heavy chain sequence has at least 93% sequence identity to SEQ ID NO: 57 and the light chain sequence has at least 93% sequence identity to SEQ ID NO: 58; the heavy chain sequence has at least 94% sequence identity to SEQ ID NO: 57 and the light chain sequence has at least 94% sequence identity to SEQ ID NO: 58; the heavy chain sequence has at least 95% sequence identity to SEQ ID NO: 57 and the light chain sequence has at least 95% sequence identity to SEQ ID NO: 58; the heavy chain sequence has at least 96% sequence identity to SEQ ID NO: 57 and the light chain sequence has at least 96% sequence identity to SEQ ID NO: 58; the heavy chain sequence has at least 97% sequence identity to SEQ ID NO: 57 and the light chain sequence has at least 97% sequence identity to SEQ ID NO: 58; the heavy chain sequence has at least 98% sequence identity to SEQ ID NO: 57 and the light chain sequence has at least 98% sequence identity to SEQ ID NO: 58; the heavy chain sequence has at least 99% sequence identity to SEQ ID NO: 57 and the light chain sequence has at least 99% sequence identity to SEQ ID NO: 58; or the heavy chain sequence comprises SEQ ID NO: 57 and the light chain sequence comprises SEQ ID NO: 58. [0157] In certain embodiments, the disclosure features an anti-PD-L1 antibody moiety including a heavy chain and a light chain sequence, where (a) the heavy chain sequence has at least 85% sequence identity to the heavy chain sequence:

(b) the light chain sequence has at least 85% sequence identity to the light chain sequence:

[0158] In various embodiments, the heavy chain sequence has at least 86% sequence identity to SEQ ID NO: 59 and the light chain sequence has at least 86% sequence identity to SEQ ID NO: 60; the heavy chain sequence has at least 87% sequence identity to SEQ ID NO: 59 and the light chain sequence has at least 87% sequence identity to SEQ ID NO: 60; the heavy chain sequence has at least 88% sequence identity to SEQ ID NO: 59 and the light chain sequence has at least 88% sequence identity to SEQ ID NO: 60; the heavy chain sequence has at least 89% sequence identity to SEQ ID NO: 59 and the light chain sequence has at least 89% sequence identity to SEQ ID NO: 60; the heavy chain sequence has at least, 90% sequence identity to SEQ ID NO: 59 and the light chain sequence has at least 90% sequence identity to SEQ ID NO: 60; the heavy chain sequence has at least 91% sequence identity to SEQ ID NO: 59 and the light chain sequence has at least 91% sequence identity to SEQ ID NO: 60; the heavy chain sequence has at least 92% sequence identity to SEQ ID NO: 59 and the light chain sequence has at least 92% sequence identity to SEQ ID NO: 60; the heavy chain sequence has at least 93% sequence identity to SEQ ID NO: 59 and the light chain sequence has at least 93% sequence identity to SEQ ID NO: 60; the heavy chain sequence has at least 94% sequence identity to SEQ ID NO: 59 and the light chain sequence has at least 94% sequence identity to SEQ ID NO: 60; the heavy chain sequence has at least 95% sequence identity to SEQ ID NO: 59 and the light chain sequence has at least 95% sequence identity to SEQ ID NO: 60; the heavy chain sequence has at least 96% sequence identity to SEQ ID NO: 59 and the light chain sequence has at least 96% sequence identity to SEQ ID NO: 60; the heavy chain sequence has at least 97% sequence identity to SEQ ID NO: 59 and the light chain sequence has at least 97% sequence identity to SEQ ID NO: 60; the heavy chain sequence has at least 98% sequence identity to SEQ ID NO: 59 and the light chain sequence has at least 98% sequence identity to SEQ ID NO: 60; the heavy chain sequence has at least 99% sequence identity to SEQ ID NO: 59 and the light chain sequence has at least 99% sequence identity to SEQ ID NO: 60; or the heavy chain sequence comprises SEQ ID NO: 59 and the light chain sequence comprises SEQ ID NO: 60. [0159] In certain embodiments, the disclosure features an anti-PD-L1 antibody moiety including a heavy chain and a light chain sequence, where (a) the heavy chain sequence has at least 85% sequence identity to the heavy chain sequence:

(b) the light chain sequence has at least 85% sequence identity to the light chain sequence:

[0160] In various embodiments, the heavy chain sequence has at least 86% sequence identity to SEQ ID NO: 61 and the light chain sequence has at least 86% sequence identity to SEQ ID NO: 62; the heavy chain sequence has at least 87% sequence identity to SEQ ID NO: 61 and the light chain sequence has at least 87% sequence identity to SEQ ID NO: 62; the heavy chain sequence has at least 88% sequence identity to SEQ ID NO: 61 and the light chain sequence has at least 88% sequence identity to SEQ ID NO: 62; the heavy chain sequence has at least 89% sequence identity to SEQ ID NO: 61 and the light chain sequence has at least 89% sequence identity to SEQ ID NO: 62; the heavy chain sequence has at least, 90% sequence identity to SEQ ID NO: 61 and the light chain sequence has at least 90% sequence identity to SEQ ID NO: 62; the heavy chain sequence has at least 91% sequence identity to SEQ ID NO: 61 and the light chain sequence has at least 91% sequence identity to SEQ ID NO: 62; the heavy chain sequence has at least 92% sequence identity to SEQ ID NO: 61 and the light chain sequence has at least 92% sequence identity to SEQ ID NO: 62; the heavy chain sequence has at least 93% sequence identity to SEQ ID NO: 61 and the light chain sequence has at least 93% sequence identity to SEQ ID NO: 62; the heavy chain sequence has at least 94% sequence identity to SEQ ID NO: 61 and the light chain sequence has at least 94% sequence identity to SEQ ID NO: 62; the heavy chain sequence has at least 95% sequence identity to SEQ ID NO: 61 and the light chain sequence has at least 95% sequence identity to SEQ ID NO: 62; the heavy chain sequence has at least 96% sequence identity to SEQ ID NO: 61 and the light chain sequence has at least 96% sequence identity to SEQ ID NO: 62; the heavy chain sequence has at least 97% sequence identity to SEQ ID NO: 61 and the light chain sequence has at least 97% sequence identity to SEQ ID NO: 62; the heavy chain sequence has at least 98% sequence identity to SEQ ID NO: 61 and the light chain sequence has at least 98% sequence identity to SEQ ID NO: 62; the heavy chain sequence has at least 99% sequence identity to SEQ ID NO: 61 and the light chain sequence has at least 99% sequence identity to SEQ ID NO: 62; or the heavy chain sequence comprises SEQ ID NO: 61 and the light chain sequence comprises SEQ ID NO: 62. [0161] Yet further exemplary anti-PD-L1 antibodies that can be used in an anti-PD- L1/TGF Trap are described in US patent publication US 7,943,743. [0162] In one embodiment of the disclosure, the anti-PD-L1 antibody is MDX-1105. [0163] In certain embodiments, the anti-PD-L1 antibody is MEDI-4736. Constant Region [0164] The proteins and peptides of the disclosure can include a constant region of an immunoglobulin or a fragment, analog, variant, mutant, or derivative of the constant region. In certain embodiments, the constant region is derived from a human immunoglobulin heavy chain, for example, IgG1, IgG2, IgG3, IgG4, or other classes. In certain embodiments, the constant region includes a CH2 domain. In certain embodiments, the constant region includes CH2 and CH3 domains or includes hinge-CH2-CH3. Alternatively, the constant region can include all or a portion of the hinge region, the CH2 domain and/or the CH3 domain. [0165] In one embodiment, the constant region contains a mutation that reduces affinity for an Fc receptor or reduces Fc effector function. For example, the constant region can contain a mutation that eliminates the glycosylation site within the constant region of an IgG heavy chain. In some embodiments, the constant region contains mutations, deletions, or insertions at an amino acid position corresponding to Leu234, Leu235, Gly236, Gly237, Asn297, or Pro331 of IgG1 (amino acids are numbered according to EU nomenclature). In a particular embodiment, the constant region contains a mutation at an amino acid position corresponding to Asn297 of IgG1. In alternative embodiments, the constant region contains mutations, deletions, or insertions at an amino acid position corresponding to Leu281, Leu282, Gly283, Gly284, Asn344, or Pro378 of IgG1. [0166] In some embodiments, the constant region contains a CH2 domain derived from a human IgG2 or IgG4 heavy chain. Preferably, the CH2 domain contains a mutation that eliminates the glycosylation site within the CH2 domain. In one embodiment, the mutation alters the asparagine within the Gln-Phe-Asn-Ser (SEQ ID NO: 15) amino acid sequence within the CH2 domain of the IgG2 or IgG4 heavy chain. Preferably, the mutation changes the asparagine to a glutamine. Alternatively, the mutation alters both the phenylalanine and the asparagine within the Gln-Phe-Asn-Ser (SEQ ID NO: 15) amino acid sequence. In one embodiment, the Gln-Phe-Asn-Ser (SEQ ID NO: 15) amino acid sequence is replaced with a Gln-Ala-Gln-Ser (SEQ ID NO: 16) amino acid sequence. The asparagine within the Gln-Phe- Asn-Ser (SEQ ID NO: 15) amino acid sequence corresponds to Asn297 of IgG1. [0167] In another embodiment, the constant region includes a CH2 domain and at least a portion of a hinge region. The hinge region can be derived from an immunoglobulin heavy chain, e.g., IgG1, IgG2, IgG3, IgG4, or other classes. Preferably, the hinge region is derived from human IgG1, IgG2, IgG3, IgG4, or other suitable classes. More preferably the hinge region is derived from a human IgG1 heavy chain. In one embodiment the cysteine in the Pro- Lys-Ser-Cys-Asp-Lys (SEQ ID NO: 17) amino acid sequence of the IgG1 hinge region is altered. In certain embodiments, the Pro-Lys-Ser-Cys-Asp-Lys (SEQ ID NO: 17) amino acid sequence is replaced with a Pro-Lys-Ser-Ser-Asp-Lys (SEQ ID NO: 18) amino acid sequence. In certain embodiments, the constant region includes a CH2 domain derived from a first antibody isotype and a hinge region derived from a second antibody isotype. In certain embodiments, the CH2 domain is derived from a human IgG2 or IgG4 heavy chain, while the hinge region is derived from an altered human IgG1 heavy chain. [0168] The alteration of amino acids near the junction of the Fc portion and the non-Fc portion can dramatically increase the serum half-life of the Fc fusion protein (PCT publication WO 0158957, the disclosure of which is hereby incorporated by reference). Accordingly, the junction region of a protein or polypeptide of the present disclosure can contain alterations that, relative to the naturally-occurring sequences of an immunoglobulin heavy chain and erythropoietin, preferably lie within about 10 amino acids of the junction point. These amino acid changes can cause an increase in hydrophobicity. In one embodiment, the constant region is derived from an IgG sequence in which the C-terminal lysine residue is replaced. Preferably, the C-terminal lysine of an IgG sequence is replaced with a non-lysine amino acid, such as alanine or leucine, to further increase serum half-life. In another embodiment, the constant region is derived from an IgG sequence in which the Leu-Ser-Leu-Ser (SEQ ID NO: 19) amino acid sequence near the C-terminus of the constant region is altered to eliminate potential junctional T-cell epitopes. For example, in one embodiment, the Leu-Ser-Leu-Ser (SEQ ID NO: 19) amino acid sequence is replaced with an Ala-Thr-Ala-Thr (SEQ ID NO: 20) amino acid sequence. In other embodiments, the amino acids within the Leu-Ser-Leu-Ser (SEQ ID NO: 19) segment are replaced with other amino acids such as glycine or proline. Detailed methods of generating amino acid substitutions of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) segment near the C-terminus of an IgG1, IgG2, IgG3, IgG4, or other immunoglobulin class molecule have been described in U.S. Patent Publication No.20030166877, the disclosure of which is hereby incorporated by reference. [0169] Suitable hinge regions for the present disclosure can be derived from IgG1, IgG2, IgG3, IgG4, and other immunoglobulin classes. The IgG1 hinge region has three cysteines, two of which are involved in disulfide bonds between the two heavy chains of the immunoglobulin. These same cysteines permit efficient and consistent disulfide bonding formation between Fc portions. Therefore, a hinge region of the present disclosure is derived from IgG1, e.g., human IgG1. In some embodiments, the first cysteine within the human IgG1 hinge region is mutated to another amino acid, preferably serine. The IgG2 isotype hinge region has four disulfide bonds that tend to promote oligomerization and possibly incorrect disulfide bonding during secretion in recombinant systems. A suitable hinge region can be derived from an IgG2 hinge; the first two cysteines are each preferably mutated to another amino acid. The hinge region of IgG4 is known to form interchain disulfide bonds inefficiently. However, a suitable hinge region for the present disclosure can be derived from the IgG4 hinge region, preferably containing a mutation that enhances correct formation of disulfide bonds between heavy chain- derived moieties (Angal S, et al. (1993) Mol. Immunol., 30:105-8). [0170] In accordance with the present disclosure, the constant region can contain CH2 and/or CH3 domains and a hinge region that are derived from different antibody isotypes, i.e., a hybrid constant region. For example, in one embodiment, the constant region contains CH2 and/or CH3 domains derived from IgG2 or IgG4 and a mutant hinge region derived from IgG1. Alternatively, a mutant hinge region from another IgG subclass is used in a hybrid constant region. For example, a mutant form of the IgG4 hinge that allows efficient disulfide bonding between the two heavy chains can be used. A mutant hinge can also be derived from an IgG2 hinge in which the first two cysteines are each mutated to another amino acid. Assembly of such hybrid constant regions has been described in U.S. Patent Publication No. 20030044423, the disclosure of which is hereby incorporated by reference. [0171] In accordance with the present disclosure, the constant region can contain one or more mutations described herein. The combinations of mutations in the Fc portion can have additive or synergistic effects on the prolonged serum half-life and increased in vivo potency of the bifunctional molecule. Thus, in one exemplary embodiment, the constant region can contain (i) a region derived from an IgG sequence in which the Leu-Ser-Leu-Ser (SEQ ID NO: 19) amino acid sequence is replaced with an Ala-Thr-Ala-Thr (SEQ ID NO: 20) amino acid sequence; (ii) a C-terminal alanine residue instead of lysine; (iii) a CH2 domain and a hinge region that are derived from different antibody isotypes, for example, an IgG2 CH2 domain and an altered IgG1 hinge region; and (iv) a mutation that eliminates the glycosylation site within the IgG2-derived CH2 domain, for example, a Gln-Ala-Gln-Ser (SEQ ID NO: 16) amino acid sequence instead of the Gln-Phe-Asn-Ser (SEQ ID NO: 15) amino acid sequence within the IgG2-derived CH2 domain. Antibody fragments [0172] The proteins and polypeptides of the disclosure can also include antigen-binding fragments of antibodies. Exemplary antibody fragments include scFv, Fv, Fab, F(ab’) 2 , and single domain VHH fragments such as those of camelid origin. [0173] Single-chain antibody fragments, also known as single-chain antibodies (scFvs), are recombinant polypeptides which typically bind antigens or receptors; these fragments contain at least one fragment of an antibody variable heavy-chain amino acid sequence (V H ) tethered to at least one fragment of an antibody variable light-chain sequence (V L ) with or without one or more interconnecting linkers. Such a linker may be a short, flexible peptide selected to assure that the proper three-dimensional folding of the V L and V H domains occurs once they are linked so as to maintain the target molecule binding-specificity of the whole antibody from which the single-chain antibody fragment is derived. Generally, the carboxyl terminus of the V L or V H sequence is covalently linked by such a peptide linker to the amino acid terminus of a complementary V L and V H sequence. Single-chain antibody fragments can be generated by molecular cloning, antibody phage display library or similar techniques. These proteins can be produced either in eukaryotic cells or prokaryotic cells, including bacteria. [0174] Single-chain antibody fragments contain amino acid sequences having at least one of the variable regions or CDRs of the whole antibodies described in this specification, but are lacking some or all of the constant domains of those antibodies. These constant domains are not necessary for antigen binding, but constitute a major portion of the structure of whole antibodies. Single-chain antibody fragments may therefore overcome some of the problems associated with the use of antibodies containing part or all of a constant domain. For example, single-chain antibody fragments tend to be free of undesired interactions between biological molecules and the heavy-chain constant region, or other unwanted biological activity.

Additionally, single-chain antibody fragments are considerably smaller than whole antibodies and may therefore have greater capillary permeability than whole antibodies, allowing single- chain antibody fragments to localize and bind to target antigen-binding sites more efficiently. Also, antibody fragments can be produced on a relatively large scale in prokaryotic cells, thus facilitating their production. Furthermore, the relatively small size of single-chain antibody fragments makes them less likely than whole antibodies to provoke an immune response in a recipient. [0175] Fragments of antibodies that have the same or comparable binding characteristics to those of the whole antibody may also be present. Such fragments may contain one or both Fab fragments or the F(ab’) 2 fragment. The antibody fragments may contain all six CDRs of the whole antibody, although fragments containing fewer than all of such regions, such as three, four or five CDRs, are also functional. Pharmaceutical Compositions [0176] The present disclosure also features pharmaceutical compositions that contain a therapeutically effective amount of a protein described herein. The composition can be formulated for use in a variety of drug delivery systems. One or more physiologically acceptable excipients or carriers can also be included in the composition for proper formulation. Suitable formulations for use in the present disclosure are found in Remington's Pharmaceutical Sciences, Mack Publishing Company, Philadelphia, Pa., 17th ed., 1985. For a brief review of methods for drug delivery, see, e.g., Langer (Science 249:1527-1533, 1990). [0177] In one aspect, the present disclosure provides an intravenous drug delivery formulation for use in a method of treating cancer or inhibiting tumor growth in a treatment naïve cancer (e.g., NSCLC with high PD-L1 expression) patient that includes 500 mg– 2400 mg of a protein including a first polypeptide and a second polypeptide, the first polypeptide includes: (a) at least a variable region of a heavy chain of an antibody that binds to human protein Programmed Death Ligand 1 (PD-L1); and (b) human Transforming Growth Factor Receptor II (TGF RII), or a fragment thereof, capable of binding Transforming Growth Factor (TGF ), the second polypeptide includes at least a variable region of a light chain of an antibody that binds PD-L1, and the heavy chain of the first polypeptide and the light chain of the second polypeptide, when combined, form an antigen binding site that binds PD-L1. [0178] In certain embodiments, a protein product of the present disclosure includes a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3, and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1. In certain embodiments, a protein product of the present disclosure includes a first polypeptide that comprises the amino acid sequences of SEQ ID NOs: 35, 36, and 37, and a second polypeptide that comprises the amino acid sequences of SEQ ID NOs: 38, 39, and 40. [0179] In certain embodiments of the present disclosure, the intravenous drug delivery formulation for use in a method of treating cancer or inhibiting tumor growth in a treatment naïve cancer (e.g., NSCLC with high PD-L1 expression) patient may include about 500 mg to about 2400 mg dose (e.g., about 500 mg to about 2300 mg, about 500 mg to about 2200 mg, about 500 mg to about 2100 mg, about 500 mg to about 2000 mg, about 500 mg to about 1900 mg, about 500 mg to about 1800 mg, about 500 mg to about 1700 mg, about 500 mg to about 1600 mg, about 500 mg to about 1500 mg, about 500 mg to about 1400 mg, about 500 mg to about 1300 mg, about 500 mg to about 1200 mg, about 500 mg to about 1100 mg, about 500 mg to about 1000 mg, about 500 mg to about 900 mg, about 500 mg to about 800 mg, about 500 mg to about 700 mg, about 500 mg to about 600 mg, about 600 mg to 2400 mg, about 700 mg to 2400 mg, about 800 mg to 2400 mg, about 900 mg to 2400 mg, about 1000 mg to 2400 mg, about 1100 mg to 2400 mg, about 1200 mg to 2400 mg, about 1300 mg to 2400 mg, about 1400 mg to 2400 mg, about 1500 mg to 2400 mg, about 1600 mg to 2400 mg, about 1700 mg to 2400 mg, about 1800 mg to 2400 mg, about 1900 mg to 2400 mg, about 2000 mg to 2400 mg, about 2100 mg to 2400 mg, about 2200 mg to 2400 mg, or about 2300 mg to 2400 mg) of a protein of the present disclosure (e.g., anti-PD-L1/TGF Trap (e.g., including a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3, and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1)). In certain embodiments, the intravenous drug delivery formulation may include about 500 to about 2000 mg dose of a protein of the present disclosure (e.g., anti-PD-L1/TGF Trap (e.g., including a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3, and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1)). In certain embodiments, the intravenous drug delivery formulation may include about 500 mg dose of a protein product of the present disclosure with a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3, and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1. In certain embodiments, the intravenous drug delivery formulation may include 500 mg dose of a protein of the present disclosure (e.g., anti-PD-L1/TGF Trap (e.g., including a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3, and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1)). In certain embodiments, the intravenous drug delivery formulation may include about 1200 mg dose of a protein product of the present disclosure with a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3, and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1. In certain embodiments, the intravenous drug delivery formulation may include 1200 mg dose of a protein of the present disclosure (e.g., anti-PD-L1/TGF Trap (e.g., including a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3, and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1)). In certain embodiments, the intravenous drug delivery formulation may include about 2400 mg dose of a protein product of the present disclosure with a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3, and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1. In certain embodiments, the intravenous drug delivery formulation may include 2400 mg dose of a protein of the present disclosure (e.g., anti-PD-L1/TGF Trap (e.g., including a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3, and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1)). In certain embodiments, the intravenous drug delivery formulation may include 2400 mg dose of a protein of the present disclosure (e.g., anti-PD-L1/TGF Trap (e.g., including a first polypeptide comprising the amino acid sequences of SEQ ID NOs: 35, 36, and 37, and a second polypeptide comprising the amino acid sequences of SEQ ID NOs: 38, 39, and 40)). [0180] In certain embodiments, the intravenous drug delivery formulation for use in a method of treating cancer or inhibiting tumor growth in a treatment naïve cancer (e.g., NSCLC with high PD-L1 expression) patient may include about 1200 mg to about 3000 mg (e.g., about 1200 mg to about 3000 mg, about 1200 mg to about 2900 mg, about 1200 mg to about 2800 mg, about 1200 mg to about 2700 mg, about 1200 mg to about 2600 mg, about 1200 mg to about 2500 mg, about 1200 mg to about 2400 mg, about 1200 mg to about 2300 mg, about 1200 mg to about 2200 mg, about 1200 mg to about 2100 mg, about 1200 mg to about 2000 mg, about 1200 mg to about 1900 mg, about 1200 mg to about 1800 mg, about 1200 mg to about 1700 mg, about 1200 mg to about 1600 mg, about 1200 mg to about 1500 mg, about 1200 mg to about 1400 mg, about 1200 mg to about 1300 mg, about 1300 mg to about 3000 mg, about 1400 mg to about 3000 mg, about 1500 mg to about 3000 mg, about 1600 mg to about 3000 mg, about 1700 mg to about 3000 mg, about 1800 mg to about 3000 mg, about 1900 mg to about 3000 mg, about 2000 mg to about 3000 mg, about 2100 mg to about 3000 mg, about 2200 mg to about 3000 mg, about 2300 mg to about 3000 mg, about 2400 mg to about 3000 mg, about 2500 mg to about 3000 mg, about 2600 mg to about 3000 mg, about 2700 mg to about 3000 mg, about 2800 mg to about 3000 mg, about 2900 mg to about 3000 mg, about 1200 mg, about 1300 mg, about 1400 mg, about 1500 mg, about 1600 mg, about 1700 mg, about 1800 mg, about 1900 mg, about 2000 mg, about 2100 mg, about 2200 mg, about 2300 mg, about 2400 mg, about 2500 mg, about 2600 mg, about 2700 mg, about 2800 mg, about 2900 mg, or about 3000 mg) of a protein product of the present disclosure (e.g., anti- PD-L1/TGF Trap). In certain embodiments, the intravenous drug delivery formulation for use in a method of treating cancer or inhibiting tumor growth in a treatment naïve cancer (e.g., NSCLC with high PD-L1 expression) patient may include about 1200 mg to about 3000 mg (e.g., about 1200 mg to about 3000 mg, about 1200 mg to about 2900 mg, about 1200 mg to about 2800 mg, about 1200 mg to about 2700 mg, about 1200 mg to about 2600 mg, about 1200 mg to about 2500 mg, about 1200 mg to about 2400 mg, about 1200 mg to about 2300 mg, about 1200 mg to about 2200 mg, about 1200 mg to about 2100 mg, about 1200 mg to about 2000 mg, about 1200 mg to about 1900 mg, about 1200 mg to about 1800 mg, about 1200 mg to about 1700 mg, about 1200 mg to about 1600 mg, about 1200 mg to about 1500 mg, about 1200 mg to about 1400 mg, about 1200 mg to about 1300 mg, about 1300 mg to about 3000 mg, about 1400 mg to about 3000 mg, about 1500 mg to about 3000 mg, about 1600 mg to about 3000 mg, about 1700 mg to about 3000 mg, about 1800 mg to about 3000 mg, about 1900 mg to about 3000 mg, about 2000 mg to about 3000 mg, about 2100 mg to about 3000 mg, about 2200 mg to about 3000 mg, about 2300 mg to about 3000 mg, about 2400 mg to about 3000 mg, about 2500 mg to about 3000 mg, about 2600 mg to about 3000 mg, about 2700 mg to about 3000 mg, about 2800 mg to about 3000 mg, about 2900 mg to about 3000 mg, about 1200 mg, about 1300 mg, about 1400 mg, about 1500 mg, about 1600 mg, about 1700 mg, about 1800 mg, about 1900 mg, about 2000 mg, about 2100 mg, about 2200 mg, about 2300 mg, about 2400 mg, about 2500 mg, about 2600 mg, about 2700 mg, about 2800 mg, about 2900 mg, or about 3000 mg) of a protein product with a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3, and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1; or a protein product with a first polypeptide that comprises the amino acid sequences of SEQ ID NOs: 35, 36, and 37, and a second polypeptide that comprises the amino acid sequences of SEQ ID NOs: 38, 39, and 40. [0181] In certain embodiments, the intravenous drug delivery formulation for use in a method of treating cancer or inhibiting tumor growth in a treatment naïve cancer (e.g., NSCLC with high PD-L1 expression) patient may include about 525 mg, about 550 mg, about 575 mg, about 600 mg, about 625 mg, about 650 mg, about 675 mg, about 700 mg, about 725 mg, about 750 mg, about 775 mg, about 800 mg, about 825 mg, about 850 mg, about 875 mg, about 900 mg, about 925 mg, about 950 mg, about 975 mg, about 1000 mg, about 1025 mg, about 1050 mg, about 1075 mg, about 1100 mg, about 1125 mg, about 1150 mg, about 1175 mg, about 1200 mg, about 1225 mg, about 1250 mg, about 1275 mg, about 1300 mg, about 1325 mg, about 1350 mg, about 1375 mg, about 1400 mg, about 1425 mg, about 1450 mg, about 1475 mg, about 1500 mg, about 1525 mg, about 1550 mg, about 1575 mg, about 1600 mg, about 1625 mg, about 1650 mg, about 1675 mg, about 1700 mg, about 1725 mg, about 1750 mg, about 1775 mg, about 1800 mg, about 1825 mg, about 1850 mg, about 1875 mg, about 1900 mg, about 1925 mg, about 1950 mg, about 1975 mg, about 2000 mg, about 2025 mg, about 2050 mg, about 2075 mg, about 2100 mg, about 2125 mg, about 2150 mg, about 2175 mg, about 2200 mg, about 2225 mg, about 2250 mg, about 2275 mg, about 2300 mg, about 2325 mg, about 2350 mg, about 2375 mg, or about 2400 mg of the protein of the present disclosure (e.g., anti-PD-L1/TGF Trap comprising a first polypeptide that comprises the amino acid sequences of SEQ ID NOs: 35, 36, and 37, and a second polypeptide that comprises the amino acid sequences of SEQ ID NOs: 38, 39, and 40). [0182] The intravenous drug delivery formulation of the present disclosure for use in a method of treating cancer or inhibiting tumor growth in a treatment naïve cancer (e.g., NSCLC with high PD-L1 expression) patient may be contained in a bag, a pen, or a syringe. In certain embodiments, the bag may be connected to a channel comprising a tube and/or a needle. In certain embodiments, the formulation may be a lyophilized formulation or a liquid formulation. In certain embodiments, the formulation may be freeze-dried (lyophilized) and contained in about 12-60 vials. In certain embodiments, the formulation may be freeze-dried and about 45 mg of the freeze-dried formulation may be contained in one vial. In certain embodiments, the about 40 mg– about 100 mg of freeze-dried formulation may be contained in one vial. In certain embodiments, freeze dried formulation from 12, 27, or 45 vials are combined to obtained a therapeutic dose of the protein in the intravenous drug formulation. In certain embodiments, the formulation may be a liquid formulation of a protein product with a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3, and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1; or a protein product with a first polypeptide that comprises the amino acid sequences of SEQ ID NOs: 35, 36, and 37, and a second polypeptide that comprises the amino acid sequences of SEQ ID NOs: 38, 39, and 40, and stored as about 250 mg/vial to about 2000 mg/vial (e.g., about 250 mg/vial to about 2000 mg/vial, about 250 mg/vial to about 1900 mg/vial, about 250 mg/vial to about 1800 mg/vial, about 250 mg/vial to about 1700 mg/vial, about 250 mg/vial to about 1600 mg/vial, about 250 mg/vial to about 1500 mg/vial, about 250 mg/vial to about 1400 mg/vial, about 250 mg/vial to about 1300 mg/vial, about 250 mg/vial to about 1200 mg/vial, about 250 mg/vial to about 1100 mg/vial, about 250 mg/vial to about 1000 mg/vial, about 250 mg/vial to about 900 mg/vial, about 250 mg/vial to about 800 mg/vial, about 250 mg/vial to about 700 mg/vial, about 250 mg/vial to about 600 mg/vial, about 250 mg/vial to about 500 mg/vial, about 250 mg/vial to about 400 mg/vial, about 250 mg/vial to about 300 mg/vial, about 300 mg/vial to about 2000 mg/vial, about 400 mg/vial to about 2000 mg/vial, about 500 mg/vial to about 2000 mg/vial, about 600 mg/vial to about 2000 mg/vial, about 700 mg/vial to about 2000 mg/vial, about 800 mg/vial to about 2000 mg/vial, about 900 mg/vial to about 2000 mg/vial, about 1000 mg/vial to about 2000 mg/vial, about 1100 mg/vial to about 2000 mg/vial, about 1200 mg/vial to about 2000 mg/vial, about 1300 mg/vial to about 2000 mg/vial, about 1400 mg/vial to about 2000 mg/vial, about 1500 mg/vial to about 2000 mg/vial, about 1600 mg/vial to about 2000 mg/vial, about 1700 mg/vial to about 2000 mg/vial, about 1800 mg/vial to about 2000 mg/vial, or about 1900 mg/vial to about 2000 mg/vial). In certain embodiments, the formulation may be a liquid formulation and stored as about 600 mg/vial. In certain embodiments, the formulation may be a liquid formulation and stored as about 1200 mg/vial. In certain embodiments, the formulation may be a liquid formulation and stored as about 1800 mg/vial. In certain embodiments, the formulation may be a liquid formulation and stored as about 250 mg/vial. [0183] This disclosure provides a liquid aqueous pharmaceutical formulation including a therapeutically effective amount of the protein of the present disclosure (e.g., anti-PD-L1/TGF Trap) in a buffered solution forming a formulation for use in a method of treating cancer or inhibiting tumor growth in a treatment naïve cancer (e.g., NSCLC with high PD-L1 expression) patient. [0184] These compositions for use in a method of treating cancer or inhibiting tumor growth in a treatment naïve cancer (e.g., NSCLC with high PD-L1 expression) patient may be sterilized by conventional sterilization techniques, or may be sterile filtered. The resulting aqueous solutions may be packaged for use as-is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration. The pH of the preparations typically will be between 3 and 11, more preferably between 5 and 9 or between 6 and 8, and most preferably between 7 and 8, such as 7 to 7.5. The resulting compositions in solid form may be packaged in multiple single dose units, each containing a fixed amount of the above-mentioned agent or agents. The composition in solid form can also be packaged in a container for a flexible quantity. [0185] In certain embodiments, the present disclosure provides for use in a method of treating cancer or inhibiting tumor growth in a treatment naïve cancer (e.g., NSCLC with high PD-L1 expression) patient, a formulation with an extended shelf life including a protein of the present disclosure (e.g., anti-PD-L1/TGF Trap (e.g., including a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3, and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1)), in combination with mannitol, citric acid monohydrate, sodium citrate, disodium phosphate dihydrate, sodium dihydrogen phosphate dihydrate, sodium chloride, polysorbate 80, water, and sodium hydroxide. [0186] In certain embodiments, an aqueous formulation for use in a method of treating cancer or inhibiting tumor growth in a treatment naïve cancer (e.g., NSCLC with high PD-L1 expression) patient is prepared including a protein of the present disclosure (e.g., anti-PD- L1/TGF Trap (e.g., including a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3, and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1; or a protein product with a first polypeptide that comprises the amino acid sequences of SEQ ID NOs: 35, 36, and 37, and a second polypeptide that comprises the amino acid sequences of SEQ ID NOs: 38, 39, and 40) in a pH-buffered solution. The buffer of this invention may have a pH ranging from about 4 to about 8, e.g., from about 4 to about 8, from about 4.5 to about 8, from about 5 to about 8, from about 5.5 to about 8, from about 6 to about 8, from about 6.5 to about 8, from about 7 to about 8, from about 7.5 to about 8, from about 4 to about 7.5, from about 4.5 to about 7.5, from about 5 to about about 7.5, from about 5.5 to about 7.5, from about 6 to about 7.5, from about 6.5 to about 7.5, from about 4 to about 7, from about 4.5 to about 7, from about 5 to about 7, from about 5.5 to about 7, from about 6 to about 7, from about 4 to about 6.5, from about 4.5 to about 6.5, from about 5 to about 6.5, from about 5.5 to about 6.5, from about 4 to about 6.0, from about 4.5 to about 6.0, from about 5 to about 6, or from about 4.8 to about 5.5, or may have a pH of about 5.0 to about 5.2. Ranges intermediate to the above recited pH's are also intended to be part of this disclosure. For example, ranges of values using a combination of any of the above recited values as upper and/or lower limits are intended to be included. Examples of buffers that will control the pH within this range include acetate (e.g. sodium acetate), succinate (such as sodium succinate), gluconate, histidine, citrate and other organic acid buffers. [0187] In certain embodiments, the formulation for use in a method of treating cancer or inhibiting tumor growth in a treatment naïve cancer (e.g., NSCLC with high PD-L1 expression) patient includes a buffer system which contains citrate and phosphate to maintain the pH in a range of about 4 to about 8. In certain embodiments the pH range may be from about 4.5 to about 6.0, or from about pH 4.8 to about 5.5, or in a pH range of about 5.0 to about 5.2. In certain embodiments, the buffer system includes citric acid monohydrate, sodium citrate, disodium phosphate dihydrate, and/or sodium dihydrogen phosphate dihydrate. In certain embodiments, the buffer system includes about 1.3 mg/ml of citric acid (e.g., 1.305 mg/ml), about 0.3 mg/ml of sodium citrate (e.g., 0.305 mg/ml), about 1.5 mg/ml of disodium phosphate dihydrate (e.g., 1.53 mg/ml), about 0.9 mg/ml of sodium dihydrogen phosphate dihydrate (e.g., 0.86), and about 6.2 mg/ml of sodium chloride (e.g., 6.165 mg/ml). In certain embodiments, the buffer system includes about 1-1.5 mg/ml of citric acid, about 0.25 to about 0.5 mg/ml of sodium citrate, about 1.25 to about 1.75 mg/ml of disodium phosphate dihydrate, about 0.7 to about 1.1 mg/ml of sodium dihydrogen phosphate dihydrate, and 6.0 to 6.4 mg/ml of sodium chloride. In certain embodiments, the pH of the formulation is adjusted with sodium hydroxide. [0188] A polyol, which acts as a tonicifier and may stabilize the antibody, may also be included in the formulation. The polyol is added to the formulation in an amount which may vary with respect to the desired isotonicity of the formulation. In certain embodiments, the aqueous formulation may be isotonic. The amount of polyol added may also alter with respect to the molecular weight of the polyol. For example, a lower amount of a monosaccharide (e.g. mannitol) may be added, compared to a disaccharide (such as trehalose). In certain

embodiments, the polyol which may be used in the formulation as a tonicity agent is mannitol. In certain embodiments, the mannitol concentration may be about 5 to about 20 mg/ml. In certain embodiments, the concentration of mannitol may be about 7.5 to about 15 mg/ml. In certain embodiments, the concentration of mannitol may be about 10– about 14 mg/ml. In certain embodiments, the concentration of mannitol may be about 12 mg/ml. In certain embodiments, the polyol sorbitol may be included in the formulation. [0189] A detergent or surfactant may also be added to the formulation. Exemplary detergents include nonionic detergents such as polysorbates (e.g. polysorbates 20, 80 etc.) or poloxamers (e.g., poloxamer 188). The amount of detergent added is such that it reduces aggregation of the formulated antibody and/or minimizes the formation of particulates in the formulation and/or reduces adsorption. In certain embodiments, the formulation may include a surfactant which is a polysorbate. In certain embodiments, the formulation may contain the detergent polysorbate 80 or Tween 80. Tween 80 is a term used to describe polyoxyethylene (20) sorbitanmonooleate (see Fiedler, Lexikon der Hilfsstoffe, Editio Cantor Verlag Aulendorf, 4th edi., 1996). In certain embodiments, the formulation may contain between about 0.1 mg/mL and about 10 mg/mL of polysorbate 80, or between about 0.5 mg/mL and about 5 mg/mL. In certain embodiments, about 0.1% polysorbate 80 may be added in the formulation. Lyophilized Formulation [0190] The lyophilized formulation for use in a method of treating cancer or inhibiting tumor growth in a treatment naïve cancer (e.g., NSCLC with high PD-L1 expression) patient of the present disclosure includes the anti-PD-L1/TGF Trap molecule and a lyoprotectant. The lyoprotectant may be sugar, e.g., disaccharides. In certain embodiments, the lycoprotectant may be sucrose or maltose. The lyophilized formulation may also include one or more of a buffering agent, a surfactant, a bulking agent, and/or a preservative. [0191] The amount of sucrose or maltose useful for stabilization of the lyophilized drug product may be in a weight ratio of at least 1:2 protein to sucrose or maltose. In certain embodiments, the protein to sucrose or maltose weight ratio may be of from 1:2 to 1:5. [0192] In certain embodiments, the pH of the formulation, prior to lyophilization, may be set by addition of a pharmaceutically acceptable acid and/or base. In certain embodiments the pharmaceutically acceptable acid may be hydrochloric acid. In certain embodiments, the pharmaceutically acceptable base may be sodium hydroxide. [0193] Before lyophilization, the pH of the solution containing the protein of the present disclosure may be adjusted between about 6 to about 8. In certain embodiments, the pH range for the lyophilized drug product may be from about 7 to about 8. [0194] In certain embodiments, a salt or buffer components may be added in an amount of about 10 mM– about 200 mM. The salts and/or buffers are pharmaceutically acceptable and are derived from various known acids (inorganic and organic) with“base forming” metals or amines. In certain embodiments, the buffer may be phosphate buffer. In certain embodiments, the buffer may be glycinate, carbonate, citrate buffers, in which case, sodium, potassium or ammonium ions can serve as counterion. [0195] In certain embodiments, a“bulking agent” may be added. A“bulking agent” is a compound which adds mass to a lyophilized mixture and contributes to the physical structure of the lyophilized cake (e.g., facilitates the production of an essentially uniform lyophilized cake which maintains an open pore structure). Illustrative bulking agents include mannitol, glycine, polyethylene glycol and sorbitol. The lyophilized formulations of the present invention may contain such bulking agents. [0196] A preservative may be optionally added to the formulations herein to reduce bacterial action. The addition of a preservative may, for example, facilitate the production of a multi-use (multiple-dose) formulation. [0197] In certain embodiments, the lyophilized drug product for use in a method of treating cancer or inhibiting tumor growth in a treatment naïve cancer (e.g., NSCLC with high PD-L1 expression) patient may be constituted with an aqueous carrier. The aqueous carrier of interest herein is one which is pharmaceutically acceptable (e.g., safe and non-toxic for administration to a human) and is useful for the preparation of a liquid formulation, after lyophilization.

Illustrative diluents include sterile water for injection (SWFI), bacteriostatic water for injection (BWFI), a pH buffered solution (e.g. phosphate-buffered saline), sterile saline solution, Ringer's solution or dextrose solution. [0198] In certain embodiments, the lyophilized drug product of the current disclosure is reconstituted with either Sterile Water for Injection, USP (SWFI) or 0.9% Sodium Chloride Injection, USP. During reconstitution, the lyophilized powder dissolves into a solution. [0199] In certain embodiments, the lyophilized protein product of the instant disclosure is constituted to about 4.5 mL water for injection and diluted with 0.9% saline solution (sodium chloride solution). Liquid Formulation [0200] In embodiments, the protein product of the present disclosure is formulated as a liquid formulation for use in a method of treating cancer or inhibiting tumor growth in a treatment naïve cancer (e.g., NSCLC with high PD-L1 expression) patient. The liquid formulation may be presented at a 10 mg/mL concentration in either a USP / Ph Eur type I 50R vial closed with a rubber stopper and sealed with an aluminum crimp seal closure. The stopper may be made of elastomer complying with USP and Ph Eur. In certain embodiments vials may be filled with about 61.2 mL of the protein product solution in order to allow an extractable volume of 60 mL. In certain embodiments, the liquid formulation may be diluted with 0.9% saline solution. In certain embodiments vials may contain about 61.2 mL of the protein product (e.g., anti-PD-L1/TGF Trap (e.g., including a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3, and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1)) solution of about 20 mg/mL to about 50 mg/mL (e.g., about 20 mg/mL, about 25 mg/mL, about 30 mg/mL, about 35 mg/mL, about 40 mg/mL, about 45 mg/mL or about 50 mg/mL) in order to allow an extractable volume of 60 mL for delivering about 1200 mg to about 3000 mg (e.g., about 1200 mg to about 3000 mg, about 1200 mg to about 2900 mg, about 1200 mg to about 2800 mg, about 1200 mg to about 2700 mg, about 1200 mg to about 2600 mg, about 1200 mg to about 2500 mg, about 1200 mg to about 2400 mg, about 1200 mg to about 2300 mg, about 1200 mg to about 2200 mg, about 1200 mg to about 2100 mg, about 1200 mg to about 2000 mg, about 1200 mg to about 1900 mg, about 1200 mg to about 1800 mg, about 1200 mg to about 1700 mg, about 1200 mg to about 1600 mg, about 1200 mg to about 1500 mg, about 1200 mg to about 1400 mg, about 1200 mg to about 1300 mg, about 1300 mg to about 3000 mg, about 1400 mg to about 3000 mg, about 1500 mg to about 3000 mg, about 1600 mg to about 3000 mg, about 1700 mg to about 3000 mg, about 1800 mg to about 3000 mg, about 1900 mg to about 3000 mg, about 2000 mg to about 3000 mg, about 2100 mg to about 3000 mg, about 2200 mg to about 3000 mg, about 2300 mg to about 3000 mg, about 2400 mg to about 3000 mg, about 2500 mg to about 3000 mg, about 2600 mg to about 3000 mg, about 2700 mg to about 3000 mg, about 2800 mg to about 3000 mg, about 2900 mg to about 3000 mg, about 1200 mg, about 1300 mg, about 1400 mg, about 1500 mg, about 1600 mg, about 1700 mg, about 1800 mg, about 1900 mg, about 2000 mg, about 2100 mg, about 2200 mg, about 2300 mg, about 2400 mg, about 2500 mg, about 2600 mg, about 2700 mg, about 2800 mg, about 2900 mg, or about 3000 mg) of the protein product (e.g., anti- PD-L1/TGF Trap (e.g., including a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3, and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1; or a protein product with a first polypeptide that comprises the amino acid sequences of SEQ ID NOs: 35, 36, and 37, and a second polypeptide that comprises the amino acid sequences of SEQ ID NOs: 38, 39, and 40)) to a subject. [0201] In certain embodiments, vials may contain about 61.2 mL of the protein product solution (protein product with a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3, and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1; or a protein product with a first polypeptide that comprises the amino acid sequences of SEQ ID NOs: 35, 36, and 37, and a second polypeptide that comprises the amino acid sequences of SEQ ID NOs: 38, 39, and 40) of about 20 mg/mL to about 50 mg/mL (e.g., about 20 mg/mL, about 25 mg/mL, about 30 mg/mL, about 35 mg/mL, about 40 mg/mL, about 45 mg/mL or about 50 mg/mL) in order to allow an extractable volume of 60 mL for delivering about 1200 mg to about 3000 mg (e.g., about 1200 mg to about 3000 mg, about 1200 mg to about 2900 mg, about 1200 mg to about 2800 mg, about 1200 mg to about 2700 mg, about 1200 mg to about 2600 mg, about 1200 mg to about 2500 mg, about 1200 mg to about 2400 mg, about 1200 mg to about 2300 mg, about 1200 mg to about 2200 mg, about 1200 mg to about 2100 mg, about 1200 mg to about 2000 mg, about 1200 mg to about 1900 mg, about 1200 mg to about 1800 mg, about 1200 mg to about 1700 mg, about 1200 mg to about 1600 mg, about 1200 mg to about 1500 mg, about 1200 mg to about 1400 mg, about 1200 mg to about 1300 mg, about 1300 mg to about 3000 mg, about 1400 mg to about 3000 mg, about 1500 mg to about 3000 mg, about 1600 mg to about 3000 mg, about 1700 mg to about 3000 mg, about 1800 mg to about 3000 mg, about 1900 mg to about 3000 mg, about 2000 mg to about 3000 mg, about 2100 mg to about 3000 mg, about 2200 mg to about 3000 mg, about 2300 mg to about 3000 mg, about 2400 mg to about 3000 mg, about 2500 mg to about 3000 mg, about 2600 mg to about 3000 mg, about 2700 mg to about 3000 mg, about 2800 mg to about 3000 mg, about 2900 mg to about 3000 mg, about 1200 mg, about 1300 mg, about 1400 mg, about 1500 mg, about 1600 mg, about 1700 mg, about 1800 mg, about 1900 mg, about 2000 mg, about 2100 mg, about 2200 mg, about 2300 mg, about 2400 mg, about 2500 mg, about 2600 mg, about 2700 mg, about 2800 mg, about 2900 mg, or about 3000 mg) of the protein product to a treatment naïve subject. [0202] In certain embodiments, the liquid formulation for use in a method of treating cancer or inhibiting tumor growth in a treatment naïve cancer (e.g., NSCLC with high PD-L1 expression) patient of the disclosure may be prepared as a 10 mg/mL concentration solution in combination with a sugar at stabilizing levels. In certain embodiments the liquid formulation may be prepared in an aqueous carrier. In certain embodiments, a stabilizer may be added in an amount no greater than that which may result in a viscosity undesirable or unsuitable for intravenous administration. In certain embodiments, the sugar may be disaccharides, e.g., sucrose. In certain embodiments, the liquid formulation may also include one or more of a buffering agent, a surfactant, and a preservative. [0203] In certain embodiments, the pH of the liquid formulation may be set by addition of a pharmaceutically acceptable acid and/or base. In certain embodiments, the pharmaceutically acceptable acid may be hydrochloric acid. In certain embodiments, the base may be sodium hydroxide. [0204] In addition to aggregation, deamidation is a common product variant of peptides and proteins that may occur during fermentation, harvest/cell clarification, purification, drug substance/drug product storage and during sample analysis. Deamidation is the loss of NH 3 from a protein forming a succinimide intermediate that can undergo hydrolysis. The succinimide intermediate results in a 17 u mass decrease of the parent peptide. The subsequent hydrolysis results in an 18 u mass increase. Isolation of the succinimide intermediate is difficult due to instability under aqueous conditions. As such, deamidation is typically detectable as 1 u mass increase. Deamidation of an asparagine results in either aspartic or isoaspartic acid. The parameters affecting the rate of deamidation include pH, temperature, solvent dielectric constant, ionic strength, primary sequence, local polypeptide conformation and tertiary structure. The amino acid residues adjacent to Asn in the peptide chain affect deamidation rates. Gly and Ser following an Asn in protein sequences results in a higher susceptibility to deamidation. [0205] In certain embodiments, the liquid formulation for use in a method of treating cancer or inhibiting tumor growth in a treatment naïve cancer (e.g., NSCLC with high PD-L1 expression) patient of the present disclosure may be preserved under conditions of pH and humidity to prevent deamination of the protein product. [0206] The aqueous carrier of interest herein is one which is pharmaceutically acceptable (safe and non-toxic for administration to a human) and is useful for the preparation of a liquid formulation. Illustrative carriers include sterile water for injection (SWFI), bacteriostatic water for injection (BWFI), a pH buffered solution (e.g. phosphate-buffered saline), sterile saline solution, Ringer's solution or dextrose solution. [0207] A preservative may be optionally added to the formulations herein to reduce bacterial action. The addition of a preservative may, for example, facilitate the production of a multi-use (multiple-dose) formulation. [0208] Intravenous (IV) formulations may be the preferred administration route in particular instances, such as when a patient is in the hospital after transplantation receiving all drugs via the IV route. In certain embodiments, the liquid formulation is diluted with 0.9% Sodium Chloride solution before administration. In certain embodiments, the diluted drug product for injection is isotonic and suitable for administration by intravenous infusion. [0209] In certain embodiments, a salt or buffer components may be added in an amount of 10 mM - 200 mM. The salts and/or buffers are pharmaceutically acceptable and are derived from various known acids (inorganic and organic) with“base forming” metals or amines. In certain embodiments, the buffer may be phosphate buffer. In certain embodiments, the buffer may be glycinate, carbonate, citrate buffers, in which case, sodium, potassium or ammonium ions can serve as counterion. [0210] A preservative may be optionally added to the formulations herein to reduce bacterial action. The addition of a preservative may, for example, facilitate the production of a multi-use (multiple-dose) formulation. [0211] The aqueous carrier of interest herein is one which is pharmaceutically acceptable (safe and non-toxic for administration to a human) and is useful for the preparation of a liquid formulation. Illustrative carriers include sterile water for injection (SWFI), bacteriostatic water for injection (BWFI), a pH buffered solution (e.g. phosphate-buffered saline), sterile saline solution, Ringer's solution or dextrose solution. [0212] A preservative may be optionally added to the formulations herein to reduce bacterial action. The addition of a preservative may, for example, facilitate the production of a multi-use (multiple-dose) formulation. Method of Treating Cancer or Inhibiting Tumor Growth [0213] In one aspect the present disclosure provides a method of treating cancer or inhibiting tumor growth in a treatment naïve subject in need thereof, the method including administering to the subject a dose of at least 500 mg of a protein including a first polypeptide and a second polypeptide. The first polypeptide includes: (a) at least a variable region of a heavy chain of an antibody that binds to human protein Programmed Death Ligand 1 (PD-L1); and (b) human Transforming Growth Factor Receptor II (TGF RII), or a fragment thereof, capable of binding Transforming Growth Factor (TGF ). The second polypeptide includes at least a variable region of a light chain of an antibody that binds PD-L1, and the heavy chain of the first polypeptide and the light chain of the second polypeptide, when combined, form an antigen binding site that binds PD-L1. [0214] In certain embodiments, the method of treating cancer or inhibiting tumor growth of the present disclosure involves administering to a treatment naïve subject a protein including two peptides in which the first polypeptide includes the amino acid sequence of SEQ ID NO: 3, and the second polypeptide includes the amino acid sequence of SEQ ID NO: 1. In certain embodiments, the protein is an anti-PD-L1/TGF Trap molecule. [0215] In an embodiment, the treatment naïve subject treated in accordance with the methods disclosed herein has not received prior therapy with the the bifunctional protein of the present disclosure (anti-PD-L1/TGF Trap molecule). In some embodiments, the treatment naïve cancer (e.g., NSCLC with high PD-L1 expression) patient to be treated in accordance with the methods of the present disclosure does not have a mutation selected from epidermal growth factor receptor (EGFR) sensitizing (activating) mutation, anaplastic lymphoma kinase (ALK) translocation, ROS1 mutation, and BRAF V600E mutation. [0216] In certain embodiments, the method of treating cancer or inhibiting tumor growth of the present disclosure involves administering to a treatment naïve subject a protein (e.g., an anti-PD-L1/TGF Trap molecule (e.g., including a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3, and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1; or a protein product with a first polypeptide that comprises the amino acid sequences of SEQ ID NOs: 35, 36, and 37, and a second polypeptide that comprises the amino acid sequences of SEQ ID NOs: 38, 39, and 40)) at a dose of about 1200 mg to about 3000 mg (e.g., about 1200 mg to about 3000 mg, about 1200 mg to about 2900 mg, about 1200 mg to about 2800 mg, about 1200 mg to about 2700 mg, about 1200 mg to about 2600 mg, about 1200 mg to about 2500 mg, about 1200 mg to about 2400 mg, about 1200 mg to about 2300 mg, about 1200 mg to about 2200 mg, about 1200 mg to about 2100 mg, about 1200 mg to about 2000 mg, about 1200 mg to about 1900 mg, about 1200 mg to about 1800 mg, about 1200 mg to about 1700 mg, about 1200 mg to about 1600 mg, about 1200 mg to about 1500 mg, about 1200 mg to about 1400 mg, about 1200 mg to about 1300 mg, about 1300 mg to about 3000 mg, about 1400 mg to about 3000 mg, about 1500 mg to about 3000 mg, about 1600 mg to about 3000 mg, about 1700 mg to about 3000 mg, about 1800 mg to about 3000 mg, about 1900 mg to about 3000 mg, about 2000 mg to about 3000 mg, about 2100 mg to about 3000 mg, about 2200 mg to about 3000 mg, about 2300 mg to about 3000 mg, about 2400 mg to about 3000 mg, about 2500 mg to about 3000 mg, about 2600 mg to about 3000 mg, about 2700 mg to about 3000 mg, about 2800 mg to about 3000 mg, about 2900 mg to about 3000 mg, about 1200 mg, about 1300 mg, about 1400 mg, about 1500 mg, about 1600 mg, about 1700 mg, about 1800 mg, about 1900 mg, about 2000 mg, about 2100 mg, about 2200 mg, about 2300 mg, about 2400 mg, about 2500 mg, about 2600 mg, about 2700 mg, about 2800 mg, about 2900 mg, or about 3000 mg). In certain embodiments, about 1200 mg of anti-PD-L1/TGF Trap molecule is administered to a treatment naïve subject once every two weeks. In certain embodiments, about 2400 mg of anti-PD-L1/TGF Trap molecule is administered to a treatment naïve subject once every three weeks. In certain embodiments, about 1200 mg of a protein product with a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3 and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1 is administered to a treatment naïve subject once every two weeks. In certain embodiments, about 2400 mg of a protein product with a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3 and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1 is administered to a treatment naïve subject once every three weeks. In certain embodiments, about 2400 mg of a protein product with a first polypeptide that comprises the amino acid sequences of SEQ ID NOs: 35, 36, and 37, and a second polypeptide that comprises the amino acid sequences of SEQ ID NOs: 38, 39, and 40 is administered to a treatment naïve subject once every three weeks. [0217] In certain embodiments, the dose administered to a treatment naïve subject may be about 500 mg, about 525 mg, about 550 mg, about 575 mg, about 600 mg, about 625 mg, about 650 mg, about 675 mg, about 700 mg, about 725 mg, about 750 mg, about 775 mg, about 800 mg, about 825 mg, about 850 mg, about 875 mg, about 900 mg, about 925 mg, about 950 mg, about 975 mg, about 1000 mg, about 1025 mg, about 1050 mg, about 1075 mg, about 1100 mg, about 1125 mg, about 1150 mg, about 1175 mg, about 1200 mg, about 1225 mg, about 1250 mg, about 1275 mg, about 1300 mg, about 1325 mg, about 1350 mg, about 1375 mg, about 1400 mg, about 1425 mg, about 1450 mg, about 1475 mg, about 1500 mg, about 1525 mg, about 1550 mg, about 1575 mg, about 1600 mg, about 1625 mg, about 1650 mg, about 1675 mg, about 1700 mg, about 1725 mg, about 1750 mg, about 1775 mg, about 1800 mg, about 1825 mg, about 1850 mg, 1875 mg, about 1900 mg, about 1925 mg, about 1950 mg, about 1975 mg, about 2000 mg, about 2025 mg, about 2050 mg, about 2075 mg, about 2100 mg, about 2125 mg, about 2150 mg, about 2175 mg, about 2200 mg, about 2225 mg, about 2250 mg, about 2275 mg, about 2300 mg, about 2325 mg, about 2350 mg, about 2375 mg, or about 2400 mg. [0218] In certain embodiments, the dose administered to a treatment naïve subject may be administered once every two weeks. In certain embodiments, the dose administered to a treatment naïve subject may be administered once every three weeks. In certain embodiments, the protein may be administered by intravenous administration, e.g., with a prefilled bag, a prefilled pen, or a prefilled syringes. In certain embodiments, the protein is administered intravenously from a 250 ml saline bag, and the intravenous infusion may be for about one hour (e.g., 50 to 80 minutes). In certain embodiments, the bag is connected to a channel comprising a tube and/or a needle. [0219] In some embodiments, the NSCLC exhibits squamous or non-squamous histology. For example, in an embodiment, the method treats squamous NSCLC. In some embodiments, the method treats non-squamous NSCLC. [0220] In certain embodiments, treatment naïve subjects or patients with advanced NSCLC (e.g., NSCLC (e.g., squamous or non-squamous NSCLC) with high PD-L1 expression) are treated by intravenously administering at least 500 mg (e.g., about 500 mg, about 600 mg, about 700 mg, about 800 mg, about 900 mg, about 1000 mg, about 1100 mg, about 1200 mg, about 1300 mg, about 1400 mg, about 1500 mg, about 1600 mg, about 1700 mg, about 1800 mg, about 1900 mg, about 2000 mg, about 2100 mg, about 2200 mg, about 2300 mg, about 2400 mg, or more) of anti-PD-L1/TGF Trap, which includes a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3, and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1. In certain embodiments, treatment naïve subjects or patients with advanced NSCLC (e.g., NSCLC (e.g., squamous or non-squamous NSCLC) with high PD-L1 expression) are treated by intravenously administering at least 500 mg (e.g., about 500 mg, about 600 mg, about 700 mg, about 800 mg, about 900 mg, about 1000 mg, about 1100 mg, about 1200 mg, about 1300 mg, about 1400 mg, about 1500 mg, about 1600 mg, about 1700 mg, about 1800 mg, about 1900 mg, about 2000 mg, about 2100 mg, about 2200 mg, about 2300 mg, about 2400 mg, or more) of anti-PD-L1/TGF Trap, which includes a first polypeptide that comprises the amino acid sequences of SEQ ID NOs: 35, 36, and 37, and a second polypeptide that comprises the amino acid sequences of SEQ ID NOs: 38, 39, and 40. [0221] In certain embodiments, treatment naïve subjects or patients with advanced NSCLC (e.g., NSCLC (e.g., squamous or non-squamous NSCLC) with high PD-L1 expression) are treated by intravenously administering about 1200 mg– about 2400 mg (e.g., about 1200 mg to about 2400 mg, about 1200 mg to about 2300 mg, about 1200 mg to about 2200 mg, about 1200 mg to about 2100 mg, about 1200 mg to about 2000 mg, about 1200 mg to about 1900 mg, about 1200 mg to about 1800 mg, about 1200 mg to about 1700 mg, about 1200 mg to about 1600 mg, about 1200 mg to about 1500 mg, about 1200 mg to about 1400 mg, about 1200 mg to about 1300 mg, about 1300 mg to about 2400 mg, about 1400 mg to about 2400 mg, about 1500 mg to about 2400 mg, about 1600 mg to about 2400 mg, about 1700 mg to about 2400 mg, about 1800 mg to about 2400 mg, about 1900 mg to about 2400 mg, about 2000 mg to about 2400 mg, about 2100 mg to about 2400 mg, about 2200 mg to about 2400 mg, or about 2300 mg to about 2400 mg) of anti-PD-L1/TGF Trap, which includes a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3, and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1. In certain embodiments, treatment naïve subjects or patients with advanced NSCLC (e.g., NSCLC (e.g., squamous or non- squamous NSCLC) with high PD-L1 expression) are treated by intravenously administering about 1200 mg– about 2400 mg (e.g., about 1200 mg to about 2400 mg, about 1200 mg to about 2300 mg, about 1200 mg to about 2200 mg, about 1200 mg to about 2100 mg, about 1200 mg to about 2000 mg, about 1200 mg to about 1900 mg, about 1200 mg to about 1800 mg, about 1200 mg to about 1700 mg, about 1200 mg to about 1600 mg, about 1200 mg to about 1500 mg, about 1200 mg to about 1400 mg, about 1200 mg to about 1300 mg, about 1300 mg to about 2400 mg, about 1400 mg to about 2400 mg, about 1500 mg to about 2400 mg, about 1600 mg to about 2400 mg, about 1700 mg to about 2400 mg, about 1800 mg to about 2400 mg, about 1900 mg to about 2400 mg, about 2000 mg to about 2400 mg, about 2100 mg to about 2400 mg, about 2200 mg to about 2400 mg, or about 2300 mg to about 2400 mg) of anti-PD-L1/TGF Trap, which includes a first polypeptide that comprises the amino acid sequences of SEQ ID NOs: 35, 36, and 37, and a second polypeptide that comprises the amino acid sequences of SEQ ID NOs: 38, 39, and 40. [0222] In some embodiments, treatment naïve subjects or patients with advanced NSCLC (e.g., NSCLC (e.g., squamous or non-squamous NSCLC) with high PD-L1 expression) are treated by intravenously administering anti-PD-L1/TGF Trap at a dose of about 1200 mg once every 2 weeks. In some embodiments, treatment naïve subjects or patients with advanced NSCLC (e.g., NSCLC (e.g., squamous or non-squamous NSCLC) with high PD-L1 expression) are treated by intravenously administering anti-PD-L1/TGF Trap at a dose of about 2400 mg once every 3 weeks. [0223] In certain embodiments, the cancer to be treated is PD-L1 positive. For example, in certain embodiments, the cancer to be treated exhibits high PD-L1 expression (“high PD-L1” or “PD-L1 high”). [0224] Methods of detecting a biomarker, such as PD-L1 for example, on a cancer or tumor, are routine in the art and are contemplated herein. Non-limiting examples include

immunohistochemistry, immunofluorescence and flourescence activated cell sorting (FACS). In some embodiments, treatment naïve subjects or patients with PD-L1 high, advanced NSCLC (e.g., squamous or non-squamous advanced NSCLC) are treated by intravenously

administering anti-PD-L1/TGF Trap at a dose of at least 500 mg. In some embodiments, treatment naïve subjects or patients with PD-L1 high, advanced NSCLC (e.g., squamous or non-squamous advanced NSCLC) are treated by intravenously administering anti-PD-L1/TGF Trap at a dose of about 1200 mg once every 2 weeks. In some embodiments, treatment naïve subjects or patients with with PD-L1 high, advanced NSCLC (e.g., squamous or non-squamous advanced NSCLC) are treated by intravenously administering anti-PD-L1/TGF Trap at a dose of about 2400 mg once every 3 weeks. [0225] In some embodiments, the treatment naïve subject or patient to be treated does not have a mutation selected from EGFR sensitizing mutation, ALK translocation, ROS1 mutation, and BRAF V600E mutation. For example, in some embodiments, treatment naïve subjects or patients with PD-L1 high, advanced NSCLC (e.g., squamous or non-squamous advanced NSCLC), but who do not have a mutation selected from EGFR sensitizing mutation, ALK translocation, ROS1 mutation, and BRAF V600E mutation, are treated by intravenously administering anti-PD-L1/TGF Trap at a dose of at least 500 mg (e.g., about 500 mg, about 600 mg, about 700 mg, about 800 mg, about 900 mg, about 1000 mg, about 1100 mg, about 1200 mg, about 1300 mg, about 1400 mg, about 1500 mg, about 1600 mg, about 1700 mg, about 1800 mg, about 1900 mg, about 2000 mg, about 2100 mg, about 2200 mg, about 2300 mg, about 2400 mg, or more). In some embodiments, treatment naïve subjects or patients with PD-L1 high, advanced NSCLC (e.g., squamous or non-squamous advanced NSCLC) who do not have a mutation selected from EGFR sensitizing mutation, ALK translocation, ROS1 mutation, and BRAF V600E mutation are treated by intravenously administering anti-PD- L1/TGF Trap at a dose of about 1200 mg once every 2 weeks. In some embodiments, treatment naïve subjects or patients with PD-L1 high, advanced NSCLC (e.g., squamous or non-squamous NSCLC) who do not have a mutation selected from EGFR sensitizing mutation, ALK translocation, ROS1 mutation, and BRAF V600E mutation are treated by intravenously administering anti-PD-L1/TGF Trap at a dose of about 2400 mg once every 3 weeks. [0226] In some embodiments, the methods of treatment disclosed herein result in a disease response or improved survival of the subject or patient. In some embodiments for example, the disease response may be a complete response, a partial response, or a stable disease. In some embodiments for example, the improved survival could be progression-free survival (PFS) or overall survival. In some embodiments, improvement (e.g., in PFS) is determined relative to a period prior to initiation of treatment with an anti-PD-L1/TGF Trap of the present disclosure. Methods of determining disease response (e.g, complete response, partial response, or stable disease) and patient survival (e.g, PFS, overall survival) for cancer or tumor therapy are routine in ther art and are contemplated herein. In some embodiments, disease response is evaluated according to RECIST 1.1 after subjecting the treated patient to contrast-enhanced computed tomography (CT) or magnetic resonance imaging (MRI) of the affected area

(e.g.,chest/abdomen and pelvis covering the area from the superior extent of the thoracic inlet to the symphysis pubis). Delivery Device [0227] In one aspect, the present disclosure provides a drug delivery device for use in a method of treating cancer or inhibiting tumor growth in a treatment naïve cancer (e.g., NSCLC with high PD-L1 expression) patient, wherein the device includes a formulation comprising about 500 mg– about 3000 mg of a protein including a first polypeptide and a second polypeptide, the first polypeptide includes: (a) at least a variable region of a heavy chain of an antibody that binds to human protein Programmed Death Ligand 1 (PD-L1); and (b) human Transforming Growth Factor Receptor II (TGF RII), or a fragment thereof, capable of binding Transforming Growth Factor (TGF ), the second polypeptide includes at least a variable region of a light chain of an antibody that binds PD-L1, and the heavy chain of the first polypeptide and the light chain of the second polypeptide, when combined, form an antigen binding site that binds PD-L1. [0228] In certain embodiments, the device may be a bag, a pen, or a syringe. In certain embodiments, the bag may be connected to a channel comprising a tube and/or a needle. [0229] In certain embodiments of the present disclosure, the drug delivery device for use in a method of treating cancer or inhibiting tumor growth in a treatment naïve cancer (e.g., NSCLC with high PD-L1 expression) patient may include about 500 mg to about 3000 mg (e.g., about 500 mg to about 3000 mg, about 500 mg to about 2900 mg, about 500 mg to about 2800 mg, about 500 mg to about 2700 mg, about 500 mg to about 2600 mg, about 500 mg to about 2500 mg, about 500 mg to about 2400 mg, about 500 mg to about 2300 mg, about 500 mg to about 2200 mg, about 500 mg to about 2100 mg, about 500 mg to about 2000 mg, about 500 mg to about 1900 mg, about 500 mg to about 1800 mg, about 500 mg to about 1700 mg, about 500 mg to about 1600 mg, about 500 mg to about 1500 mg, about 500 mg to about 1400 mg, about 500 mg to about 1300 mg, about 500 mg to about 1200 mg, about 500 mg to about 1100 mg, about 500 mg to about 1000 mg, about 500 mg to about 900 mg, about 500 mg to about 800 mg, about 500 mg to about 700 mg, about 500 mg to about 600 mg, about 600 mg to about 3000 mg, about 700 mg to about 3000 mg, about 800 mg to about 3000 mg, about 900 mg to about 3000 mg, about 1000 mg to about 3000 mg, about 1100 mg to about 3000 mg, about 1200 mg to about 3000 mg, about 1300 mg to about 3000 mg, about 1400 mg to about 3000 mg, about 1500 mg to about 3000 mg, about 1600 mg to about 3000 mg, about 1700 mg to about 3000 mg, about 1800 mg to about 3000 mg, about 1900 mg to about 3000 mg, about 2000 mg to about 3000 mg, about 2100 mg to about 3000 mg, about 2200 mg to about 3000 mg, about 2300 mg to about 3000 mg, about 2400 mg to about 3000 mg, about 2500 mg to about 3000 mg, about 2600 mg to about 3000 mg, about 2700 mg to about 3000 mg, about 2800 mg to about 3000 mg, or about 2900 mg to about 3000 mg) of a protein of the present disclosure (e.g., anti-PD-L1/TGF Trap, which includes a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3, and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1; or a protein product with a first polypeptide that comprises the amino acid sequences of SEQ ID NOs: 35, 36, and 37, and a second polypeptide that comprises the amino acid sequences of SEQ ID NOs: 38, 39, and 40). In certain embodiments, the drug delivery device may include about 500 to about 1200 mg dose of a protein of the present disclosure (e.g., anti-PD-L1/TGF Trap, which includes a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3, and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1). In certain embodiments, the drug delivery device may include about 500 mg dose of the protein of the present disclosure (e.g., anti-PD-L1/TGF Trap, which includes a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3, and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1; or a protein product with a first polypeptide that comprises the amino acid sequences of SEQ ID NOs: 35, 36, and 37, and a second polypeptide that comprises the amino acid sequences of SEQ ID NOs: 38, 39, and 40). [0230] In certain embodiments, the drug delivery device includes about 1200 mg dose of a protein of the present disclosure (e.g., anti-PD-L1/TGF Trap, which includes a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3, and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1; or a protein product with a first polypeptide that comprises the amino acid sequences of SEQ ID NOs: 35, 36, and 37, and a second polypeptide that comprises the amino acid sequences of SEQ ID NOs: 38, 39, and 40). In certain embodiments, the drug delivery device for use in a method of treating cancer or inhibiting tumor growth in a treatment naïve cancer (e.g., NSCLC with high PD-L1 expression) patient includes about 2400 mg dose of a protein of the present disclosure (e.g., anti-PD- L1/TGF Trap, which includes a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3, and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1; or a protein product with a first polypeptide that comprises the amino acid sequences of SEQ ID NOs: 35, 36, and 37, and a second polypeptide that comprises the amino acid sequences of SEQ ID NOs: 38, 39, and 40). In certain embodiments, the drug delivery device for use in a method of treating cancer or inhibiting tumor growth in a treatment naïve cancer (e.g., NSCLC with high PD-L1 expression) patient includes about 1200 mg or about 2400 mg dose of the protein product with a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3, and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1; or a protein product with a first polypeptide that comprises the amino acid sequences of SEQ ID NOs: 35, 36, and 37, and a second polypeptide that comprises the amino acid sequences of SEQ ID NOs: 38, 39, and 40. [0231] In certain embodiments, the drug delivery device for use in a method of treating cancer or inhibiting tumor growth in a treatment naïve cancer (e.g., NSCLC with high PD-L1 expression) patient includes about 1200 mg dose of the protein of the present disclosure (e.g., anti-PD-L1/TGF Trap (e.g., including a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3, and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1; or a protein product with a first polypeptide that comprises the amino acid sequences of SEQ ID NOs: 35, 36, and 37, and a second polypeptide that comprises the amino acid sequences of SEQ ID NOs: 38, 39, and 40)). In certain embodiments, the drug delivery device for use in a method of treating cancer or inhibiting tumor growth in a treatment naïve cancer (e.g., NSCLC with high PD-L1 expression) patient includes about 2400 mg dose of the protein of the present disclosure (e.g., anti-PD-L1/TGF Trap (e.g., including a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3, and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1; or a protein product with a first polypeptide that comprises the amino acid sequences of SEQ ID NOs: 35, 36, and 37, and a second polypeptide that comprises the amino acid sequences of SEQ ID NOs: 38, 39, and 40)). In certain embodiments, the drug delivery device for use in a method of treating cancer or inhibiting tumor growth in a treatment naïve cancer (e.g., NSCLC with high PD-L1 expression) patient may include about 500 mg, about 525 mg, about 550 mg, about 575 mg, about 600 mg, about 625 mg, about 650 mg, about 675 mg, about 700 mg, about 725 mg, about 750 mg, about 775 mg, about 800 mg, about 825 mg, about 850 mg, about 875 mg, about 900 mg, about 925 mg, about 950 mg, about 975 mg, about 1000 mg, about 1025 mg, about 1050 mg, about 1075 mg, about 1100 mg, about 1125 mg, about 1150 mg, about 1175 mg, about 1200 mg, about 1225 mg, about 1250 mg, about 1275 mg, about 1300 mg, about 1325 mg, about 1350 mg, about 1375 mg, about 1400 mg, about 1425 mg, about 1450 mg, about 1475 mg, about 1500 mg, about 1525 mg, about 1550 mg, about 1575 mg, about 1600 mg, about 1625 mg, about 1650 mg, about 1675 mg, about 1700 mg, about 1725 mg, about 1750 mg, about 1775 mg, about 1800 mg, about 1825 mg, about 1850 mg, about 1875 mg, about 1900 mg, about 1925 mg, about 1950 mg, about 1975 mg, about 2000 mg, about 2025 mg, about 2050 mg, about 2075 mg, about 2100 mg, about 2125 mg, about 2150 mg, about 2175 mg, about 2200 mg, about 2225 mg, about 2250 mg, about 2275 mg, about 2300 mg, about 2325 mg, about 2350 mg, about 2375 mg, or about 2400 mg of the protein of the present disclosure (e.g., anti-PD- L1/TGF Trap, e.g., a protein product with a first polypeptide that comprises the amino acid sequences of SEQ ID NOs: 35, 36, and 37, and a second polypeptide that comprises the amino acid sequences of SEQ ID NOs: 38, 39, and 40). Protein Production [0232] The antibody-cytokine Trap proteins are generally produced recombinantly, using mammalian cells containing a nucleic acid engineered to express the protein. Although one example of a suitable cell line and protein production method is described in Examples 1 and 2 of US 20150225483 A1, a wide variety of suitable vectors, cell lines and protein production methods have been used to produce antibody-based biopharmaceuticals and could be used in the synthesis of these antibody-cytokine Trap proteins. Therapeutic Indications [0233] The anti-PD-L1/TGF Trap proteins described in the application (e.g., including a first polypeptide that includes the amino acid sequence of SEQ ID NO: 3, and a second polypeptide that includes the amino acid sequence of SEQ ID NO: 1), as well as the disclosed intravenous drug delivery formulations and delivery devices comprising said anti-PD-L1/TGF Trap proteins, can be used to treat cancer or reduce tumor growth in a treatment naïve patient. Exemplary cancers include non-small cell lung cancer (NSCLC), melanoma, pancreatic cancer, colorectal cancer (e.g., pretreated colorectal cancer (CRC)), ovarian cancer, glioblastoma, gastric cancer (e.g., pretreated recurrent or refractory unresectable Stage IV gastric cancer), biliary tract cancer, esophageal cancer (squamous cell carcinoma or adenocarcinoma), adenoma of the head or the neck, and squamous carcinoma of the head or the neck. In a specific embodiment, the treated cancer is advanced NSCLC (e.g., squamous or non-squamous advanced NSCLC). [0234] The cancer or tumor to be treated with an anti-PD-L1/ TGF Trap may be selected based on the expression or elevated expression of PD-L1 and/or TGF in the tumor, the correlation of their expression levels with prognosis or disease progression, and preclinical and clinical experience on the sensitivity of the tumor to treatments targeting PD-L1 and TGF . Such cancers or tumors include but are not limited to colorectal, breast, ovarian, pancreatic, gastric, prostate, renal, cervical, bladder, head and neck, liver, non-small cell lung cancer, advanced non-small cell lung cancer, melanoma, Merkel cell carcinoma, and mesothelioma. For example, in a specific embodiment, a treatment naïve patient with a PD-L1 positive (e.g., PD-L1 high) advanced NSCLC is treated in accordance with the methods of the present disclosure. [0235] In some embodiments, the treatment naïve cancer (e.g., advanced NSCLC (i.e., metastatic NSCLC)) with high PD-L1 expression) patient to be treated in accordance with the methods of the present disclosure does not have a mutation selected from epidermal growth factor receptor (EGFR) sensitizing (activating) mutation, anaplastic lymphoma kinase (ALK) translocation, ROS1 mutation, and BRAF V600E mutation. In some embodiments, the treatment naïve cancer (e.g., advanced NSCLC (i.e., metastatic NSCLC)) with high PD-L1 expression) patient to be treated in accordance with the methods of the present disclosure does not have epidermal growth factor receptor (EGFR) sensitizing (activating) mutation. In some embodiments, the treatment naïve cancer (e.g., advanced NSCLC (i.e., metastatic NSCLC)) with high PD-L1 expression) patient to be treated in accordance with the methods of the present disclosure does not have anaplastic lymphoma kinase (ALK) translocation. In some embodiments, the treatment naïve cancer (e.g., advanced NSCLC (i.e., metastatic NSCLC)) with high PD-L1 expression) patient to be treated in accordance with the methods of the present disclosure does not have ROS1 mutation. In some embodiments, the treatment naïve cancer (e.g., advanced NSCLC (i.e., metastatic NSCLC)) with high PD-L1 expression) patient to be treated in accordance with the methods of the present disclosure does not have BRAF V600E mutation. EXAMPLES

[0236] The disclosure now being generally described, will be more readily understood by reference to the following examples, which are included merely for purposes of illustration of certain aspects and embodiments of the present disclosure, and are not intended to limit the scope of the disclosure in any way. EXAMPLE 1: Packaging of Intravenous Drug Formulation [0237] The formulation of anti-PD-L1/TGF Trap is prepared as a lyophilized formulation or a liquid formulation. For preparing the lyophilized formulation, freeze-dried anti-PD- L1/TGF Trap is sterilized and stored in single-use glass vials. Several such glass vials are then packaged in a kit for delivering a specific body weight independent dose to a subject diagnosed with a cancer or a tumor. Depending on the dose requirement, the kit contains 12-60 vials. Alternatively, the formulation is prepared and packaged as a liquid formulation and stored as 250 mg/vial to 1000 mg/vial. For example, the formulation is a liquid formulation and stored as 600 mg/vial, or stored as 250 mg/vial. In another example, the anti-PD-L1/TGF Trap is formulated as a 10mg/mL solution and is supplied in USP/Ph Eur type I vials filled to allow an extractable volume of 60 mL (600 mg/60 mL) and closed with rubber stoppers in serum format complying with USP and Ph Eur with an aluminum crimpseal closure. [0238] A subject diagnosed with advanced NSCLC is intravenously administered a formulation containing 500 mg to 2400 mg of anti-PD-L1/TGF Trap. For example, the subject is intravenously administered 1200 mg of anti-PD-L1/TGF Trap once in two weeks or 2400 mg of anti-PD-L1/TGF Trap once in three weeks. The intravenous administration is from a saline bag. The amount of the anti-PD-L1/TGF Trap administered to a subject is independent of the subject’s body weight. EXAMPLE 2: BW-Independent Dosing Regimen of a Treatment Naïve, Advanced NSCLC Patient Cohort [0239] In one exemplary embodiment, the BW-independent dose of 1200 mg is administered to cancer patients with advanced non-small cell lung cancer (NSCLC) once every two weeks. The administration is performed intravenously for about an hour (-10 minutes / +20 minutes, i.e., 50 minutes to 80 minutes). In one exemplary embodiment, the BW- independent dose of 2400 mg is administered to cancer patients with advanced non-small cell lung cancer (NSCLC) once every three weeks. The administration is performed intravenously for about an hour (-10 minutes / +20 minutes, i.e., 50 minutes to 80 minutes). In order to mitigate potential infusion-related reactions, premedication with an antihistamine and with paracetamol (acetaminophen) (for example, 25-50 mg diphenhydramine and 500-650 mg paracetamol [acetaminophen] IV or oral equivalent) approximately 30 to 60 minutes prior to each anti-PD-L1/TGF Trap dose is administered for the first 2 infusions. If Grade 2 infusion reactions are observed during the first two infusions, premedication is not stopped. Steroids as premedication are not permitted. [0240] The following describes the inclusion criteria for patients used in this example. Patients: - are 18 years

- Histologically confirmed diagnosis of advanced NSCLC with high PD-L1 expression on tumor cells

- have not received prior systemic therapy treatment for their advanced NSCLC (completion of treatment with cytotoxic chemotherapy, biological therapy, and/or radiation as part of neoadjuvant/adjuvant therapy is allowed as long as therapy was completed at least 6 months prior to the diagnosis of metastatic disease)

- have measurable disease based on RECIST 1.1 (see Eisenhauer et al., EJC.

2009; 45:228-247) - have a life expectancy of at least 3 months

- have archival tumor material (less than 6 months old) or produce fresh biopsies to determine PD-L1 expression

- have Eastern Cooperative Oncology Group Performance Status (ECOG PS) of 0 to 1

- have adequate organ function and life expectancy 3 months

- have adequate hematological function defined by absolute neutrophil count (ANC) 1.5 × 10 9 /L, platelet count 100 × 10 9 /L, and Hgb 9 g/ dL

- have adequate hepatic function defined by a total bilirubin level the upper limit of normal (ULN), an AST level 1.5 × ULN, and an ALT level 1.5 × ULN. For participants with liver involvement in their tumor, aspartate aminotransferase (AST) 5.0 × ULN, alanine aminotransferase (ALT) 5.0 × ULN, and bilirubin 3.0 × ULN is acceptable

- have adequate renal function defined by creatinine 1.5 × ULN or a calculated creatinine clearance > 30 mL/min; and

- have adequate coagulation function defined as international normalized ratio (INR) or prothrombin time (PT) 1.5× ULN unless the participant is receiving anticoagulant therapy, and activated partial thromboplastin time (aPTT) 1.5× ULN unless the participant is receiving anticoagulant therapy. [0241] Selected patients do not have active tuberculosis or an autoimmune disease that might deteriorate when receiving an immunostimulatory agent. Selected patients have not received prior therapy with an anti-PD-1, anti-PD-L1, anti-PD-L2, anti-CD137, or anti- Cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) antibody (including ipilimumab), or any other antibody or drug specifically targeting T-cell co-stimulation or checkpoint pathways. EXAMPLE 3: Treatment of Advanced NSCLC Patients with anti-PD-L1/TGF Trap [0242] Objective: The purpose of this study is to evaluate whether anti-PD-L1/TGF Trap improves progression-free survival (PFS) time and/or best overall response (BOR) as a first- line (1L) treatment for patients with, advanced non-small cell lung cancer (NSCLC) with PD- L1 tumor expression. The rationale for using anti-PD-L1/TGF Trap in this NSCLC patient cohort is that anti-PD-L1/TGF Trap targets PD-L1 and TGF , two major mechanisms of immunosuppression in the tumor microenvironment. Preclinical data suggest that anti-PD- L1/TGF Trap strongly enhances antitumor activity and prolongs survival in mouse tumor models above the effect of either the anti PD-L1 antibody avelumab or the TGF Trap control alone. Thus, simultaneous neutralization of TGF- , a molecule known to inhibit tumor immune activation, might stimulate clinical response in patients. [0243] Study Design: This study evaluates disease response and survival primary endpoints to assess clinical benefit of an anti-PD-L1/TGF Trap as first line treatment for patients with advanced NSCLC with high PD-L1-tumor expression. For purposes of this study, PD-L1 high is 80% PD-L1 positive tumor cells as determined by the Dako 73-10 assay. Patients with tumor proportion score (TPS) 50% as determined by the PD-L1 Dako IHC 22C3 PharmDx assay performed according to local laboratory regulations prior to study enrollment are also eligible. Both Dako 73-10 assay and Dako IHC 22C3 PharmDx assay select a similar patient population at their respective cutoffs. Approximately 300 patients who have not received previous treatment for their advanced NSCLC (patients are treatment naïve) are enrolled in this study. The patients in this study meet the inclusion criteria of patients described in Example 2, have not received prior systemic therapy treatment for their advanced NSCLC (patients are treatment naïve), and do not have epidermal growth factor receptor (EGFR) sensitizing (activating) mutation, anaplastic lymphoma kinase (ALK) translocation, ROS1 mutation, or BRAF V600E mutation, where targeted therapy is locally approved. The patients are stratified according to tumor histology (squamous versus nonsquamous) and smoking history as follows: squamous history, nonsquamous history and never smoked, nonsquamous history and ever smoker. The patients are intravenously administered an anti-PD-L1/TGF Trap dose of 1200 mg once every two weeks, or 2400 mg once every three weeks. Treatment is continued until confirmed progressive disease (PD) per Response Evaluation Criteria in Solid Tumors version 1.1 (RECIST 1.1), unacceptable toxicity, or for up to 24 months. In the case of PD, treatment may continue past the initial determination of PD or confirmed PD if the patient's Eastern Cooperative Oncology Group Performance Status (ECOG PS) remains stable, and if the participant will benefit from continued treatment. Patients who experience stable disease (SD), partial response (PR), or complete response (CR) will continue treatment until the end of 24 months, although additional treatment is be possible. [0244] Throughout treatment, safety is assessed through the recording, reporting and analysis of baseline medical conditions, adverse events (AEs), physical examination findings, including vital signs, ECOG performance status, and laboratory tests. [0245] Efficacy Assessments: Tumor response to anti-PD-L1/TGF Trap is assessed by CT scan or MRI. Scans performed at baseline are repeated at subsequent visits. In general, lesions detected at baseline are followed using the same imaging methodology and preferably the same imaging equipment at subsequent tumor evaluation visits. Skin metastasis can be used as target lesions according to RECIST 1.1 using measurements by caliper, if they fulfill RECIST 1.1 for target lesions. [0246] Results: Objective tumor response is evaluated by the overall response rate (ORR), defined as the number of participants having reached a best overall response (BOR) of complete response (CR) or partial response (PR) divided by the number of participants in the analysis population. Progression-free survival is defined as the time from randomization to the date of the first documentation of objective progression of disease (PD) as assessed according to RECIST 1.1 or death due to any cause, whichever occurs first. It is contemplated that treatment with anti-PD-L1/TGF Trap results in initial clinical activity in treatment naïve, advanced NSCLC patients with PD-L1 high status. Tumor biomarkers are evaluated before and after treatment from blood and tumor samples obtained from the patients who are administered the anti-PD-L1/TGF Trap. Treated patients exhibit disease response (e.g., partial response, complete response, stable disease) and/or improved survival (e.g., progression-free survival and/or overall survival). [0247] In summary, the examples described above provide treatment regimens involving anti-PD-L1/TGF Trap for treating advanced NSCLC with high PD-L1 expression in treatment naïve patients.

SEQUENCES SEQ ID NO: 1 Peptide sequence of the secreted anti-PD-L1 lambda light chain

SEQ ID NO: 2 Peptide sequence of the secreted H chain of anti-PDL1

SEQ ID NO: 3 Peptide sequence of the secreted H chain of anti-PDL1/TGF Trap

SEQ ID NO: 4 DNA sequence from the translation initiation codon to the translation stop codon of the anti- PD-L1 lambda light chain (the leader sequence preceding the VL is the signal peptide from urokinase plasminogen activator)

SEQ ID NO: 5

SEQ ID NO: 6 Polypeptide sequence of the secreted lambda light chain of anti-PD-L1(mut)/ TGF Trap, with mutations A31G, D52E, R99Y

SEQ ID NO: 7 Polypeptide sequence of the secreted heavy chain of anti-PD-L1(mut)/ TGF Trap

SEQ ID NO: 8 Human TGF RII Isoform A Precursor Polypeptide (NCBI RefSeq Accession No: NP_001020018)

SEQ ID NO: 9 Human TGF RII Isoform B Precursor Polypeptide (NCBI RefSeq Accession No: NP_003233)

SEQ ID NO: 10 A Human TGF RII Isoform B Extracellular Domain Polypeptide

SEQ ID NO: 11 (Gly 4 Ser) 4 Gly linker

SEQ ID NO: 12 Polypeptide sequence of the secreted heavy chain variable region of anti-PD-L1 antibody MPDL3289A

SEQ ID NO: 13 Polypeptide sequence of the secreted light chain variable region of anti-PD-L1 antibody MPDL3289A

SEQ ID NO: 14 Polypeptide sequence of the secreted heavy chain variable region of anti-PD-L1 antibody YW243.55S70

SEQ ID NO: 50 A Truncated Human TGF RII Isoform B Extracellular Domain Polypeptide

SEQ ID NO: 51 A Truncated Human TGF RII Isoform B Extracellular Domain Polypeptide

SEQ ID NO: 52 A Truncated Human TGF RII Isoform B Extracellular Domain Polypeptide

SEQ ID NO: 53 A Truncated Human TGF RII Isoform B Extracellular Domain Polypeptide

SEQ ID NO: 54 A Mutated Human TGF RII Isoform B Extracellular Domain Polypeptide

SEQ ID NO: 55 Polypeptide sequence of the heavy chain variable region of anti-PD-L1 antibody SEQ ID NO: 56 Polypeptide sequence of the light chain variable region of anti-PD-L1 antibody

SEQ ID NO: 57 Polypeptide sequence of the heavy chain variable region of anti-PD-L1 antibody

SEQ ID NO: 58 Polypeptide sequence of the light chain variable region of anti-PD-L1 antibody

SEQ ID NO: 59 Polypeptide sequence of the heavy chain of anti-PD-L1 antibody

SEQ ID NO: 60 Polypeptide sequence of the light chain of anti-PD-L1 antibody

SEQ ID NO: 61 Polypeptide sequence of the heavy chain of anti-PD-L1 antibody

SEQ ID NO: 62 Polypeptide sequence of the light chain of anti-PD-L1 antibody INCORPORATION BY REFERENCE [0248] The entire disclosure of each of the patent documents and scientific articles referred to herein is incorporated by reference for all purposes. EQUIVALENTS [0249] The disclosure may be embodied in other specific forms without departing from the spirit or essential characteristics thereof. The foregoing embodiments are therefore to be considered in all respects illustrative rather than limiting the disclosure described herein. Various structural elements of the different embodiments and various disclosed method steps may be utilized in various combinations and permutations, and all such variants are to be considered forms of the disclosure. Scope of the disclosure is thus indicated by the appended claims rather than by the foregoing description, and all changes that come within the meaning and range of equivalency of the claims are intended to be embraced therein.