The instant application contains a Sequence Listing which has been submitted in ASCII format via EFS-Web and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Jul. 3, 2020, is named “27522-0225PCT Sequence Listing_ST25.txt” and is 34 kilobytes in size.
The present disclosure describes compositions and methods directed to treating cancer where the compositions include utilizing oncolytic viruses, such as Sindbis virus, and antibodies directed against a co-stimulatory molecule or to an immunesystem agonist molecule, such as OX-40 and 4-1BB (CD137).
Immune checkpoint modulation has shown remarkable promise in treating cancer. Although, high response rates with immune checkpoint blockade have been documented in patients with highly immunogenic tumors, often the proportion of patients that respond to treatment is still low. Major challenges to overcome are the lack of T cell infiltration into the tumor microenvironment as well as the immunosuppressive nature of the tumor, which inhibits the intratumoral immune response. Further, tumors tend to quickly escape the immune response by mutating or losing the expression of drug targets or tumor antigens targeted by the immune response. Thus there is a need in the art for compositions and methods that overcome these limitations. The present disclosure addresses these needs.
The present disclosure provides methods for treating cancer in a subject in need thereof comprising administering to the subject a therapeutically effective amount of (a) a oncolytic viral vector and (b) an antibody directed against a co-stimulatory molecule or a nucleic acid encoding same; or an antibody to an immunesystem agonist molecule or a nucleic acid encoding same.
The oncolytic viral vector can be a Sindbis viral vector. The Sindbis viral vector can be replication defective. The Sindbis viral vector can comprise at least one nucleic acid encoding a therapeutic protein. The Sindbis viral vector can comprise at least one nucleic acid encoding an immunostimulatory or an immunomodulatory protein. The immunostimulatory or immunomodulatory protein can be IL-1, IL-2, IL-3, IL-4, IL-5, IL-6 IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, IL-19, IL-20, IL-21, IL-22, IL-23, IL-24, IL-25, IL-26, IL-27, IL-28, IL-29, IL-30, IL-31, IL-32, IL-33, IL-34, IL-35, IL-36, or any combination thereof. In a preferred aspect, the immunostimulatory or immunomodulatory protein is IL-12. The Sindbis viral vector can comprise at least one nucleic acid encoding LacZ, Flue or GFP.
The antibody can be an anti-OX40 antibody, an anti-4-1BB antibody, an anti-CD28 antibody, an anti-GITR antibody, an anti-CD137 antibody, an anti-CD37 antibody, an anti-HVEM antibody, or a combination thereof.
The Sindbis viral vector and the antibody can induce an immune response in a tumor associated antigen (TAA) nonspecific manner. The induced and nonspecific immune response can be a first immune response. The first immune response can be followed by a secondary immuneresponse. The secondary immune response can be the result of one or more TAAs released from the dead tumor cells. The secondary immune response can comprise memory T cells directed against one or more TAAs released from the dead tumor cells.
The present disclosure also provides methods for treating cancer in a subject in need thereof comprising administering to the subject a therapeutically effective amount of (a) a Sindbis viral vector comprising a nucleic acid encoding interleukin-12 and (b) an anti-OX40 monoclonal antibody or a nucleic acid encoding same, thereby treating cancer in the subject.
The Sindbis viral vector can be replication defective. The Sindbis viral vector can comprise the nucleic acid encoding interleukin-12 and can further comprise the nucleic acid encoding the anti-OX40 monoclonal antibody. The method can comprise administering a Sindbis viral vector comprising the nucleic acid encoding interleukin-12 and administering a Sindbis viral vector comprising the nucleic acid encoding the anti-OX40 monoclonal antibody. The Sindbis viral vector can comprise the nucleic acid encoding interleukin-12 alpha subunit (IL-12 α, IL-12, p35 subunit) and interleukin-12 beta subunit (IL-12 β, IL-12, p40 subunit). The Sindbis viral vector can comprise the nucleic acid encoding interleukin-12 alpha subunit of GenBank accession no. M86672 and interleukin-12 beta subunit (IL-12 β, IL-12, p40 subunit) GenBank accession no. M86671. The nucleic acid encoding interleukin-12 alpha subunit comprises the nucleic acid sequence of SEQ ID NO: 1. The nucleic acid encoding interleukin-12 beta subunit comprises the nucleic acid sequence of SEQ ID NO: 2.
The Sindbis viral vector can comprise a nucleic acid encoding interleukin-12 alpha subunit of amino acid sequence of GenBank accession no. AAA39292.1 and comprise a nucleic acid encoding interleukin-12 beta subunit of amino acid sequence of GenBank accession no. AAA39296.1. The amino acid sequence of the interleukin-12 alpha subunit is of amino acid sequence of SEQ ID NO: 3. The amino acid sequence of the interleukin-12 beta subunit is of amino acid sequence of SEQ ID NO: 4.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 variable heavy chain comprising the amino acid sequence of SEQ ID NO: 5. The nucleic acid sequence encoding the anti-OX40 variable heavy chain is SEQ ID NO: 6. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 variable light chain comprising the amino acid sequence of SEQ ID NO: 7. The nucleic acid sequence encoding the anti-OX40 variable light chain is SEQ ID NO: 8.
The Sindbis viral vector can comprise a nucleic acid encoding a mouse anti-OX40 heavy chain of amino acid sequence of SEQ ID NO; 5 and a nucleic acid encoding a mouse anti-OX40 light chain of amino acid sequence of SEQ ID NO: 7. The Sindbis viral vector can comprise a nucleic acid encoding a mouse anti-OX40 heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to amino acid sequence of SEQ ID NO: 5 and a nucleic acid encoding a mouse anti-OX40 light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to amino acid sequence of SEQ ID NO: 7.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain comprising the amino acid sequence of SEQ ID NO: 9. The nucleic acid sequence encoding the an anti-OX40 antibody heavy chain is SEQ ID NO: 10.
The Sindbis viral vector can comprise a nucleic acid encoding a mouse anti-OX40 antibody light chain comprising the amino acid sequence of SEQ ID No: 11. The nucleic acid sequence encoding the anti-OX40 antibody light chain is SEQ ID NO: 12.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence of SEQ ID NO: 9 and an anti-OX40 antibody light chain with an amino acid sequence of SEQ ID NO: 11. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain of SEQ ID NO: 10 and an anti-OX40 antibody light chain with an amino acid sequence of SEQ ID NO: 12.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 9 and an anti-OX40 antibody light chain with an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 11. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 10 and an anti-OX40 antibody light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 12.
The Sindbis viral vector can comprise the nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody variable heavy chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody variable light chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise the nucleic acid encoding an anti-OX40 antibody light chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody variable heavy chain amino acid sequence, and an anti-OX40 antibody variable light chain amino acid sequence, that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain amino acid sequence, and an anti-OX40 antibody light chain amino acid sequence, that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence of SEQ ID NO: 31. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain is SEQ ID NO: 32. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence of SEQ ID NO: 33. The nucleic acid sequence encoding the human anti-OX40 variable light chain is SEQ ID NO: 34.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 31. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to is SEQ ID NO: 32. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 33. The nucleic acid sequence encoding the human anti-OX40 variable light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 34.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence of SEQ ID NO: 35. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain is SEQ ID NO: 36. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence of SEQ ID NO: 37. The nucleic acid sequence encoding the human anti-OX40 variable light chain is SEQ ID NO: 38.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 35. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to is SEQ ID NO: 36. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 37. The nucleic acid sequence encoding the human anti-OX40 variable light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 38.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence of SEQ ID NO: 39. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain is SEQ ID NO: 40. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence of SEQ ID NO: 41. The nucleic acid sequence encoding the human anti-OX40 variable light chain is SEQ ID NO: 42.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 39. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to is SEQ ID NO: 40. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 41. The nucleic acid sequence encoding the human anti-OX40 variable light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 42.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence of SEQ ID NO: 43. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain is SEQ ID NO: 44. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence of SEQ ID NO: 45. The nucleic acid sequence encoding the human anti-OX40 variable light chain is SEQ ID NO: 46.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 43. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to is SEQ ID NO: 44. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 45. The nucleic acid sequence encoding the human anti-OX40 variable light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 46.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence of SEQ ID NO: 47. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain is SEQ ID NO: 48. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence of SEQ ID NO: 49. The nucleic acid sequence encoding the human anti-OX40 variable light chain is SEQ ID NO: 50.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 47. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to is SEQ ID NO: 48. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 49. The nucleic acid sequence encoding the human anti-OX40 variable light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 50.
The Sindbis viral vector and the anti-OX40 monoclonal antibody can be administered sequentially or concurrently. The Sindbis viral vector can be administered systemically. The anti-OX40 monoclonal antibody can be administered systemically. Both the Sindbis viral vector and the anti-OX40 monoclonal antibody, or a nucleic acid encoding same, can be administered systemically. The Sindbis viral vector can be administered parenterally. The anti-OX40 monoclonal antibody can be administered parenterally. Both the Sindbis viral vector and the anti-OX40 monoclonal antibody, or a nucleic acid encoding same, can be administered parenterally. The Sindbis viral vector can be administered intraperitoneally. The anti-OX40 monoclonal antibody can be administered intraperitoneally. Both the Sindbis viral vector and the anti-OX40 monoclonal antibody, or a nucleic acid encoding same, can be administered intraperitoneally.
An antibody of the present disclosure, or a fragment thereof, can be derived from any species, including, but not limited to, a human, a mouse, a rat, a hamster, a dog, a rabbit, a frog, a sheep, a goat, a cow, a horse, a pig, a bird, a donkey, a chicken, a camel, a llama, a dromedary, an alpaca, a shark, a bovine and a turtle. In some aspects, an antibody of the present disclosure, or a fragment thereof, is derived from a human. In some aspects, an antibody of the present disclosure, or a fragment thereof, is derived from a mouse. In some aspects, an antibody of the present disclosure, or a fragment thereof, is derived from a camel, a llama or an alpaca. In some aspects, an antibody of the present disclosure, or a fragment thereof, is derived from a shark. In some aspects, an antibody of the present disclosure, or a fragment thereof, of the present disclosure is a chimeric antibody that is derived from two or more of the aforementioned species. In a non-limiting example, an antibody of the present disclosure, or fragment thereof, can be a chimeric antibody that is derived from a human and a mouse. In some aspects, an antibody of the present disclosure, or a fragment thereof, can be derived from any species other than human and can be further humanized using standard methods known in the art as to reduce the immunogenicity of the antibody.
A “monoclonal antibody” as disclosed herein, can be a full-length antibody or an antigen binding fragment thereof, wherein the “antigen binding fragment” is a fragment of the full length antibody that retains binding to the target antigen of the said monoclonal antibody. For example, a monoclonal antibody against OX-40 or an anti-OX40 monoclonal antibody, as described herein, can be a full length antibody against OX40 antibody or an antigen binding fragment thereof, wherein the fragment retains binding to the OX40 receptor on a cell surface. An “antigen-binding fragment” of an anti-OX-40 antibody, as described herein can include any fragment selected from the group consisting of Fv, Fav, F(ab′)2, Fab′, dsFv, scFv, sc(Fv)2, scFv-CH3, scFv-Fc, and diabody fragments.
The cancer can be a solid cancer or a liquid/hematologic cancer. The cancer can comprise metastatic cancer. The cancer can comprise a solid tumor. The cancer can be a carcinoma, a lymphoma, a blastoma, a sarcoma, a leukemia, a brain cancer, a breast cancer, a blood cancer, a bone cancer, a lung cancer, a skin cancer, a liver cancer, an ovarian cancer, a bladder cancer, a renal cancer, a gastric cancer, a thyroid cancer, a pancreatic cancer, an esophageal cancer, a prostate cancer, a cervical cancer or a colorectal cancer. In one preferred aspect, the cancer is colon cancer. In one preferred aspect, the cancer is prostate cancer. In one preferred aspect, the cancer is ovarian cancer.
The present disclosure further provides a Sindbis viral vector comprising a nucleic acid encoding interleukin-12 and a nucleic acid encoding an anti-OX40 monoclonal antibody. The present disclosure also provides a composition comprising (a) a Sindbis viral vector comprising a nucleic acid encoding interleukin-12 and (b) an anti-OX40 monoclonal antibody or a nucleic acid encoding same. The present disclosure also provides a composition comprising (a) a Sindbis viral vector comprising a nucleic acid encoding interleukin-12 and (b) a Sindbis viral vector comprising a nucleic acid encoding an anti-OX40 monoclonal antibody.
The Sindbis viral vector can comprise the nucleic acid encoding interleukin-12 alpha subunit (IL-12 α, IL-12, p35 subunit) and interleukin-12 beta subunit (IL-12 β, IL-12, p40 subunit). The Sindbis viral vector can comprise the nucleic acid encoding interleukin-12 alpha subunit of GenBank accession no. M86672 and interleukin-12 beta subunit (IL-12 β, IL-12, p40 subunit) GenBank accession no. M86671. The nucleic acid encoding interleukin-12 alpha subunit comprises the nucleic acid sequence of SEQ ID NO: 1. The nucleic acid encoding interleukin-12 beta subunit comprises the nucleic acid sequence of SEQ ID NO: 2.
The Sindbis viral vector can comprise a nucleic acid encoding interleukin-12 alpha subunit of amino acid sequence of GenBank accession no. AAA39292.1 and comprise a nucleic acid encoding interleukin-12 beta subunit of amino acid sequence of GenBank accession no. AAA39296.1. The amino acid sequence of the interleukin-12 alpha subunit is of amino acid sequence of SEQ ID NO: 3. The amino acid sequence of the interleukin-12 beta subunit is of amino acid sequence of SEQ ID NO: 4.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 variable heavy chain comprising the amino acid sequence of SEQ ID NO: 5. The nucleic acid sequence encoding the anti-OX40 variable heavy chain is SEQ ID NO: 6. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 variable light chain comprising the amino acid sequence of SEQ ID NO: 7. The nucleic acid sequence encoding the anti-OX40 variable light chain is SEQ ID NO: 8.
The Sindbis viral vector can comprise a nucleic acid encoding a mouse anti-OX40 heavy chain of amino acid sequence of SEQ ID NO; 5 and a nucleic acid encoding a mouse anti-OX40 light chain of amino acid sequence of SEQ ID NO: 7. The Sindbis viral vector can comprise a nucleic acid encoding a mouse anti-OX40 heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to amino acid sequence of SEQ ID NO: 5 and a nucleic acid encoding a mouse anti-OX40 light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to amino acid sequence of SEQ ID NO: 7.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain comprising the amino acid sequence of SEQ ID NO: 9. The nucleic acid sequence encoding the an anti-OX40 antibody heavy chain is SEQ ID NO: 10.
The Sindbis viral vector can comprise a nucleic acid encoding a mouse anti-OX40 antibody light chain comprising the amino acid sequence of SEQ ID No: 11. The nucleic acid sequence encoding the anti-OX40 antibody light chain is SEQ ID NO: 12.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence of SEQ ID NO: 9 and an anti-OX40 antibody light chain with an amino acid sequence of SEQ ID NO: 11. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain of SEQ ID NO: 10 and an anti-OX40 antibody light chain with an amino acid sequence of SEQ ID NO: 12.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 9 and an anti-OX40 antibody light chain with an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 11. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 10 and an anti-OX40 antibody light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 12.
The Sindbis viral vector can comprise the nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody variable heavy chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody variable light chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise the nucleic acid encoding an anti-OX40 antibody light chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody variable heavy chain amino acid sequence, and an anti-OX40 antibody variable light chain amino acid sequence, that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain amino acid sequence, and an anti-OX40 antibody light chain amino acid sequence, that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence of SEQ ID NO: 31. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain is SEQ ID NO: 32. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence of SEQ ID NO: 33. The nucleic acid sequence encoding the human anti-OX40 variable light chain is SEQ ID NO: 34.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 31. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to is SEQ ID NO: 32. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 33. The nucleic acid sequence encoding the human anti-OX40 variable light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 34.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence of SEQ ID NO: 35. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain is SEQ ID NO: 36. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence of SEQ ID NO: 37. The nucleic acid sequence encoding the human anti-OX40 variable light chain is SEQ ID NO: 38.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 35. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to is SEQ ID NO: 36. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 37. The nucleic acid sequence encoding the human anti-OX40 variable light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 38.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence of SEQ ID NO: 39. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain is SEQ ID NO: 40. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence of SEQ ID NO: 41. The nucleic acid sequence encoding the human anti-OX40 variable light chain is SEQ ID NO: 42.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 39. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to is SEQ ID NO: 40. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 41. The nucleic acid sequence encoding the human anti-OX40 variable light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 42.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence of SEQ ID NO: 43. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain is SEQ ID NO: 44. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence of SEQ ID NO: 45. The nucleic acid sequence encoding the human anti-OX40 variable light chain is SEQ ID NO: 46.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 43. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to is SEQ ID NO: 44. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 45. The nucleic acid sequence encoding the human anti-OX40 variable light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 46.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence of SEQ ID NO: 47. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain is SEQ ID NO: 48. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence of SEQ ID NO: 49. The nucleic acid sequence encoding the human anti-OX40 variable light chain is SEQ ID NO: 50.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 47. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to is SEQ ID NO: 48. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 49. The nucleic acid sequence encoding the human anti-OX40 variable light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 50.
An antibody of the present disclosure, or a fragment thereof, can be derived from any species, including, but not limited to, a human, a mouse, a rat, a hamster, a dog, a rabbit, a frog, a sheep, a goat, a cow, a horse, a pig, a bird, a donkey, a chicken, a camel, a llama, a dromedary, an alpaca, a shark, a bovine and a turtle. In some aspects, an antibody of the present disclosure, or a fragment thereof, is derived from a human. In some aspects, an antibody of the present disclosure, or a fragment thereof, is derived from a mouse. In some aspects, an antibody of the present disclosure, or a fragment thereof, is derived from a camel, a llama or an alpaca. In some aspects, an antibody of the present disclosure, or a fragment thereof, is derived from a shark. In some aspects, an antibody of the present disclosure, or a fragment thereof, of the present disclosure is a chimeric antibody that is derived from two or more of the aforementioned species. In a non-limiting example, an antibody of the present disclosure, or fragment thereof, can be a chimeric antibody that is derived from a human and a mouse. In some aspects, an antibody of the present disclosure, or a fragment thereof, can be derived from any species other than human and can be further humanized using standard methods known in the art as to reduce the immunogenicity of the antibody.
A “monoclonal antibody” as disclosed herein, can be a full-length antibody or an antigen binding fragment thereof, wherein the “antigen binding fragment” is a fragment of the full length antibody that retains binding to the target antigen of the said monoclonal antibody. For example, a monoclonal antibody against OX-40 or an anti-OX40 monoclonal antibody, as described herein, can be a full length antibody against OX40 antibody or an antigen binding fragment thereof, wherein the fragment retains binding to the OX40 receptor on a cell surface. An “antigen-binding fragment” of an anti-OX-40 antibody, as described herein can include any fragment selected from the group consisting of Fv, Fav, F(ab′)2, Fab′, dsFv, scFv, sc(Fv)2, scFv-CH3, scFv-Fc, and diabody fragments.
The present disclosure also provides methods for treating cancer in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a Sindbis viral vector comprising a nucleic acid encoding interleukin-12 and a Sindbis viral vector comprising a nucleic acid encoding NY-ESO-1, thereby treating cancer in the subject. The present disclosure also provides methods for treating cancer in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a Sindbis viral vector comprising a nucleic acid encoding interleukin-12 and a nucleic acid encoding NY-ESO-1, thereby treating cancer in the subject.
The Sindbis viral vector can comprise the nucleic acid encoding interleukin-12 alpha subunit (IL-12 α, IL-12, p35 subunit) and interleukin-12 beta subunit (IL-12 β, IL-12, p40 subunit). The Sindbis viral vector can comprise the nucleic acid encoding interleukin-12 alpha subunit of GenBank accession no. M86672 and interleukin-12 beta subunit (IL-12 β, IL-12, p40 subunit) GenBank accession no. M86671. The nucleic acid encoding interleukin-12 alpha subunit comprises the nucleic acid sequence of SEQ ID NO: 1. The nucleic acid encoding interleukin-12 beta subunit comprises the nucleic acid sequence of SEQ ID NO: 2.
The Sindbis viral vector can comprise a nucleic acid encoding interleukin-12 alpha subunit of amino acid sequence of GenBank accession no. AAA39292.1 and comprise a nucleic acid encoding interleukin-12 beta subunit of amino acid sequence of GenBank accession no. AAA39296.1. The amino acid sequence of the interleukin-12 alpha subunit is of amino acid sequence of SEQ ID NO: 3. The amino acid sequence of the interleukin-12 beta subunit is of amino acid sequence of SEQ ID NO: 4.
The Sindbis viral vector can comprise the nucleic acid encoding NY-ESO-1 of NCBI Reference accession no. NM_001327.1. The nucleic acid encoding NY-ESO-1 comprises the nucleic acid sequence of SEQ ID NO: 14. The Sindbis viral vector can comprise the nucleic acid encoding NY-ESO-1 of amino acid sequence of NCBI Reference accession no. NP_001318.1. The amino acid sequence of the NY-ESO-1 comprises the amino acid sequence of SEQ ID NO: 15.
The Sindbis viral vector comprising a nucleic acid encoding interleukin-12 and the Sindbis viral vector comprising a nucleic acid encoding NY-ESO-1 can be administered sequentially or concurrently. The Sindbis viral vector comprising a nucleic acid encoding interleukin-12 can be administered systemically. The Sindbis viral vector comprising a nucleic acid encoding NY-ESO-1 can be administered systemically. Both the Sindbis viral vector comprising a nucleic acid encoding interleukin-12 and the Sindbis viral vector comprising a nucleic acid encoding NY-ESO-1 can be administered systemically. The Sindbis viral vector comprising a nucleic acid encoding interleukin-12 can be administered parenterally. The Sindbis viral vector comprising a nucleic acid encoding NY-ESO-1 can be administered parenterally. Both the Sindbis viral vector comprising a nucleic acid encoding interleukin-12 and the Sindbis viral vector comprising a nucleic acid encoding NY-ESO-1 can be administered parenterally. The Sindbis viral vector comprising a nucleic acid encoding interleukin-12 can be administered intraperitoneally. The Sindbis viral vector comprising a nucleic acid encoding NY-ESO-1 can be administered intraperitoneally. Both the Sindbis viral vector comprising a nucleic acid encoding interleukin-12 and the Sindbis viral vector comprising a nucleic acid encoding NY-ESO-1 can be administered intraperitoneally.
The cancer can be a solid cancer or a liquid/hematologic cancer. The cancer can comprise metastatic cancer. The cancer can comprise a solid tumor. The cancer can be a carcinoma, a lymphoma, a blastoma, a sarcoma, a leukemia, a brain cancer, a breast cancer, a blood cancer, a bone cancer, a lung cancer, a skin cancer, a liver cancer, an ovarian cancer, a bladder cancer, a renal cancer, a gastric cancer, a thyroid cancer, a pancreatic cancer, an esophageal cancer, a prostate cancer, a cervical cancer or a colorectal cancer. In one preferred aspect, the cancer is colon cancer. In one preferred aspect, the cancer is prostate cancer. In one preferred aspect, the cancer is ovarian cancer.
The present disclosure further provides a Sindbis viral vector comprising a nucleic acid encoding interleukin-12 and a nucleic acid encoding NY-ESO-1. The present disclosure also provides a composition comprising (a) a Sindbis viral vector comprising a nucleic acid encoding interleukin-12 and (b) a Sindbis viral vector comprising a nucleic acid encoding NY-ESO-1.
The Sindbis viral vector can comprise the nucleic acid encoding interleukin-12 alpha subunit (IL-12 α, IL-12, p35 subunit) and interleukin-12 beta subunit (IL-12 β, IL-12, p40 subunit). The Sindbis viral vector can comprise the nucleic acid encoding interleukin-12 alpha subunit of GenBank accession no. M86672 and interleukin-12 beta subunit (IL-12 β, IL-12, p40 subunit) GenBank accession no. M86671. The nucleic acid encoding interleukin-12 alpha subunit comprises the nucleic acid sequence of SEQ ID NO: 1. The nucleic acid encoding interleukin-12 beta subunit comprises the nucleic acid sequence of SEQ ID NO: 2.
The Sindbis viral vector can comprise a nucleic acid encoding interleukin-12 alpha subunit of amino acid sequence of GenBank accession no. AAA39292.1 and comprise a nucleic acid encoding interleukin-12 beta subunit of amino acid sequence of GenBank accession no. AAA39296.1. The amino acid sequence of the interleukin-12 alpha subunit is of amino acid sequence of SEQ ID NO: 3. The amino acid sequence of the interleukin-12 beta subunit is of amino acid sequence of SEQ ID NO: 4.
The Sindbis viral vector can comprise the nucleic acid encoding NY-ESO-1 of NCBI Reference accession no. NM_001327.1. The nucleic acid encoding NY-ESO-1 comprises the nucleic acid sequence of SEQ ID NO: 14. The Sindbis viral vector can comprise the nucleic acid encoding NY-ESO-1 of amino acid sequence of NCBI Reference accession no. NP_001318.1. The amino acid sequence of the NY-ESO-1 comprises the amino acid sequence of SEQ ID NO: 15.
The present disclosure provides methods for treating cancer in a subject in need thereof comprising administering to the subject a therapeutically effective amount of (a) a Sindbis viral vector comprising a nucleic acid encoding NY-ESO-1 and (b) an anti-OX40 monoclonal antibody or a nucleic acid encoding same, thereby treating cancer in the subject.
The Sindbis viral vector can be replication defective. The Sindbis viral vector can comprise the nucleic acid encoding NY-ESO-1 and can further comprise the nucleic acid encoding the anti-OX40 monoclonal antibody. The method can comprise administering a Sindbis viral vector comprising the nucleic acid encoding NY-ESO-1 and administering a Sindbis viral vector comprising the nucleic acid encoding the anti-OX40 monoclonal antibody. The Sindbis viral vector can comprise the nucleic acid encoding NY-ESO-1 of NCBI Reference accession no. NM_001327.1. The nucleic acid encoding NY-ESO-1 comprises the nucleic acid sequence of SEQ ID NO: 14. The Sindbis viral vector can comprise the nucleic acid encoding NY-ESO-1 of amino acid sequence of NCBI Reference accession no. NP_001318.1. The amino acid sequence of the NY-ESO-1 comprises the amino acid sequence of SEQ ID NO: 15.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 variable heavy chain comprising the amino acid sequence of SEQ ID NO: 5. The nucleic acid sequence encoding the anti-OX40 variable heavy chain is SEQ ID NO: 6. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 variable light chain comprising the amino acid sequence of SEQ ID NO: 7. The nucleic acid sequence encoding the anti-OX40 variable light chain is SEQ ID NO: 8.
The Sindbis viral vector can comprise a nucleic acid encoding a mouse anti-OX40 heavy chain of amino acid sequence of SEQ ID NO; 5 and a nucleic acid encoding a mouse anti-OX40 light chain of amino acid sequence of SEQ ID NO: 7. The Sindbis viral vector can comprise a nucleic acid encoding a mouse anti-OX40 heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to amino acid sequence of SEQ ID NO: 5 and a nucleic acid encoding a mouse anti-OX40 light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to amino acid sequence of SEQ ID NO: 7.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain comprising the amino acid sequence of SEQ ID NO: 9. The nucleic acid sequence encoding the an anti-OX40 antibody heavy chain is SEQ ID NO: 10.
The Sindbis viral vector can comprise a nucleic acid encoding a mouse anti-OX40 antibody light chain comprising the amino acid sequence of SEQ ID No: 11. The nucleic acid sequence encoding the anti-OX40 antibody light chain is SEQ ID NO: 12.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence of SEQ ID NO: 9 and an anti-OX40 antibody light chain with an amino acid sequence of SEQ ID NO: 11. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain of SEQ ID NO: 10 and an anti-OX40 antibody light chain with an amino acid sequence of SEQ ID NO: 12.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 9 and an anti-OX40 antibody light chain with an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 11. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 10 and an anti-OX40 antibody light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 12.
The Sindbis viral vector can comprise the nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody variable heavy chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody variable light chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise the nucleic acid encoding an anti-OX40 antibody light chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody variable heavy chain amino acid sequence, and an anti-OX40 antibody variable light chain amino acid sequence, that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain amino acid sequence, and an anti-OX40 antibody light chain amino acid sequence, that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence of SEQ ID NO: 31. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain is SEQ ID NO: 32. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence of SEQ ID NO: 33. The nucleic acid sequence encoding the human anti-OX40 variable light chain is SEQ ID NO: 34.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 31. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to is SEQ ID NO: 32. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 33. The nucleic acid sequence encoding the human anti-OX40 variable light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 34.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence of SEQ ID NO: 35. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain is SEQ ID NO: 36. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence of SEQ ID NO: 37. The nucleic acid sequence encoding the human anti-OX40 variable light chain is SEQ ID NO: 38.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 35. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to is SEQ ID NO: 36. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 37. The nucleic acid sequence encoding the human anti-OX40 variable light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 38.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence of SEQ ID NO: 39. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain is SEQ ID NO: 40. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence of SEQ ID NO: 41. The nucleic acid sequence encoding the human anti-OX40 variable light chain is SEQ ID NO: 42.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 39. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to is SEQ ID NO: 40. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 41. The nucleic acid sequence encoding the human anti-OX40 variable light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 42.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence of SEQ ID NO: 43. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain is SEQ ID NO: 44. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence of SEQ ID NO: 45. The nucleic acid sequence encoding the human anti-OX40 variable light chain is SEQ ID NO: 46.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 43. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to is SEQ ID NO: 44. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 45. The nucleic acid sequence encoding the human anti-OX40 variable light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 46.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence of SEQ ID NO: 47. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain is SEQ ID NO: 48. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence of SEQ ID NO: 49. The nucleic acid sequence encoding the human anti-OX40 variable light chain is SEQ ID NO: 50.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 47. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to is SEQ ID NO: 48. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 49. The nucleic acid sequence encoding the human anti-OX40 variable light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 50.
The Sindbis viral vector and the anti-OX40 monoclonal antibody can be administered sequentially or concurrently. The Sindbis viral vector can be administered systemically. The anti-OX40 monoclonal antibody can be administered systemically. Both the Sindbis viral vector and the anti-OX40 monoclonal antibody, or a nucleic acid encoding same, can be administered systemically. The Sindbis viral vector can be administered parenterally. The anti-OX40 monoclonal antibody can be administered parenterally. Both the Sindbis viral vector and the anti-OX40 monoclonal antibody, or a nucleic acid encoding same, can be administered parenterally. The Sindbis viral vector can be administered intraperitoneally. The anti-OX40 monoclonal antibody can be administered intraperitoneally. Both the Sindbis viral vector and the anti-OX40 monoclonal antibody, or a nucleic acid encoding same, can be administered intraperitoneally.
An antibody of the present disclosure, or a fragment thereof, can be derived from any species, including, but not limited to, a human, a mouse, a rat, a hamster, a dog, a rabbit, a frog, a sheep, a goat, a cow, a horse, a pig, a bird, a donkey, a chicken, a camel, a llama, a dromedary, an alpaca, a shark, a bovine and a turtle. In some aspects, an antibody of the present disclosure, or a fragment thereof, is derived from a human. In some aspects, an antibody of the present disclosure, or a fragment thereof, is derived from a mouse. In some aspects, an antibody of the present disclosure, or a fragment thereof, is derived from a camel, a llama or an alpaca. In some aspects, an antibody of the present disclosure, or a fragment thereof, is derived from a shark. In some aspects, an antibody of the present disclosure, or a fragment thereof, of the present disclosure is a chimeric antibody that is derived from two or more of the aforementioned species. In a non-limiting example, an antibody of the present disclosure, or fragment thereof, can be a chimeric antibody that is derived from a human and a mouse. In some aspects, an antibody of the present disclosure, or a fragment thereof, can be derived from any species other than human and can be further humanized using standard methods known in the art as to reduce the immunogenicity of the antibody.
A “monoclonal antibody” as disclosed herein, can be a full-length antibody or an antigen binding fragment thereof, wherein the “antigen binding fragment” is a fragment of the full length antibody that retains binding to the target antigen of the said monoclonal antibody. For example, a monoclonal antibody against OX-40 or an anti-OX40 monoclonal antibody, as described herein, can be a full length antibody against OX40 antibody or an antigen binding fragment thereof, wherein the fragment retains binding to the OX40 receptor on a cell surface. An “antigen-binding fragment” of an anti-OX-40 antibody, as described herein can include any fragment selected from the group consisting of Fv, Fav, F(ab′)2, Fab′, dsFv, scFv, sc(Fv)2, scFv-CH3, scFv-Fc, and diabody fragments.
The cancer can be a solid cancer or a liquid/hematologic cancer. The cancer can comprise metastatic cancer. The cancer can comprise a solid tumor. The cancer can be a carcinoma, a lymphoma, a blastoma, a sarcoma, a leukemia, a brain cancer, a breast cancer, a blood cancer, a bone cancer, a lung cancer, a skin cancer, a liver cancer, an ovarian cancer, a bladder cancer, a renal cancer, a gastric cancer, a thyroid cancer, a pancreatic cancer, an esophageal cancer, a prostate cancer, a cervical cancer or a colorectal cancer. In one preferred aspect, the cancer is colon cancer. In one preferred aspect, the cancer is prostate cancer. In one preferred aspect, the cancer is ovarian cancer.
The present disclosure further provides a Sindbis viral vector comprising a nucleic acid encoding NY-ESO-1 and a nucleic acid encoding an anti-OX40 monoclonal antibody. The present disclosure also provides a composition comprising (a) a Sindbis viral vector comprising a nucleic acid encoding NY-ESO-1 and (b) an anti-OX40 monoclonal antibody or a nucleic acid encoding same. The present disclosure also provides a composition comprising (a) a Sindbis viral vector comprising a nucleic acid encoding NY-ESO-1 and (b) a Sindbis viral vector comprising a nucleic acid encoding an anti-OX40 monoclonal antibody. The Sindbis viral vector can comprise the nucleic acid encoding NY-ESO-1 of NCBI Reference accession no. NM_001327.1. The nucleic acid encoding NY-ESO-1 comprises the nucleic acid sequence of SEQ ID NO: 14. The Sindbis viral vector can comprise the nucleic acid encoding NY-ESO-1 of amino acid sequence of NCBI Reference accession no. NP_001318.1. The amino acid sequence of the NY-ESO-1 comprises the amino acid sequence of SEQ ID NO: 15.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 variable heavy chain comprising the amino acid sequence of SEQ ID NO: 5. The nucleic acid sequence encoding the anti-OX40 variable heavy chain is SEQ ID NO: 6. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 variable light chain comprising the amino acid sequence of SEQ ID NO: 7. The nucleic acid sequence encoding the anti-OX40 variable light chain is SEQ ID NO: 8.
The Sindbis viral vector can comprise a nucleic acid encoding a mouse anti-OX40 heavy chain of amino acid sequence of SEQ ID NO; 5 and a nucleic acid encoding a mouse anti-OX40 light chain of amino acid sequence of SEQ ID NO: 7. The Sindbis viral vector can comprise a nucleic acid encoding a mouse anti-OX40 heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to amino acid sequence of SEQ ID NO: 5 and a nucleic acid encoding a mouse anti-OX40 light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to amino acid sequence of SEQ ID NO: 7.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain comprising the amino acid sequence of SEQ ID NO: 9. The nucleic acid sequence encoding the an anti-OX40 antibody heavy chain is SEQ ID NO: 10.
The Sindbis viral vector can comprise a nucleic acid encoding a mouse anti-OX40 antibody light chain comprising the amino acid sequence of SEQ ID No: 11. The nucleic acid sequence encoding the anti-OX40 antibody light chain is SEQ ID NO: 12.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence of SEQ ID NO: 9 and an anti-OX40 antibody light chain with an amino acid sequence of SEQ ID NO: 11. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain of SEQ ID NO: 10 and an anti-OX40 antibody light chain with an amino acid sequence of SEQ ID NO: 12.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 9 and an anti-OX40 antibody light chain with an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 11. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 10 and an anti-OX40 antibody light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 12.
The Sindbis viral vector can comprise the nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody variable heavy chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody variable light chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise the nucleic acid encoding an anti-OX40 antibody light chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody variable heavy chain amino acid sequence, and an anti-OX40 antibody variable light chain amino acid sequence, that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain amino acid sequence, and an anti-OX40 antibody light chain amino acid sequence, that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13.
An antibody of the present disclosure, or a fragment thereof, can be derived from any species, including, but not limited to, a human, a mouse, a rat, a hamster, a dog, a rabbit, a frog, a sheep, a goat, a cow, a horse, a pig, a bird, a donkey, a chicken, a camel, a llama, a dromedary, an alpaca, a shark, a bovine and a turtle. In some aspects, an antibody of the present disclosure, or a fragment thereof, is derived from a human. In some aspects, an antibody of the present disclosure, or a fragment thereof, is derived from a mouse. In some aspects, an antibody of the present disclosure, or a fragment thereof, is derived from a camel, a llama or an alpaca. In some aspects, an antibody of the present disclosure, or a fragment thereof, is derived from a shark. In some aspects, an antibody of the present disclosure, or a fragment thereof, of the present disclosure is a chimeric antibody that is derived from two or more of the aforementioned species. In a non-limiting example, an antibody of the present disclosure, or fragment thereof, can be a chimeric antibody that is derived from a human and a mouse. In some aspects, an antibody of the present disclosure, or a fragment thereof, can be derived from any species other than human and can be further humanized using standard methods known in the art as to reduce the immunogenicity of the antibody.
A “monoclonal antibody” as disclosed herein, can be a full-length antibody or an antigen binding fragment thereof, wherein the “antigen binding fragment” is a fragment of the full length antibody that retains binding to the target antigen of the said monoclonal antibody. For example, a monoclonal antibody against OX-40 or an anti-OX40 monoclonal antibody, as described herein, can be a full length antibody against OX40 antibody or an antigen binding fragment thereof, wherein the fragment retains binding to the OX40 receptor on a cell surface. An “antigen-binding fragment” of an anti-OX40 antibody, as described herein can include any fragment selected from the group consisting of Fv, Fav, F(ab′)2, Fab′, dsFv, scFv, sc(Fv)2, scFv-CH3, scFv-Fc, and diabody fragments.
The present disclosure also provides methods for treating cancer in a subject in need thereof comprising administering to the subject a therapeutically effective amount (a) a Sindbis viral vector and (b) an anti-4-1BB (CD137) monoclonal antibody or a nucleic acid encoding same, thereby treating cancer in the subject. The present disclosure further provides in vitro or ex vivo methods for treating cancer or assessing the treatment of cancer in a subject comprising contacting a biological sample from the subject with (a) a Sindbis viral vector and (b) an anti-4-1BB monoclonal antibody or a nucleic acid encoding same. The Sindbis viral vector does not comprise an endogenous nucleic acid encoding any protein.
The Sindbis viral vector is replication defective. The Sindbis viral vector can comprise a nucleic acid sequence encoding a therapeutic protein, an immunostimulatory protein or an immunomodulatory protein. The Sindbis viral vector can comprise the nucleic acid encoding the therapeutic protein, an immunostimulatory protein or an immunomodulatory protein and further comprise the nucleic acid encoding the anti-4-1BB monoclonal antibody. The Sindbis viral vector can comprise a nucleic acid sequence encoding LacZ (lac operon structural gene lacZ encoding β-galactosidase), Flue (firefly luciferase) or GFP (green fluorescent protein). The Sindbis viral vector can comprise the nucleic acid encoding LacZ, Flue or GFP and further comprise the nucleic acid encoding the anti-4-1BB monoclonal antibody.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-4-1BB antibody heavy chain comprising the heavy chain complementarity determining region 1 (HCDR1), HCDR2 and HCDR3 amino acid sequences of SEQ ID NOs: 16, 17 and 18, respectively. The Sindbis viral vector can comprise the nucleic acid encoding an anti-4-1BB antibody heavy chain comprising the amino acid sequence of SEQ ID NO: 19. The Sindbis viral vector can comprise a nucleic acid encoding an anti-4-1BB antibody heavy chain comprising the light chain complementarity determining region 1 (LCDR1), LCDR2 and LCDR3 amino acid sequences of SEQ ID NOs: 20, 21 and 22, respectively. The Sindbis viral vector can comprise the nucleic acid encoding an anti-4-1BB antibody light chain comprising the amino acid sequence of SEQ ID NO: 23.
The Sindbis viral vector can comprise the nucleic acid encoding anti-4-1BB antibody heavy chain CDR1, CDR2 and CDR3 amino acid sequences that binds to a target antigen of the amino acid sequence of SEQ ID NO: 24. The Sindbis viral vector can comprise the nucleic acid encoding anti-4-1BB antibody light chain CDR1, CDR2 and CDR3 amino acid sequences that binds to a target antigen of the amino acid sequence of SEQ ID NO: 24. The Sindbis viral vector can comprise the nucleic acid encoding anti-4-1BB antibody heavy chain amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 24. The Sindbis viral vector can comprise the nucleic acid encoding anti-4-1BB antibody light chain amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 24.
The immunostimulatory or immunomodulatory protein can be IL-1, IL-2, IL-3, IL-4, IL-5, IL-6 IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, IL-19, IL-20, IL-21, IL-22, IL-23, IL-24, IL-25, IL-26, IL-27, IL-28, IL-29, IL-30, IL-31, IL-32, IL-33, IL-34, IL-35, IL-36, or any combination thereof. In a preferred aspect, the immunostimulatory or immunomodulatory protein is IL-12. The anti-4-1BB antibody can be urelumab, utomilumab or a combination thereof. The anti-4-1BB antibody can be InVivoMAb anti-mouse 4-1BB (BioXCell, Clone: LOB12.3, Cat. No. BE0169).
The Sindbis viral vector and the anti-4-1BB monoclonal antibody can be administered sequentially or concurrently. The Sindbis viral vector can be administered systemically. The anti-4-1BB monoclonal antibody can be administered systemically. Both the Sindbis viral vector and the anti-4-1BB monoclonal antibody, or a nucleic acid encoding same, can be administered systemically. The Sindbis viral vector can be administered parenterally. The anti-4-1BB monoclonal antibody can be administered parenterally. Both the Sindbis viral vector and the anti-4-1BB monoclonal antibody, or a nucleic acid encoding same, can be administered parenterally. The Sindbis viral vector can be administered intraperitoneally. The anti-4-1BB monoclonal antibody can be administered intraperitoneally. Both the Sindbis viral vector and the anti-4-1BB monoclonal antibody, or a nucleic acid encoding same, can be administered intraperitoneally.
An antibody of the present disclosure, or a fragment thereof, can be derived from any species, including, but not limited to, a human, a mouse, a rat, a hamster, a dog, a rabbit, a frog, a sheep, a goat, a cow, a horse, a pig, a bird, a donkey, a chicken, a camel, a llama, a dromedary, an alpaca, a shark, a bovine and a turtle. In some aspects, an antibody of the present disclosure, or a fragment thereof, is derived from a human. In some aspects, an antibody of the present disclosure, or a fragment thereof, is derived from a mouse. In some aspects, an antibody of the present disclosure, or a fragment thereof, is derived from a camel, a llama or an alpaca. In some aspects, an antibody of the present disclosure, or a fragment thereof, is derived from a shark. In some aspects, an antibody of the present disclosure, or a fragment thereof, of the present disclosure is a chimeric antibody that is derived from two or more of the aforementioned species. In a non-limiting example, an antibody of the present disclosure, or fragment thereof, can be a chimeric antibody that is derived from a human and a mouse. In some aspects, an antibody of the present disclosure, or a fragment thereof, can be derived from any species other than human and can be further humanized using standard methods known in the art as to reduce the immunogenicity of the antibody.
A “monoclonal antibody” as disclosed herein, can be a full-length antibody or an antigen binding fragment thereof, wherein the “antigen binding fragment” is a fragment of the full length antibody that retains binding to the target antigen of the said monoclonal antibody. For example, a monoclonal antibody against 4-1BB or an anti-4-1BB monoclonal antibody, as described herein, can be a full length antibody against 4-1BB antibody or an antigen binding fragment thereof, wherein the fragment retains binding to the 4-1BB receptor on a cell surface. An “antigen-binding fragment” of an anti-4-1BB antibody, as described herein can include any fragment selected from the group consisting of Fv, Fav, F(ab′)2, Fab′, dsFv, scFv, sc(Fv)2, scFv-CH3, scFv-Fc, and diabody fragments.
The cancer can be a solid cancer or a liquid/hematologic cancer. The cancer can comprise metastatic cancer. The cancer can comprise a solid tumor. The cancer can be a carcinoma, a lymphoma, a blastoma, a sarcoma, a leukemia, a brain cancer, a breast cancer, a blood cancer, a bone cancer, a lung cancer, a skin cancer, a liver cancer, an ovarian cancer, a bladder cancer, a renal cancer, a gastric cancer, a thyroid cancer, a pancreatic cancer, an esophageal cancer, a prostate cancer, a cervical cancer or a colorectal cancer. In one preferred aspect, the cancer is a lymphoma. In one preferred aspect, the cancer is a B cell lymphoma.
The present disclosure provides a Sindbis viral vector comprising a nucleic acid encoding a therapeutic protein, an immunostimulatory protein or an immunomodulatory protein and a nucleic acid encoding an anti-4-1BB monoclonal antibody. The present disclosure provides a Sindbis viral vector comprising a nucleic acid encoding encoding LacZ, Flue or GFP and a nucleic acid encoding an anti-4-1BB monoclonal antibody. The present disclosure also provides a composition comprising (a) a Sindbis viral vector comprising a nucleic acid encoding a therapeutic protein, an immunostimulatory protein or an immunomodulatory protein and (b) a Sindbis viral vector comprising a nucleic acid encoding an anti-4-1BB monoclonal antibody. The present disclosure also provides a composition comprising (a) a Sindbis viral vector comprising a nucleic acid encoding LacZ, Flue or GFP and (b) a Sindbis viral vector comprising a nucleic acid encoding an anti-4-1BB monoclonal antibody. The present disclosure further provides a composition comprising (a) a Sindbis viral vector comprising a nucleic acid encoding a therapeutic protein, an immunostimulatory protein or an immunomodulatory protein and (b) an anti-4-1BB monoclonal antibody or a nucleic acid encoding same. The present disclosure provides a composition comprising (a) a Sindbis viral vector comprising a nucleic acid encoding encoding LacZ, Flue or GFP and (b) an anti-4-1BB monoclonal antibody or a nucleic acid encoding same.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-4-1BB antibody heavy chain comprising the heavy chain complementarity determining region 1 (HCDR1), HCDR2 and HCDR3 amino acid sequences of SEQ ID NOs: 16, 17 and 18, respectively. The Sindbis viral vector can comprise the nucleic acid encoding an anti-4-1BB antibody heavy chain comprising the amino acid sequence of SEQ ID NO: 19. The Sindbis viral vector can comprise a nucleic acid encoding an anti-4-1BB antibody heavy chain comprising the light chain complementarity determining region 1 (LCDR1), LCDR2 and LCDR3 amino acid sequences of SEQ ID NOs: 20, 21 and 22, respectively. The Sindbis viral vector can comprise the nucleic acid encoding an anti-4-1BB antibody light chain comprising the amino acid sequence of SEQ ID NO: 23.
The Sindbis viral vector can comprise the nucleic acid encoding anti-4-1BB antibody heavy chain CDR1, CDR2 and CDR3 amino acid sequences that binds to a target antigen of the amino acid sequence of SEQ ID NO: 24. The Sindbis viral vector can comprise the nucleic acid encoding anti-4-1BB antibody light chain CDR1, CDR2 and CDR3 amino acid sequences that binds to a target antigen of the amino acid sequence of SEQ ID NO: 24. The Sindbis viral vector can comprise the nucleic acid encoding anti-4-1BB antibody heavy chain amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 24. The Sindbis viral vector can comprise the nucleic acid encoding anti-4-1BB antibody light chain amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 24.
The immunostimulatory or immunomodulatory protein can be IL-1, IL-2, IL-3, IL-4, IL-5, IL-6 IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, IL-19, IL-20, IL-21, IL-22, IL-23, IL-24, IL-25, IL-26, IL-27, IL-28, IL-29, IL-30, IL-31, IL-32, IL-33, IL-34, IL-35, IL-36, or any combination thereof. In a preferred aspect, the immunostimulatory or immunomodulatory protein is IL-12.
An antibody of the present disclosure, or a fragment thereof, can be derived from any species, including, but not limited to, a human, a mouse, a rat, a hamster, a dog, a rabbit, a frog, a sheep, a goat, a cow, a horse, a pig, a bird, a donkey, a chicken, a camel, a llama, a dromedary, an alpaca, a shark, a bovine and a turtle. In some aspects, an antibody of the present disclosure, or a fragment thereof, is derived from a human. In some aspects, an antibody of the present disclosure, or a fragment thereof, is derived from a mouse. In some aspects, an antibody of the present disclosure, or a fragment thereof, is derived from a camel, a llama or an alpaca. In some aspects, an antibody of the present disclosure, or a fragment thereof, is derived from a shark. In some aspects, an antibody of the present disclosure, or a fragment thereof, of the present disclosure is a chimeric antibody that is derived from two or more of the aforementioned species. In a non-limiting example, an antibody of the present disclosure, or fragment thereof, can be a chimeric antibody that is derived from a human and a mouse. In some aspects, an antibody of the present disclosure, or a fragment thereof, can be derived from any species other than human and can be further humanized using standard methods known in the art as to reduce the immunogenicity of the antibody.
A “monoclonal antibody” as disclosed herein, can be a full-length antibody or an antigen binding fragment thereof, wherein the “antigen binding fragment” is a fragment of the full length antibody that retains binding to the target antigen of the said monoclonal antibody. For example, a monoclonal antibody against 4-1BB or an anti-4-1BB monoclonal antibody, as described herein, can be a full length antibody against 4-1BB antibody or an antigen binding fragment thereof, wherein the fragment retains binding to the 4-1BB receptor on a cell surface. An “antigen-binding fragment” of an anti-4-1BB antibody, as described herein can include any fragment selected from the group consisting of Fv, Fav, F(ab′)2, Fab′, dsFv, scFv, sc(Fv)2, scFv-CH3, scFv-Fc, and diabody fragments.
Any of the above aspects can be combined with any other aspect.
Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. In the Specification, the singular forms also include the plural unless the context clearly dictates otherwise; as examples, the terms “a,” “an,” and “the” are understood to be singular or plural and the term “or” is understood to be inclusive. By way of example, “an element” means one or more element. Throughout the specification the word “comprising,” or variations such as “comprises” or “comprising,” will be understood to imply the inclusion of a stated element, integer or step, or group of elements, integers or steps, but not the exclusion of any other element, integer or step, or group of elements, integers or steps. About can be understood as within 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.1%, 0.05%, or 0.01% of the stated value. Unless otherwise clear from the context, all numerical values provided herein are modified by the term “about.”
Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present disclosure, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. The references cited herein are not admitted to be prior art to the claimed invention. In the case of conflict, the present Specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and are not intended to be limiting. Other features and advantages of the disclosure will be apparent from the following detailed description and claims.
Oncolytic virus (OV) therapy has become a novel immunotherapeutic approach to treat cancer. A rationale for oncolytic virus is that they can infect and lyse the tumor cell. They have been made to selectively replicate in tumor cells either through the direction of tumor specific promoters or through direct intratumoral administration. Most OVs encounter a number of barriers to systemic administration. Once lysed by OVs, tumor cells release tumor associated antigens (TAAs) that can stimulate cytotoxic T cells. OV infection also induces an inflammatory response that helps to trigger an immune anti-tumor response. Several OV clinical trials are underway and have shown promising results. However, whether OV therapy can effectively treat tumors that they are unable to infect remains an unresolved limitation.
Sindbis virus (SV) belongs to alphavirus genus and is one type of OV. Although it does not lyse infected tumor cells, it can cause their apoptotic death. It offers several important benefits. SV is known as one of the least virulent alphaviruses with clinical signs and symptoms usually unapparent. It has been estimated that there are 17 times more subclinical than symptomatic SV infections. In general, when symptoms do occur in humans they consist of a self-limiting, mild, febrile disease with vesicular exanthema and arthralgia from which most patients recover within 14 days. The disease is in part self-limiting because SV is an RNA virus that does not integrate in the host genome and hence its presence is transitory. The lack of an integrative step in its replication cycle also avoids insertional mutagenesis risks. In addition, SV vectors of the present disclosure were generated from the laboratory strain AR339, which is not known to cause disease in humans. These vectors were further attenuated by rendering them replication-defective.
SV vectors can target tumors systemically and can reach metastatic tumor cells throughout the body. They can target tumors without infecting normal tissues. However, susceptibility to infection by SV vectors depends on a number of factors including laminin receptor expression and distribution, as well as, defects in IFN signaling in tumors. The present disclosure demonstrates that SV vectors can effectively help cure tumors that they are unable to infect and further demonstrates that the combination antibodies and SV vectors provide a surprising synergistic therapeutic effect against cancer.
The present disclosure provides methods for treating cancer in a subject in need thereof comprising administering to the subject a therapeutically effective amount of (a) a oncolytic viral vector and (b) an antibody directed against a co-stimulatory molecule or a nucleic acid encoding same; or an antibody to an immune system agonist molecule or a nucleic acid encoding same.
The oncolytic viral vector can be a Sindbis viral vector. The Sindbis viral vector can be replication defective. Sindbis viral vectors were produced as described in U.S. Pat. No. 8,093,021 (incorporated herein by reference in its entirety). The Sindbis viral vector can comprise at least one nucleic acid encoding a therapeutic protein. The Sindbis viral vector can comprise at least one nucleic acid encoding an immunostimulatory or an immunomodulatory protein. The immunostimulatory or immunomodulatory protein can be IL-1, IL-2, IL-3, IL-4, IL-5, IL-6 IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, IL-19, IL-20, IL-21, IL-22, IL-23, IL-24, IL-25, IL-26, IL-27, IL-28, IL-29, IL-30, IL-31, IL-32, IL-33, IL-34, IL-35, IL-36, or any combination thereof. In a preferred aspect, the immunostimulatory or immunomodulatory protein is IL-12. The Sindbis viral vector can comprise at least one nucleic acid encoding LacZ, Flue or GFP.
The antibody can be an anti-OX40 antibody, an anti-4-1BB antibody, an anti-CD28 antibody, an anti-GITR antibody, an anti-CD137 antibody, an anti-cd37 antibody, an anti-HVEM antibody, or a combination thereof.
The Sindbis viral vector and the antibody can induce an immune response in a tumor associated antigen (TAA) nonspecific manner. The induced and nonspecific immune response can be a first immune response. The first immune response can be followed by a secondary immuneresponse. The secondary immune response can be the result of one or more TAAs released from the dead tumor cells. The secondary immune response can comprise memory T cells directed against one or more TAAs released from the dead tumor cells.
The present disclosure also provides methods for treating cancer in a subject in need thereof comprising administering to the subject a therapeutically effective amount of (a) a Sindbis viral vector comprising a nucleic acid encoding interleukin-12 and (b) an anti-OX40 monoclonal antibody or a nucleic acid encoding same, thereby treating cancer in the subject.
The Sindbis viral vector can be replication defective. The Sindbis viral vector can comprise the nucleic acid encoding interleukin-12 and can further comprise the nucleic acid encoding the anti-OX40 monoclonal antibody. The method can comprise administering a Sindbis viral vector comprising the nucleic acid encoding interleukin-12 and administering a Sindbis viral vector comprising the nucleic acid encoding the anti-OX40 monoclonal antibody.
The Sindbis viral vector can comprise the nucleic acid encoding interleukin-12 alpha subunit (IL-12 α, IL-12, p35 subunit) and interleukin-12 beta subunit (IL-12 β, IL-12, p40 subunit). The Sindbis viral vector can comprise the nucleic acid encoding interleukin-12 alpha subunit of GenBank accession no. M86672 and interleukin-12 beta subunit of GenBank accession no. M86671. The nucleic acid encoding interleukin-12 alpha subunit comprises the nucleic acid sequence of SEQ ID NO: 1. The nucleic acid encoding interleukin-12 beta subunit comprises the nucleic acid sequence of SEQ ID NO: 2.
The nucleic acid encoding interleukin-12 alpha subunit (IL-12 α, IL-12, p35 subunit) comprises the nucleic acid sequence of SEQ ID NO: 1 shown in the following Table.
The nucleic acid encoding interleukin-12 beta subunit (IL-12 β, IL-12, p40 subunit) comprises the nucleic acid sequence of SEQ ID NO: 2 shown in the following Table.
The Sindbis viral vector can comprise the nucleic acid encoding interleukin-12 alpha subunit of amino acid sequence of GenBank accession no. AAA39292.1 and interleukin-12 beta subunit of amino acid sequence of GenBank accession no. AAA39296.1. The amino acid sequence of the interleukin-12 alpha subunit is of amino acid sequence of SEQ ID NO: 3. The amino acid sequence of the interleukin-12 beta subunit is of amino acid sequence of SEQ ID NO: 4.
The Sindbis viral vector can comprise the nucleic acid encoding the interleukin-12 alpha subunit of amino acid sequence of GenBank accession no. AAA39292.1 and a nucleic acid encoding the interleukin-12 beta subunit of amino acid sequence of GenBank accession no. AAA39296.1. The amino acid sequence of the interleukin-12 alpha subunit comprises the amino acid sequence of SEQ ID NO: 3 shown in the following Table.
The amino acid sequence of the interleukin-12 beta subunit comprises the amino acid sequence of SEQ ID NO: 4 shown in the following Table.
The Sindbis viral vector can comprise a nucleic acid encoding an interleukin-12 alpha subunit that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to amino acid sequence of SEQ ID NO: 3 and a nucleic acid encoding an interleukin-12 alpha subunit that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to amino acid sequence of SEQ ID NO: 4.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 variable heavy chain comprising the amino acid sequence of SEQ ID NO: 5. The nucleic acid sequence encoding the anti-OX40 variable heavy chain is SEQ ID NO: 6. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 variable light chain comprising the amino acid sequence of SEQ ID NO: 7. The nucleic acid sequence encoding the anti-OX40 variable light chain is SEQ ID NO: 8.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 variable heavy chain comprising the amino acid sequence of SEQ ID NO:5 shown in the following Table.
The nucleic acid sequence encoding the anti-OX40 variable heavy chain comprises the nucleic acid sequence of SEQ ID NO:6 shown in the following Table.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 variable light chain comprising the amino acid sequence of SEQ ID NO:7 shown in the following Table.
The nucleic acid sequence encoding the anti-OX40 variable light chain comprises the nucleic acid sequence of SEQ ID NO:8 shown in the following Table.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 variable heavy chain of amino acid sequence of SEQ ID NO: 5 and a nucleic acid encoding an anti-OX40 variable light chain of amino acid sequence of SEQ ID NO: 7. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 variable heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to amino acid sequence of SEQ ID NO: 5 and a nucleic acid encoding an anti-OX40 variable light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%˜ or 100% identical to amino acid sequence of SEQ ID NO: 7.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain comprising the amino acid sequence of SEQ ID NO:9 shown in the following Table.
MGQSRYLLFLATLALLNHLSLA
MAEVQLVESGGGLVQPGGSLRLSCAASG
FTFSNYTMNWVRQAPGKGLEWVSAISGSGGSTYYADSVKGRFTISRDNSK
NTLYLQMNSLRAEDTAVYYCAKDRYSQVHYALDYWGQGTLVTVAAKTTAP
PAPNLLGGPSVFIFPPKIKDVLMISLSPIVTCVVV
The nucleic acid sequence encoding the anti-OX40 antibody heavy chain comprises the nucleic acid sequence of SEQ ID NO: 10 shown in the following Table.
ggccgaggtgcagctggtggagagcggcggcggcctggtgcagcccggcggcagcctgaggctgagctgcgccgcc
agcggcttcaccttcagcaactacaccatgaactgggtgaggcaggcccccggcaagggcctggagtgggtgagcg
ccatcagcggcagcggcggcagcacctactacgccgacagcgtgaagggcaggttcaccatcagcagggacaacag
caagaacaccctgtacctgcagatgaacagcctgagggccgaggacaccgccgtgtactactgcgccaaggacagg
tacagccaggtgcactacgccctggactactggggccagggcaccctggtgaccgtggccgccaagaccaccgccc
In SEQ ID NOs: 9 and 10, the underlined residues indicate IL-2 signal peptide; the Bold residues indicate variable antigen binding region; the non-underlined residues indicate mouse heavy chain IgG2a constant region, GB Accession BC080671; and the bold and underlined residues indicate the Hinge and disulfide bond region. The double underlined residue in SEQ ID NO: 9 indicates change from C to T to remove ApaI site. The dotted underlined residues in SEQ ID NO: 9 indicate Kozak sequence.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody light chain comprising the amino acid sequence of SEQ ID NO:11 shown in the following Table.
MGQSRYLLFLATLALLNHLSLA
DIQMTQSPDSLPVTPGEPASISCRSSQS
LLHSNGYNYLDWYLQKAGQSPQLLIYLGSNRASGVPDRFSGSGSGTDFTL
The nucleic acid sequence encoding the mouse anti-OX40 antibody light chain comprises the nucleic acid sequence of SEQ ID NO 12 shown in the following Table
acatccagatgacccagtcccccgactccctgcccgtgacccccggcgagcccgcctccatctcctgccggtcct
cccagtccctgctgcactccaacggctacaactacctggactggtacctgcagaaggccggccagtccccccagc
tgctgatctacctgggctccaaccgggcctccggcgtgcccgaccggttctccggctccggctccggcaccgact
tcaccctgaagatctcccgggtggaggccgaggacgtgggcgtgtactactgccagcagtactacaaccacccca
ccaccttcggccagggcaccaagctggagatcaagcgggccgacgccgcccccaccgtgtccatcttccccccct
In SEQ ID NOs: 11 and 12, the underlined residues indicate IL-2 signal peptide; the Bold residues indicate variable antigen binding region; the non-underlined residues indicate light constant region, GB Accession BC091750.1. The dotted underlined residues in SEQ ID NO: 12 indicate Kozak sequence.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence of SEQ ID NO: 9 and an anti-OX40 antibody light chain with an amino acid sequence of SEQ ID NO: 11. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain of SEQ ID NO: 10 and an anti-OX40 antibody light chain with an amino acid sequence of SEQ ID NO: 12.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 9 and an anti-OX40 antibody light chain with an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 11. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 10 and an anti-OX40 antibody light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 12.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO:13 shown in the following Table.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 antibody variable heavy chain comprising the amino acid sequence of SEQ ID NO: 31 shown in the following Table.
MTMITPSLVP SSDPLVTAAS VLEFALLIRL TIGQAVVSTQ
The nucleic acid sequence encoding the human anti-OX40 antibody heavy chain variable region comprises the nucleic acid sequence of SEQ ID NO: 32 shown in the following Table.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 antibody variable light chain comprising the amino acid sequence of SEQ ID NO: 33 shown in the following Table.
MEAPAQLLFL LLLWLPDTTG EIVLTQSPAT LSLSPGERAT
The nucleic acid sequence encoding the human anti-OX40 antibody light chain variable region comprises the nucleic acid sequence of SEQ ID NO: 34 shown in the following Table.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 antibody variable heavy chain comprising the amino acid sequence of SEQ ID NO: 35 shown in the following Table.
MEWGPCWVFL VVILEGVQCG VQLVESGGGL VQPGGSLRLS
The nucleic acid sequence encoding the human anti-OX40 antibody heavy chain variable region comprises the nucleic acid sequence of SEQ ID NO: 36 shown in the following Table.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 antibody variable light chain comprising the amino acid sequence of SEQ ID NO: 37 shown in the following Table.
MDMRVLAQLL GLLLLCFPGA RCDIQMTQSP SSLSASVGNR
The nucleic acid sequence encoding the human anti-OX40 antibody light chain variable region comprises the nucleic acid sequence of SEQ ID NO: 38 shown in the following Table.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 antibody variable heavy chain comprising the amino acid sequence of SEQ ID NO: 39 shown in the following Table.
MDTLCSTLLL LTIPSWVLSQ ITLKESGPTL VKPTQTLTLT
The nucleic acid sequence encoding the human anti-OX40 antibody heavy chain variable region comprises the nucleic acid sequence of SEQ ID NO: 40 shown in the following Table.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 antibody variable light chain comprising the amino acid sequence of SEQ ID NO: 41 shown in the following Table.
MEAPAQLLFL LLLWLPDTTG EIVLTQSPAT LSLSPGERAT LSCRASQGVS SYLAWYQQKP
The nucleic acid sequence encoding the human anti-OX40 antibody light chain variable region comprises the nucleic acid sequence of SEQ ID NO: 42 shown in the following Table.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 antibody variable heavy chain comprising the amino acid sequence of SEQ ID NO: 43 shown in the following Table.
MDTLCSTLLL LTIPSWVLSQ ITLKESGPTL VKPKQTLTLT CTFSGFSLST SGMGVGWIRQ
The nucleic acid sequence encoding the human anti-OX40 antibody heavy chain variable region comprises the nucleic acid sequence of SEQ ID NO: 44 shown in the following Table.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 antibody variable light chain comprising the amino acid sequence of SEQ ID NO: 45 shown in the following Table.
METPAQLLFL LLLWLPDTTG EIVLTQSPGT LSLSPGERAT LSCRASQSVS SSYLAWYQQK
The nucleic acid sequence encoding the human anti-OX40 antibody light chain variable region comprises the nucleic acid sequence of SEQ ID NO: 46 shown in the following Table.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 antibody variable heavy chain comprising the amino acid sequence of SEQ ID NO: 47 shown in the following Table.
MDTLCSTLLL LTIPSWVLSQ ITLKESGPTL VKPTQTLTLS CTFSGFSLST SGVGVGWIRQ
The nucleic acid sequence encoding the human anti-OX40 antibody heavy chain variable region comprises the nucleic acid sequence of SEQ ID NO: 48 shown in the following Table.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 antibody variable light chain comprising the amino acid sequence of SEQ ID NO: 49 shown in the following Table.
METPAQLLFL LLLWLPDTTG EIVLTQSPGT LSLSPGERAI LSCRASQSVS SSFLAWYQQK
The nucleic acid sequence encoding the human anti-OX40 antibody light chain variable region comprises the nucleic acid sequence of SEQ ID NO: 50 shown in the following Table.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody variable heavy chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 IgG2a antibody heavy chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody variable light chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise the nucleic acid encoding an anti-OX40 IgG2a antibody light chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody variable heavy chain amino acid sequence, and a mouse anti-OX40 antibody variable light chain amino acid sequence, that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain amino acid sequence, and an anti-OX40 antibody light chain amino acid sequence, that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 variable heavy chain comprising the amino acid sequence of SEQ ID NO: 5. The nucleic acid sequence encoding the anti-OX40 variable heavy chain is SEQ ID NO: 6. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 variable light chain comprising the amino acid sequence of SEQ ID NO: 7. The nucleic acid sequence encoding the anti-OX40 variable light chain is SEQ ID NO: 8.
The Sindbis viral vector can comprise a nucleic acid encoding a mouse anti-OX40 heavy chain of amino acid sequence of SEQ ID NO; 5 and a nucleic acid encoding a mouse anti-OX40 light chain of amino acid sequence of SEQ ID NO: 7. The Sindbis viral vector can comprise a nucleic acid encoding a mouse anti-OX40 heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to amino acid sequence of SEQ ID NO: 5 and a nucleic acid encoding a mouse anti-OX40 light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to amino acid sequence of SEQ ID NO: 7.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain comprising the amino acid sequence of SEQ ID NO: 9. The nucleic acid sequence encoding the an anti-OX40 antibody heavy chain is SEQ ID NO: 10.
The Sindbis viral vector can comprise a nucleic acid encoding a mouse anti-OX40 antibody light chain comprising the amino acid sequence of SEQ ID No: 11. The nucleic acid sequence encoding the anti-OX40 antibody light chain is SEQ ID NO: 12.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence of SEQ ID NO: 9 and an anti-OX40 antibody light chain with an amino acid sequence of SEQ ID NO: 11. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain of SEQ ID NO: 10 and an anti-OX40 antibody light chain with an amino acid sequence of SEQ ID NO: 12.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 9 and an anti-OX40 antibody light chain with an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 11. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 10 and an anti-OX40 antibody light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 12.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence of SEQ ID NO: 31. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain is SEQ ID NO: 32. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence of SEQ ID NO: 33. The nucleic acid sequence encoding the human anti-OX40 variable light chain is SEQ ID NO: 34.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 31. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to is SEQ ID NO: 32. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 33. The nucleic acid sequence encoding the human anti-OX40 variable light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 34.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence of SEQ ID NO: 35. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain is SEQ ID NO: 36. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence of SEQ ID NO: 37. The nucleic acid sequence encoding the human anti-OX40 variable light chain is SEQ ID NO: 38.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 35. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to is SEQ ID NO: 36. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 37. The nucleic acid sequence encoding the human anti-OX40 variable light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 38.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence of SEQ ID NO: 39. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain is SEQ ID NO: 40. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence of SEQ ID NO: 41. The nucleic acid sequence encoding the human anti-OX40 variable light chain is SEQ ID NO: 42.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 39. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to is SEQ ID NO: 40. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 41. The nucleic acid sequence encoding the human anti-OX40 variable light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 42.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence of SEQ ID NO: 43. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain is SEQ ID NO: 44. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence of SEQ ID NO: 45. The nucleic acid sequence encoding the human anti-OX40 variable light chain is SEQ ID NO: 46.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 43. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to is SEQ ID NO: 44. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 45. The nucleic acid sequence encoding the human anti-OX40 variable light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 46.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence of SEQ ID NO: 47. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain is SEQ ID NO: 48. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence of SEQ ID NO: 49. The nucleic acid sequence encoding the human anti-OX40 variable light chain is SEQ ID NO: 50.
The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable heavy chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 47. The nucleic acid sequence encoding the human anti-OX40 variable heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to is SEQ ID NO: 48. The Sindbis viral vector can comprise a nucleic acid encoding a human anti-OX40 variable light chain comprising the amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 49. The nucleic acid sequence encoding the human anti-OX40 variable light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 50.
The Sindbis viral vector can comprise the nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody variable heavy chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody variable light chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise the nucleic acid encoding an anti-OX40 antibody light chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody variable heavy chain amino acid sequence, and an anti-OX40 antibody variable light chain amino acid sequence, that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain amino acid sequence, and an anti-OX40 antibody light chain amino acid sequence, that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13.
The Sindbis viral vector and the anti-OX40 monoclonal antibody can be administered sequentially or concurrently. The Sindbis viral vector can be administered systemically. The anti-OX40 monoclonal antibody can be administered systemically. Both the Sindbis viral vector and the anti-OX40 monoclonal antibody, or a nucleic acid encoding same, can be administered systemically. The Sindbis viral vector can be administered parenterally. The anti-OX40 monoclonal antibody can be administered parenterally. Both the Sindbis viral vector and the anti-OX40 monoclonal antibody, or a nucleic acid encoding same, can be administered parenterally. The Sindbis viral vector can be administered intraperitoneally. The anti-OX40 monoclonal antibody can be administered intraperitoneally. Both the Sindbis viral vector and the anti-OX40 monoclonal antibody, or a nucleic acid encoding same, can be administered intraperitoneally.
A “monoclonal antibody” as disclosed herein, can be a full-length antibody or an antigen binding fragment thereof, wherein the “antigen binding fragment” is a fragment of the full length antibody that retains binding to the target antigen of the said monoclonal antibody. For example, a monoclonal antibody against OX-40 or an anti-OX40 monoclonal antibody, as described herein, can be a full length antibody against OX40 antibody or an antigen binding fragment thereof, wherein the fragment retains binding to the OX40 receptor on a cell surface. An “antigen-binding fragment” of an anti-OX-40 antibody, as described herein can include any fragment selected from the group consisting of Fv, Fav, F(ab′)2, Fab′, dsFv, scFv, sc(Fv)2, scFv-CH3, scFv-Fc, and diabody fragments.
The cancer can be a solid cancer or a liquid/hematologic cancer. The cancer can comprise metastatic cancer. The cancer can comprise a solid tumor. The cancer can be a carcinoma, a lymphoma, a blastoma, a sarcoma, a leukemia, a brain cancer, a breast cancer, a blood cancer, a bone cancer, a lung cancer, a skin cancer, a liver cancer, an ovarian cancer, a bladder cancer, a renal cancer, a gastric cancer, a thyroid cancer, a pancreatic cancer, an esophageal cancer, a prostate cancer, a cervical cancer or a colorectal cancer. In one preferred aspect, the cancer is colon cancer. In one preferred aspect, the cancer is prostate cancer. In one preferred aspect, the cancer is ovarian cancer.
Sindbis Viral Vector and Anti-OX40 Monoclonal Antibody
The present disclosure further provides a Sindbis viral vector comprising a nucleic acid encoding interleukin-12 and a nucleic acid encoding an anti-OX40 monoclonal antibody. The present disclosure also provides a composition comprising (a) a Sindbis viral vector comprising a nucleic acid encoding interleukin-12 and (b) an anti-OX40 monoclonal antibody or a nucleic acid encoding same. The present disclosure also provides a composition comprising (a) a Sindbis viral vector comprising a nucleic acid encoding interleukin-12 and (b) a Sindbis viral vector comprising a nucleic acid encoding an anti-OX40 monoclonal antibody.
The Sindbis viral vector can comprise the nucleic acid encoding interleukin-12 alpha subunit (IL-12 α, IL-12, p35 subunit) and interleukin-12 beta subunit (IL-12 β, IL-12, p40 subunit). The Sindbis viral vector can comprise the nucleic acid encoding interleukin-12 alpha subunit of GenBank accession no. M86672 and interleukin-12 beta subunit of GenBank accession no. M86671. The nucleic acid encoding interleukin-12 alpha subunit comprises the nucleic acid sequence of SEQ ID NO: 1. The nucleic acid encoding interleukin-12 beta subunit comprises the nucleic acid sequence of SEQ ID NO: 2.
The Sindbis viral vector can comprise the nucleic acid encoding interleukin-12 alpha subunit of amino acid sequence of GenBank accession no. AAA39292.1 and interleukin-12 beta subunit of amino acid sequence of GenBank accession no. AAA39296.1. The amino acid sequence of the interleukin-12 alpha subunit is of amino acid sequence of SEQ ID NO: 3. The amino acid sequence of the interleukin-12 beta subunit is of amino acid sequence of SEQ ID NO: 4.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 variable heavy chain comprising the amino acid sequence of SEQ ID NO: 5. The nucleic acid sequence encoding the anti-OX40 variable heavy chain is SEQ ID NO: 6. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 variable light chain comprising the amino acid sequence of SEQ ID NO: 7. The nucleic acid sequence encoding the anti-OX40 variable light chain is SEQ ID NO: 8.
The Sindbis viral vector can comprise a nucleic acid encoding a mouse anti-OX40 heavy chain of amino acid sequence of SEQ ID NO; 5 and a nucleic acid encoding a mouse anti-OX40 light chain of amino acid sequence of SEQ ID NO: 7. The Sindbis viral vector can comprise a nucleic acid encoding a mouse anti-OX40 heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to amino acid sequence of SEQ ID NO: 5 and a nucleic acid encoding a mouse anti-OX40 light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to amino acid sequence of SEQ ID NO: 7.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain comprising the amino acid sequence of SEQ ID NO: 9. The nucleic acid sequence encoding the an anti-OX40 antibody heavy chain is SEQ ID NO: 10.
The Sindbis viral vector can comprise a nucleic acid encoding a mouse anti-OX40 antibody light chain comprising the amino acid sequence of SEQ ID No: 11. The nucleic acid sequence encoding the anti-OX40 antibody light chain is SEQ ID NO: 12.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence of SEQ ID NO: 9 and an anti-OX40 antibody light chain with an amino acid sequence of SEQ ID NO: 11. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain of SEQ ID NO: 10 and an anti-OX40 antibody light chain with an amino acid sequence of SEQ ID NO: 12.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 9 and an anti-OX40 antibody light chain with an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 11. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 10 and an anti-OX40 antibody light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 12.
The Sindbis viral vector can comprise the nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody variable heavy chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody variable light chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise the nucleic acid encoding an anti-OX40 antibody light chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody variable heavy chain amino acid sequence, and an anti-OX40 antibody variable light chain amino acid sequence, that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain amino acid sequence, and an anti-OX40 antibody light chain amino acid sequence, that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13.
The present disclosure also provides methods for treating cancer in a subject in need thereof comprising administering to the subject a therapeutically effective amount of (a) a Sindbis viral vector comprising a nucleic acid encoding interleukin-12 and (b) an anti-OX40 monoclonal antibody or a nucleic acid encoding same, thereby treating cancer in the subject.
The Sindbis viral vector can be replication defective. The Sindbis viral vector can comprise the nucleic acid encoding interleukin-12 and can further comprise the nucleic acid encoding the anti-OX40 monoclonal antibody. The method can comprise administering a Sindbis viral vector comprising the nucleic acid encoding interleukin-12 and administering a Sindbis viral vector comprising the nucleic acid encoding the anti-OX40 monoclonal antibody.
The Sindbis viral vector can comprise the nucleic acid encoding interleukin-12 alpha subunit (IL-12 α, IL-12, p35 subunit) and interleukin-12 beta subunit (IL-12 β, IL-12, p40 subunit). The Sindbis viral vector can comprise the nucleic acid encoding interleukin-12 alpha subunit of GenBank accession no. M86672 and interleukin-12 beta subunit of GenBank accession no. M86671. The nucleic acid encoding interleukin-12 alpha subunit comprises the nucleic acid sequence of SEQ ID NO: 1. The nucleic acid encoding interleukin-12 beta subunit comprises the nucleic acid sequence of SEQ ID NO: 2.
The Sindbis viral vector can comprise the nucleic acid encoding interleukin-12 alpha subunit of amino acid sequence of GenBank accession no. AAA39292.1 and interleukin-12 beta subunit of amino acid sequence of GenBank accession no. AAA39296.1. The amino acid sequence of the interleukin-12 alpha subunit is of amino acid sequence of SEQ ID NO: 3. The amino acid sequence of the interleukin-12 beta subunit is of amino acid sequence of SEQ ID NO: 4.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 variable heavy chain comprising the amino acid sequence of SEQ ID NO: 5. The nucleic acid sequence encoding the anti-OX40 variable heavy chain is SEQ ID NO: 6. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 variable light chain comprising the amino acid sequence of SEQ ID NO: 7. The nucleic acid sequence encoding the anti-OX40 variable light chain is SEQ ID NO: 8.
The Sindbis viral vector can comprise a nucleic acid encoding a mouse anti-OX40 heavy chain of amino acid sequence of SEQ ID NO; 5 and a nucleic acid encoding a mouse anti-OX40 light chain of amino acid sequence of SEQ ID NO: 7. The Sindbis viral vector can comprise a nucleic acid encoding a mouse anti-OX40 heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to amino acid sequence of SEQ ID NO: 5 and a nucleic acid encoding a mouse anti-OX40 light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to amino acid sequence of SEQ ID NO: 7.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain comprising the amino acid sequence of SEQ ID NO: 9. The nucleic acid sequence encoding the an anti-OX40 antibody heavy chain is SEQ ID NO: 10.
The Sindbis viral vector can comprise a nucleic acid encoding a mouse anti-OX40 antibody light chain comprising the amino acid sequence of SEQ ID No: 11. The nucleic acid sequence encoding the anti-OX40 antibody light chain is SEQ ID NO: 12.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence of SEQ ID NO: 9 and an anti-OX40 antibody light chain with an amino acid sequence of SEQ ID NO: 11. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain of SEQ ID NO: 10 and an anti-OX40 antibody light chain with an amino acid sequence of SEQ ID NO: 12.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 9 and an anti-OX40 antibody light chain with an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 11. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 10 and an anti-OX40 antibody light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 12.
The Sindbis viral vector can comprise the nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody variable heavy chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody variable light chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise the nucleic acid encoding an anti-OX40 antibody light chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody variable heavy chain amino acid sequence, and an anti-OX40 antibody variable light chain amino acid sequence, that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain amino acid sequence, and an anti-OX40 antibody light chain amino acid sequence, that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13.
A “monoclonal antibody” as disclosed herein, can be a full-length antibody or an antigen binding fragment thereof, wherein the “antigen binding fragment” is a fragment of the full length antibody that retains binding to the target antigen of the said monoclonal antibody. For example, a monoclonal antibody against OX-40 or an anti-OX40 monoclonal antibody, as described herein, can be a full length antibody against OX40 antibody or an antigen binding fragment thereof, wherein the fragment retains binding to the OX40 receptor on a cell surface. An “antigen-binding fragment” of an anti-OX-40 antibody, as described herein can include any fragment selected from the group consisting of Fv, Fav, F(ab′)2, Fab′, dsFv, scFv, sc(Fv)2, scFv-CH3, scFv-Fc, and diabody fragments.
The Sindbis viral vector and the anti-OX40 monoclonal antibody can be administered sequentially or concurrently. The Sindbis viral vector can be administered systemically. The anti-OX40 monoclonal antibody can be administered systemically. Both the Sindbis viral vector and the anti-OX40 monoclonal antibody, or a nucleic acid encoding same, can be administered systemically. The Sindbis viral vector can be administered parenterally. The anti-OX40 monoclonal antibody can be administered parenterally. Both the Sindbis viral vector and the anti-OX40 monoclonal antibody, or a nucleic acid encoding same, can be administered parenterally. The Sindbis viral vector can be administered intraperitoneally. The anti-OX40 monoclonal antibody can be administered intraperitoneally. Both the Sindbis viral vector and the anti-OX40 monoclonal antibody, or a nucleic acid encoding same, can be administered intraperitoneally.
The cancer can be a solid cancer or a liquid/hematologic cancer. The cancer can comprise metastatic cancer. The cancer can comprise a solid tumor. The cancer can be a carcinoma, a lymphoma, a blastoma, a sarcoma, a leukemia, a brain cancer, a breast cancer, a blood cancer, a bone cancer, a lung cancer, a skin cancer, a liver cancer, an ovarian cancer, a bladder cancer, a renal cancer, a gastric cancer, a thyroid cancer, a pancreatic cancer, an esophageal cancer, a prostate cancer, a cervical cancer or a colorectal cancer. In one preferred aspect, the cancer is colon cancer. In one preferred aspect, the cancer is prostate cancer. In one preferred aspect, the cancer is ovarian cancer.
Sindbis virus can be administered at least one time, at least two times, at least three times, at least four times or at least five times per week. Sindbis virus can be administered for at least one week, at least two weeks, at least three weeks, or at least four weeks. Sindbis virus can be administered from 106-109 TU/mL. Preferably, Sindbis virus can be administered from 106-109 TU/mL.
An anti-OX40 monoclonal antibody can be administered at least one time, at least two times, at least three times, at least four times or at least five times per week. An anti-OX40 monoclonal antibody can be administered for at least one week, at least two weeks, at least three weeks, or at least four weeks. An anti-OX40 monoclonal antibody can be administered from 25 μg-500 μg, 25 μg-450 μg, 50 μg-400 μg, from 50 μg-350 μg, from 50 μg-300 μg, from 50 μg-250 μg, from 50 μg-200 μg, from 50 μg-150 μg or from 50 μg-100 μg. An anti-OX40 monoclonal antibody can be administered at 250 μg. An anti-OX40 monoclonal antibody can be administered at 250 μg once a week for one week. An anti-OX40 monoclonal antibody can be administered at 250 μg once a week for two weeks. An anti-OX40 monoclonal antibody can be administered at 250 μg once a week for three weeks. An anti-OX40 monoclonal antibody can be administered at 250 μg three times a week for one week. An anti-OX40 monoclonal antibody can be administered at 250 μg three times a week for two weeks. An anti-OX40 monoclonal antibody can be administered at 250 μg three times a week for three weeks.
The results provided in the instant disclosure demonstrate that administration of the a Sindbis virus expressing IL-12 (SV.IL12) markedly increases the expression of OX40 on CD4 T cells and demonstrate that administration of a combination of SV.IL12 and anti-OX40 monoclonal antibody resulted in complete tumor regression in colon cancer, prostate cancer and ovarian cancer in vivo models and led to a greater than 60% survival rate (in some instances to a greater than 90% survival rate). This combined therapeutic effect was dramatically more effective when compared to either SV.IL12 or anti-OX40 monoclonal antibody treatment alone. These results also confirm that the oncolytic activity of the Sindbis virus is not required to induce a robust and effective anti-tumor response.
The results provided in the instant disclosure demonstrate that the combination of SV.IL12 or anti-OX40 monoclonal antibody treatment markedly changes the transcriptome signature of T cells and favors the differentiation of terminal effector T cells (e.g., effector T cells with a Th1 type phenotype). In particular, pathways upregulated by the combination treatment were dominated by DNA replication, chromosomal organization and cell cycle regulation, but also included various metabolic and immunological processes, such as mitochondrial respiration, nucleotide metabolism and adaptive immune responses. Specifically, only T cells from combined therapy expressed the gene signature of terminally differentiated effector T cells, which are characterized by high expression of the killer lectin-like receptor (KLRG1) and low expression of the interleukin 7 receptor (IL-7R). Furthermore, genes encoding products associated with the differentiation and function of effector cells, such as Batf Id2, Tbet, Gzmb and Ifng, were also highly expressed in T cells following combination therapy. Furthermore, CD4 T cells also expressed a marked anti-tumor effector phenotype (ICOS*Tbet*) which was on average 2 to 3-fold higher during combined therapy compared with SV.IL12 or anti-OX40 treatment.
The tumor microenvironment can be a very challenging milieu for an effector T cell as it is characterized by hypoxia, acidosis and low levels of nutrient sources such as glucose and glutamine. Even if T cell activation and initiation of effector function is allowed, T cells may be unable to generate the bioenergetics intermediates necessary to carry out effector function in the tumor microenvironment. Thus, providing a metabolic support for T cells is crucial for the success of cancer treatments. The results provided in the instant disclosure demonstrate that the combination of SV.IL12 or anti-OX40 monoclonal antibody promotes metabolic reprogramming of T cells. Specifically, the basal rate of oxygen consumption (OCR) was enhanced and spare respiratory capacity was dramatically increased in T cells following combination treatment. The combination also induced elevated protein expression of c-MYC as well as rate of extracellular acidification (ECAR). Collectively, these results show that SV.IL12 induces enhanced oxidative phosphorylation in CD8 T cells and the combination treatment is required to push CD4 T cells towards glycolysis by increasing the protein expression of c-MYC. Thus, the combination of SV.IL12 or anti-OX40 monoclonal antibody metabolically rewires T cells to an energetic state using both metabolic pathways, oxidative phosphorylation and glycolysis.
The results provided in the instant disclosure demonstrate that metabolic reprogrammed T cells display enhanced CD4 mediated cytokine production and anti-tumor activity following treatment with the combination of SV.IL12 and anti-OX40 monoclonal antibody. Specifically, genes encoding pro-inflammatory cytokines ifng and il2 were upregulated in T cells and the secretion of interferon-γ (IFNγ) by splenocytes was increased following combination treatment. Additional, the levels of the cytotoxic proteases, granzyme A and B, were upregulated following combination treatment. Further, granzyme B positive cells were detected in CD8 as well as CD4 T cells, indicating the presence of cytotoxic CD4 T cells following combination treatment. In addition, tumor growth was markedly reduced when co-cultured with splenocytes from mice receiving combined therapy. Surprisingly, tumor growth inhibition was mediated by CD4 T cells. Together, these results clearly show that T cells from combined therapy elicit enhanced anti-tumor and functional activity, such as granzyme B and IFNγ production driven by CD4 T cells.
The results provided in the instant disclosure demonstrate that treatment with the combination of SV.IL12 and anti-OX40 monoclonal antibody results in enhanced T cell migration and intratumoral T cell immunity. Specifically, CXCR3 levels were significantly upregulated on CD4 T cells following combination therapy. In contrast, CXCR3 expression on CD8 T cells only appeared later on in treatment, indicating that CD4 T cells are first recruited to the inflamed site followed by CD8 T cells. Combination therapy also enhanced the production of CXCL9 and CXCL10 in the tumor microenvironment, indicating that CXCR3 positive T cells migrate to the tumor site. These results clearly show that the combination of SV.IL12 and anti-OX40 monoclonal antibody alter the tumor microenvironment by facilitating T cell infiltration via modulation of the CXCR3/CXCL9-11 axis. Not only did combination therapy increase T cell infiltration but CD4 as well as CD8 T cells also demonstrated enhanced functional activity in the tumor, as judged by the Ki-67 and granzyme B expression. These results indicate that the presence of activated T cells in the tumor microenvironment exert anti-tumor activity which inhibits tumor growth. Enhanced iNOS production was also demonstrated in tumors treated with combination therapy. Interestingly, the amount of iNOS inversely correlated with arginase1 production, indicating a repolarization of tumor associated macrophages from the M2-like (pro-tumor) into Ml-like (anti-tumor) phenotype during combination therapy.
Thus, the data provided herein clearly shows that even in absence of direct Sindbis virus infectivity, SV.IL12 in combination with an anti-OX40 monoclonal antibody alter the tumor microenvironment by enhancing T cell infiltration and intratumoral T cell immunity, especially against low immunogenic tumors. The synergistic therapeutic efficacy of the systemic administration of the combination is driven by T cell modulation and reprogramming of its metabolic state, in order to enhance the anti-tumor response in the periphery and in the tumor microenvironment. Furthermore, the use of Sindbis virus allows these metabolically reprogrammed T cells to better infiltrate the tumor microenvironment, which is crucial for an adequate immunotherapy.
Sindbis Viral Vector and Anti-4-1BB Monoclonal Antibody
The present disclosure also provides methods for treating cancer in a subject in need thereof comprising administering to the subject a therapeutically effective amount (a) a Sindbis viral vector and (b) an anti-4-1BB monoclonal antibody or a nucleic acid encoding same, thereby treating cancer in the subject. The present disclosure further provides in vitro or ex vivo methods for treating cancer or assessing the treatment of cancer in a subject comprising contacting a biological sample from the subject with (a) a Sindbis viral vector and (b) an anti-4-1BB monoclonal antibody or a nucleic acid encoding same. Preferably, the Sindbis viral vector does not comprise an endogenous nucleic acid encoding any protein. Sindbis viral vectors were produced as described in U.S. Pat. No. 8,093,021 (incorporated herein by reference in its entirety).
The Sindbis viral vector is replication defective. The Sindbis viral vector can comprise a nucleic acid sequence encoding a therapeutic protein, an immunostimulatory protein or an immunomodulatory protein. The Sindbis viral vector can comprise the nucleic acid encoding the therapeutic protein, an immunostimulatory protein or an immunomodulatory protein and further comprise the nucleic acid encoding the anti-4-1BB monoclonal antibody. The Sindbis viral vector can comprise a nucleic acid sequence encoding LacZ (lac operon structural gene lacZ encoding β-galactosidase), Flue (firefly luciferase) or GFP (green fluorescent protein). The Sindbis viral vector can comprise the nucleic acid encoding LacZ, Flue or GFP and further comprise the nucleic acid encoding the anti-4-1BB monoclonal antibody.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-4-1BB antibody heavy chain comprising the heavy chain complementarity determining region 1 (HCDR1), HCDR2 and HCDR3 amino acid sequences as follows: HCDR1: GFIFSYFDMA (SEQ ID NO: 16), HCDR2: SISPDGSIPYYRDSVK (SEQ ID NO: 17) and HCDR3: RSYGGYSELDY (SEQ ID NO: 18).
The Sindbis viral vector can comprise a nucleic acid encoding an anti-4-1BB heavy chain comprising the amino acid sequence of SEQ ID NO:19 shown in the following Table.
ISPDGSIPYYRDSVKGRFTVSRENAKSSLYLQMDSLRSEDTATYYCARRS
YGGYSELDYWGQGVMVTVSS.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-4-1BB antibody light chain comprising the light chain complementarity determining region 1 (LCDR1), LCDR2 and LCDR3 amino acid sequences as follows: LCDR1: QASQDIGNWLA (SEQ ID NO: 20), LCDR2: GSTSLAD (SEQ ID NO: 21) and LCDR3: LQAYGAPW (SEQ ID NO: 22).
The Sindbis viral vector can comprise a nucleic acid encoding an anti-4-1BB light chain comprising the amino acid sequence of SEQ ID NO:23 shown in the following Table.
The Sindbis viral vector can comprise a nucleic acid encoding anti-4-1BB antibody heavy chain CDR1, CDR2 and CDR3 amino acid sequences that binds to a target antigen comprising the amino acid sequence of SEQ ID NO:24 shown in the following Table.
The Sindbis viral vector can comprise a nucleic acid encoding a 4-1BB heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to amino acid sequence of SEQ ID NO: 19 and a nucleic acid encoding a 4-1BB light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to amino acid sequence of SEQ ID NO: 23.
The Sindbis viral vector can comprise the nucleic acid encoding anti-4-1BB antibody light chain CDR1, CDR2 and CDR3 amino acid sequences that binds to a target antigen (4-1BB antigen) of the amino acid sequence of (SEQ ID NO: 24). The Sindbis viral vector can comprise the nucleic acid encoding anti-4-1BB antibody heavy chain amino acid sequence that binds to a target antigen (4-1BB antigen) of the amino acid sequence of (SEQ ID NO: 24). The Sindbis viral vector can comprise the nucleic acid encoding anti-4-1BB antibody light chain amino acid sequence that binds to a target antigen (4-1BB antigen) of the amino acid sequence of (SEQ ID NO: 24).
The Sindbis viral vector can comprise a nucleic acid encoding an anti-4-1BB antibody heavy chain comprising the heavy chain complementarity determining region 1 (HCDR1), HCDR2 and HCDR3 amino acid sequences of SEQ ID NOs: 16, 17 and 18, respectively. The Sindbis viral vector can comprise the nucleic acid encoding an anti-4-1BB antibody heavy chain comprising the amino acid sequence of SEQ ID NO: 19. The Sindbis viral vector can comprise a nucleic acid encoding an anti-4-1BB antibody heavy chain comprising the light chain complementarity determining region 1 (LCDR1), LCDR2 and LCDR3 amino acid sequences of SEQ ID NOs: 20, 21 and 22, respectively. The Sindbis viral vector can comprise the nucleic acid encoding an anti-4-1BB antibody light chain comprising the amino acid sequence of SEQ ID NO: 23.
The Sindbis viral vector can comprise the nucleic acid encoding anti-4-1BB antibody heavy chain CDR1, CDR2 and CDR3 amino acid sequences that binds to a target antigen of the amino acid sequence of SEQ ID NO: 24. The Sindbis viral vector can comprise the nucleic acid encoding anti-4-1BB antibody light chain CDR1, CDR2 and CDR3 amino acid sequences that binds to a target antigen of the amino acid sequence of SEQ ID NO: 24. The Sindbis viral vector can comprise the nucleic acid encoding anti-4-1BB antibody heavy chain amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 24. The Sindbis viral vector can comprise the nucleic acid encoding anti-4-1BB antibody light chain amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 24.
The immunostimulatory or immunomodulatory protein can be IL-1, IL-2, IL-3, IL-4, IL-5, IL-6 IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, IL-19, IL-20, IL-21, IL-22, IL-23, IL-24, IL-25, IL-26, IL-27, IL-28, IL-29, IL-30, IL-31, IL-32, IL-33, IL-34, IL-35, IL-36, or any combination thereof. In a preferred aspect, the immunostimulatory or immunomodulatory protein is IL-12. The anti-4-1BB antibody can be urelumab, utomilumab or a combination thereof. The anti-4-1BB antibody can be InVivoMAb anti-mouse 4-1BB (BioXCell, Clone: LOB12.3, Cat. No. BE0169).
The Sindbis viral vector and the anti-4-1BB monoclonal antibody can be administered sequentially or concurrently. The Sindbis viral vector can be administered systemically. The anti-4-1BB monoclonal antibody can be administered systemically. Both the Sindbis viral vector and the anti-4-1BB monoclonal antibody, or a nucleic acid encoding same, can be administered systemically. The Sindbis viral vector can be administered parenterally. The anti-4-1BB monoclonal antibody can be administered parenterally. Both the Sindbis viral vector and the anti-4-1BB monoclonal antibody, or a nucleic acid encoding same, can be administered parenterally. The Sindbis viral vector can be administered intraperitoneally. The anti-4-1BB monoclonal antibody can be administered intraperitoneally. Both the Sindbis viral vector and the anti-4-1BB monoclonal antibody, or a nucleic acid encoding same, can be administered intraperitoneally.
A “monoclonal antibody” as disclosed herein, can be a full-length antibody or an antigen binding fragment thereof, wherein the “antigen binding fragment” is a fragment of the full length antibody that retains binding to the target antigen of the said monoclonal antibody. For example, a monoclonal antibody against 4-1BB or an anti-4-1BB monoclonal antibody, as described herein, can be a full length antibody against 4-1BB antibody or an antigen binding fragment thereof, wherein the fragment retains binding to the 4-1BB receptor on a cell surface. An “antigen-binding fragment” of an anti-4-1BB antibody, as described herein can include any fragment selected from the group consisting of Fv, Fav, F(ab′)2, Fab′, dsFv, scFv, sc(Fv)2, scFv-CH3, scFv-Fc, and diabody fragments.
The cancer can be a solid cancer or a liquid/hematologic cancer. The cancer can comprise metastatic cancer. The cancer can comprise a solid tumor. The cancer can be a carcinoma, a lymphoma, a blastoma, a sarcoma, a leukemia, a brain cancer, a breast cancer, a blood cancer, a bone cancer, a lung cancer, a skin cancer, a liver cancer, an ovarian cancer, a bladder cancer, a renal cancer, a gastric cancer, a thyroid cancer, a pancreatic cancer, an esophageal cancer, a prostate cancer, a cervical cancer or a colorectal cancer. In one preferred aspect, the cancer is a lymphoma. In one preferred aspect, the cancer is a B cell lymphoma.
The present disclosure provides a Sindbis viral vector comprising a nucleic acid encoding a therapeutic protein, an immunostimulatory protein or an immunomodulatory protein and a nucleic acid encoding an anti-4-1BB monoclonal antibody. The present disclosure provides a Sindbis viral vector comprising a nucleic acid encoding LacZ, Flue or GFP and a nucleic acid encoding an anti-4-1BB monoclonal antibody. The present disclosure also provides a composition comprising (a) a Sindbis viral vector comprising a nucleic acid encoding a therapeutic protein, an immunostimulatory protein or an immunomodulatory protein and (b) a Sindbis viral vector comprising a nucleic acid encoding an anti-4-1BB monoclonal antibody. The present disclosure also provides a composition comprising (a) a Sindbis viral vector comprising a nucleic acid encoding LacZ, Flue or GFP and (b) a Sindbis viral vector comprising a nucleic acid encoding an anti-4-1BB monoclonal antibody. The present disclosure further provides a composition comprising (a) a Sindbis viral vector comprising a nucleic acid encoding a therapeutic protein, an immunostimulatory protein or an immunomodulatory protein and (b) an anti-4-1BB monoclonal antibody or a nucleic acid encoding same. The present disclosure provides a composition comprising (a) a Sindbis viral vector comprising a nucleic acid encoding encoding LacZ, Flue or GFP and (b) an anti-4-1BB monoclonal antibody or a nucleic acid encoding same.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-4-1 in antibody heavy chain comprising the heavy chain complementarity determining region 1 (HCDR1), HCDR2 and HCDR3 amino acid sequences of SEQ ID NOs: 16, 17 and 18, respectively. The Sindbis viral vector can comprise the nucleic acid encoding an anti-4-1BB antibody heavy chain comprising the amino acid sequence of SEQ ID NO: 19. The Sindbis viral vector can comprise a nucleic acid encoding an anti-4-1BB antibody heavy chain comprising the light chain complementarity determining region 1 (LCDR1), LCDR2 and LCDR3 amino acid sequences of SEQ ID NOs: 20, 21 and 22, respectively. The Sindbis viral vector can comprise the nucleic acid encoding an anti-4-1BB antibody light chain comprising the amino acid sequence of SEQ ID NO: 23.
The Sindbis viral vector can comprise the nucleic acid encoding anti-4-1BB antibody heavy chain CDR1, CDR2 and CDR3 amino acid sequences that binds to a target antigen of the amino acid sequence of SEQ ID NO: 24. The Sindbis viral vector can comprise the nucleic acid encoding anti-4-1BB antibody light chain CDR1, CDR2 and CDR3 amino acid sequences that binds to a target antigen of the amino acid sequence of SEQ ID NO: 24. The Sindbis viral vector can comprise the nucleic acid encoding anti-4-1BB antibody heavy chain amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 24. The Sindbis viral vector can comprise the nucleic acid encoding anti-4-1BB antibody light chain amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 24.
The immunostimulatory or immunomodulatory protein can be IL-1, IL-2, IL-3, IL-4, IL-5, IL-6 IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, IL-19, IL-20, IL-21, IL-22, IL-23, IL-24, IL-25, IL-26, IL-27, IL-28, IL-29, IL-30, IL-31, IL-32, IL-33, IL-34, IL-35, IL-36, or any combination thereof. In a preferred aspect, the immunostimulatory or immunomodulatory protein is IL-12. Additional cytokines include IL-I8-IL-36. In addition to CCL17, other chemokines can also be used, including, but not limited to, CCL1-CCL27 and other CC chemokines, CXCLI-CXCL13 and other CXC chemokines, C chemokines, and CX3C chemkines. Cytokine or chemokine receptors and soluble receptors can also be used. Additional immune modulators that can be used include TGF-β and TNFα. In addition, different combinations of the above-mentioned (or alternative) cytokines can be used.
A “monoclonal antibody” as disclosed herein, can be a full-length antibody or an antigen binding fragment thereof, wherein the “antigen binding fragment” is a fragment of the full length antibody that retains binding to the target antigen of the said monoclonal antibody. For example, a monoclonal antibody against 4-1BB or an anti-4-1BB monoclonal antibody, as described herein, can be a full length antibody against 4-1BB antibody or an antigen binding fragment thereof, wherein the fragment retains binding to the 4-1BB receptor on a cell surface. An “antigen-binding fragment” of an anti-4-1BB antibody, as described herein can include any fragment selected from the group consisting of Fv, Fav, F(ab′)2, Fab′, dsFv, scFv, sc(Fv)2, scFv-CH3, scFv-Fc, and diabody fragments.
Sindbis virus can be administered at least one time, at least two times, at least three times, at least four times or at least five times per week. Sindbis virus can be administered for at least one week, at least two weeks, at least three weeks, or at least four weeks. Sindbis virus can be administered from 106-109 TU/mL. Preferably, Sindbis virus can be administered from 106-109 TU/mL.
An anti-4-1BB monoclonal antibody can be administered at least one time, at least two times, at least three times, at least four times or at least five times per week. An anti-4-1BB monoclonal antibody can be administered for at least one week, at least two weeks, at least three weeks, or at least four weeks. An anti-4-1BB monoclonal antibody can be administered from 25 μg-500 μg, 25 μg-450 μg, 50 μg-400 μg, from 50 μg-350 μg, from 50 μg-300 μg, from 50 μg-250 μg, from 50 μg-200 μg, from 50 μg-150 μg or from 50 μg-100 μg. An anti-4-1BB monoclonal antibody can be administered at 50 μg. An anti-4-1BB monoclonal antibody can be administered at 50 μg once a week for three weeks. An anti-4-1BB monoclonal antibody can be administered at 250 μg. An anti-4-1BB monoclonal antibody can be administered at 250 μg three times week for two weeks. An anti-4-1BB monoclonal antibody can be administered at 350 μg. An anti-4-1BB monoclonal antibody can be administered at 350 μg three times week for two weeks.
The results provided in the instant disclosure demonstrate that administration of the combination of Sindbis virus and anti-4-1BB monoclonal antibody resulted in complete tumor regression in an lymphoma in vivo model and that this therapeutic effect was dramatically more effective when compared to either Sindbis virus or anti-4-1BB monoclonal antibody treatment alone. Tumor elimination involves a synergistic effect of the combination that significantly boosts T cell cytotoxicity, IFNγ production, T cell proliferation, migration, and glycolysis. As described in more detail below, the data identified the molecular pathways, including upregulated cytokines, chemokines and metabolic pathways in T cells that are triggered by the combined therapy and help to achieve a highly effective anti-tumor response.
The results provided in the instant disclosure demonstrate that administration of the combination of Sindbis virus and anti-4-1BB monoclonal antibody resulted in increased T cell cycle progression, cytokine production and activation. T cell proliferation is critical for an effective anti-tumor response.
The results provided in the instant disclosure demonstrate that administration of the combination of Sindbis virus and anti-4-1BB monoclonal antibody resulted in increased cytotoxicity (e.g., increased cytotoxic T cell function). Specifically, genes such as Gzmb (granzyme B), Prfl (perforin) and Klrkl (NKG2D) are significantly upregulated in T cells (particularly CD8 T cells) following administration of Sindbis virus and anti-4-1BB monoclonal antibody.
The results provided in the instant disclosure demonstrate that administration of the combination of Sindbis virus and anti-4-1BB monoclonal antibody resulted in increased IFNγ production from T cells and Th1 differentiation. The combination of Sindbis virus and anti-4-1BB monoclonal antibody upregulated the expression of STAT4, Ccr5, Cxcr3, Havcr2(Tim3), IL12rbl and Klrcl in T cells, which are required for the development of Th1 cells from naïve CD4+ T cells and IFNγ production. This increase was independent of the presence or absence of TAA. The combination of Sindbis virus and anti-4-1BB monoclonal antibody increased IFNγ production from both CD4 and CD8 T cells (with a larger portion CD4 T cells producing IFNγ) and demonstrated that antigen presenting cells (APCs) are essenTh1tial for helping T cells product IFNγ. The combination of Sindbis virus and anti-4-1BB monoclonal antibody also increased T-bet in T cells. T-bet is a key transcription factor which is essential for type I immune response (IFNγ production, T cell cytotoxicity) and memory T cell differentiation. Thus, this indicates that the combination of Sindbis virus and anti-4-1BB monoclonal antibody boosts the type I immune response, which is critical for controlling tumor growth. The combination of Sindbis virus and anti-4-1BB monoclonal antibody also increased Eomesodermin (EOMES) in T cells. EOMES, another important transcription factor, is upregulated in activated T cells and is essential for memory CD8 T cell development.
The results provided in the instant disclosure demonstrate that administration of the combination of Sindbis virus and anti-4-1BB monoclonal antibody resulted in increased chemotaxis, adhesion and enhanced T cell infiltration and activation in tumors. Specifically, the combination significantly upregulates CD11a and ICAM-1(CD54) in both CD4 and CD8 T cells, which are two adhesion molecules expressed on activated T cells and are essential for the formation of immune synapses between T cells and APCs and are also required for T cell/T cell homotypic aggregation and activation. The combination of Sindbis virus and anti-4-1BB monoclonal antibody also significantly upregulated OX40 and ICOS in T cells. OX40 engagement promotes effector T cell function and survival and ICOS is another key CD4 T cell costimulatory molecule. Tumor infiltrating lymphocytes play a critical anti-tumor role and are an important marker for prognosis. The percentage of CD3 and CD8 T cells increased about two-fold following combination treatment. Thus, these results demonstrate that combination treatment enhanced T cell infiltration, division, activation, cytotoxicity and downregulated the inhibitory Treg population.
The results provided in the instant disclosure demonstrate that administration of the combination of Sindbis virus and anti-4-1BB monoclonal antibody resulted in enhanced T cell glycolysis and oxidative phosphorylation. T cell activation requires a quick consumption of energy through both enhanced glycolysis and oxidative phosphorylation. Metabolic switch is a major feature of T cell activation and memory T cell development. Upregulation of glycolysis genes quickly produce ATP and supports T cell migration and cytotoxicity in hypoxic or acidific microenvironments (such as in and around a tumor). The instant results demonstrate that combination treatment significantly increased both oxygen consumption rate (OCR, represents oxidative phosphorylation) and extracellular acidification rate (ECAR, represents glycolysis). This indicates that both glycolysis and oxidative phosphorylation are activated in combination treated T cells.
The results provided in the instant disclosure demonstrate that mice cured by the administration of the combination of Sindbis virus and anti-4-1BB monoclonal antibody are completely protected from cancer rechallenge demonstrating that these mice acquired long lasting antitumor immunity.
The conventional view of oncolytic virus therapy against tumors is that it requires selective infection of cancer cells resulting in the induction of cancer cell lysis and apoptosis. Tumor specific antigens (TAAs), released from dead tumor cells, attract and further stimulate an antitumor immune response. The data presented herein demonstrates that encoding a TAA is not necessary for the combination of Sindbis virus and anti-4-1BB monoclonal antibody to be fully successful in eradicating growing tumors.
The quick inhibition of tumor growth is critical for cancer therapy because tumor cells undergo exponentially rapid division. However, the induction of adaptive immunity and establishment of tumor specific immunity takes a long time. An ideal therapy requires an early, quick reduction of tumor burden, and a later induction of anti-tumor specificity that prevents relapse. The data presented herein demonstrates that the combination of Sindbis virus and anti-4-1BB monoclonal antibody treatment induced massive T cell activation due to viral induced immune response. This massive activation helps to control the tumor in a TAA nonspecific manner.
It was shown herein that both NKG2D (KLRKI) and granzyme B are highly expressed under combination treatment. This massive nonspecific activation is critical for controlling tumor growth at an early time point (day 7). This step is also important for inducing anti-tumor specificity that is mediated by TAAs released from dead tumor cells due to nonspecific killing. After tumor regression, T cells from treated animals were able maintain the ability to produce IFNγ and acquired immunological memory to rapidly reject tumor rechallenges. IFNγ production from purified T cells of cured mice was significantly enhanced after encountering tumor cells. This demonstrates that anti-tumor specificity is fully established in cured mice.
The data also shows that Sindbis viral infection of tumor cells, inclusion of dendtric cells and lymphodepletion are not necessary for successful cancer treatment. The omission of these additional features decreases costs, any risks related to toxicity and infection.
Thus, the data provided herein demonstrates that the combination of Sindbis virus and anti-4-1BB monoclonal antibody completely eradicated a B-cell lymphoma in a preclinical mouse model, a result that could not be achieved with either treatment alone. Tumor elimination involves a synergistic effect of the combination that significantly boosts T cell cytotoxicity, IFN-γ production, migration, tumor infiltration and oxidative phosphorylation. In addition, all mice that survived after treatment developed long lasting antitumor immunity.
Sindbis Viral Vector and Sindbis Viral Vector NY-ESO-1
The present disclosure also provides methods for treating cancer in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a Sindbis viral vector comprising a nucleic acid encoding interleukin-12 and a Sindbis viral vector comprising a nucleic acid encoding NY-ESO-1, thereby treating cancer in the subject. The present disclosure also provides methods for treating cancer in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a Sindbis viral vector comprising a nucleic acid encoding interleukin-12 and a nucleic acid encoding NY-ESO-1, thereby treating cancer in the subject.
The Sindbis viral vector can comprise the nucleic acid encoding interleukin-12 alpha subunit (IL-12 α, IL-12, p35 subunit) and interleukin-12 beta subunit (IL-12 β, IL-12, p40 subunit). The Sindbis viral vector can comprise the nucleic acid encoding interleukin-12 alpha subunit of GenBank accession no. M86672 and interleukin-12 beta subunit of GenBank accession no. M86671. The nucleic acid encoding interleukin-12 alpha subunit comprises the nucleic acid sequence of SEQ ID NO: 1. The nucleic acid encoding interleukin-12 beta subunit comprises the nucleic acid sequence of SEQ ID NO: 2.
The Sindbis viral vector can comprise the nucleic acid encoding interleukin-12 alpha subunit of amino acid sequence of GenBank accession no. AAA39292.1 and interleukin-12 beta subunit of amino acid sequence of GenBank accession no. AAA39296.1. The amino acid sequence of the interleukin-12 alpha subunit is of amino acid sequence of SEQ ID NO: 3. The amino acid sequence of the interleukin-12 beta subunit is of amino acid sequence of SEQ ID NO: 4.
The Sindbis viral vector can comprise the nucleic acid encoding NY-ESO-1 of NCBI Reference accession no. NM_001327.1. The nucleic acid encoding NY-ESO-1 comprises the nucleic acid sequence of SEQ ID NO:14 shown in the following Table.
The Sindbis viral vector can comprise the nucleic acid encoding NY-ESO-1 of amino acid sequence of NCBI Reference accession no. NP_001318.1. The amino acid sequence of the NY-ESO-1 comprises the amino acid sequence of SEQ ID NO:15 shown in the following Table.
The Sindbis viral vector can comprise the nucleic acid encoding NY-ESO-1 of NCBI Reference accession no. NM_001327.1. The nucleic acid encoding NY-ESO-1 comprises the nucleic acid sequence of SEQ ID NO: 14. The Sindbis viral vector can comprise the nucleic acid encoding NY-ESO-1 of amino acid sequence of NCBI Reference accession no. NP_001318.1. The amino acid sequence of the NY-ESO-1 comprises the amino acid sequence of SEQ ID NO: 15.
A replication defective Sindbis viral vector as described herein can be any replication defective Sindbis viral vector including a replication defective viral vector described, for example, in U.S. Pat. Nos. 7,303,898, 7,306,792, and 8,093,021. Replication defective vectors are preferred for use in the present invention in order to prevent infection of healthy tissues.
The Sindbis viral vector comprising a nucleic acid encoding interleukin-12 and the Sindbis viral vector comprising a nucleic acid encoding NY-ESO-1 can be administered sequentially or concurrently. The Sindbis viral vector comprising a nucleic acid encoding interleukin-12 can be administered systemically. The Sindbis viral vector comprising a nucleic acid encoding NY-ESO-1 can be administered systemically. Both the Sindbis viral vector comprising a nucleic acid encoding interleukin-12 and the Sindbis viral vector comprising a nucleic acid encoding NY-ESO-1 can be administered systemically. The Sindbis viral vector comprising a nucleic acid encoding interleukin-12 can be administered parenterally. The Sindbis viral vector comprising a nucleic acid encoding NY-ESO-1 can be administered parenterally. Both the Sindbis viral vector comprising a nucleic acid encoding interleukin-12 and the Sindbis viral vector comprising a nucleic acid encoding NY-ESO-1 can be administered parenterally. The Sindbis viral vector comprising a nucleic acid encoding interleukin-12 can be administered intraperitoneally. The Sindbis viral vector comprising a nucleic acid encoding NY-ESO-1 can be administered intraperitoneally. Both the Sindbis viral vector comprising a nucleic acid encoding interleukin-12 and the Sindbis viral vector comprising a nucleic acid encoding NY-ESO-1 can be administered intraperitoneally.
The cancer can be a solid cancer or a liquid/hematologic cancer. The cancer can comprise metastatic cancer. The cancer can comprise a solid tumor. The cancer can be a carcinoma, a lymphoma, a blastoma, a sarcoma, a leukemia, a brain cancer, a breast cancer, a blood cancer, a bone cancer, a lung cancer, a skin cancer, a liver cancer, an ovarian cancer, a bladder cancer, a renal cancer, a gastric cancer, a thyroid cancer, a pancreatic cancer, an esophageal cancer, a prostate cancer, a cervical cancer or a colorectal cancer. In one preferred aspect, the cancer is colon cancer. In one preferred aspect, the cancer is prostate cancer. In one preferred aspect, the cancer is ovarian cancer.
The present disclosure further provides a Sindbis viral vector comprising a nucleic acid encoding interleukin-12 and a nucleic acid encoding NY-ESO-1. The present disclosure also provides a composition comprising (a) a Sindbis viral vector comprising a nucleic acid encoding interleukin-12 and (b) a Sindbis viral vector comprising a nucleic acid encoding NY-ESO-1.
The Sindbis viral vector can comprise the nucleic acid encoding interleukin-12 alpha subunit (IL-12 α, IL-12, p35 subunit) and interleukin-12 beta subunit (IL-12 β, IL-12, p40 subunit). The Sindbis viral vector can comprise the nucleic acid encoding interleukin-12 alpha subunit of GenBank accession no. M86672 and interleukin-12 beta subunit of GenBank accession no. M86671. The nucleic acid encoding interleukin-12 alpha subunit comprises the nucleic acid sequence of SEQ ID NO: 1. The nucleic acid encoding interleukin-12 beta subunit comprises the nucleic acid sequence of SEQ ID NO: 2.
The Sindbis viral vector can comprise the nucleic acid encoding interleukin-12 alpha subunit of amino acid sequence of GenBank accession no. AAA39292.1 and interleukin-12 beta subunit of amino acid sequence of GenBank accession no. AAA39296.1. The amino acid sequence of the interleukin-12 alpha subunit is of amino acid sequence of SEQ ID NO: 3. The amino acid sequence of the interleukin-12 beta subunit is of amino acid sequence of SEQ ID NO: 4.
The Sindbis viral vector can comprise the nucleic acid encoding NY-ESO-1 of NCBI Reference accession no. NM_001327.1. The nucleic acid encoding NY-ESO-1 comprises the nucleic acid sequence of SEQ ID NO: 14. The Sindbis viral vector can comprise the nucleic acid encoding NY-ESO-1 of amino acid sequence of NCBI Reference accession no. NP_001318.1. The amino acid sequence of the NY-ESO-1 comprises the amino acid sequence of SEQ ID NO: 15.
The present disclosure provides methods for treating cancer in a subject in need thereof comprising administering to the subject a therapeutically effective amount of (a) a Sindbis viral vector comprising a nucleic acid encoding NY-ESO-1 and (b) an anti-OX40 monoclonal antibody or a nucleic acid encoding same, thereby treating cancer in the subject.
The Sindbis viral vector can be replication defective. The Sindbis viral vector can comprise the nucleic acid encoding NY-ESO-1 and can further comprise the nucleic acid encoding the anti-OX40 monoclonal antibody. The method can comprise administering a Sindbis viral vector comprising the nucleic acid encoding NY-ESO-1 and administering a Sindbis viral vector comprising the nucleic acid encoding the anti-OX40 monoclonal antibody. The Sindbis viral vector can comprise the nucleic acid encoding NY-ESO-1 of NCBI Reference accession no. NM_001327.1. The nucleic acid encoding NY-ESO-1 comprises the nucleic acid sequence of SEQ ID NO: 14. The Sindbis viral vector can comprise the nucleic acid encoding NY-ESO-1 of amino acid sequence of NCBI Reference accession no. NP_001318.1. The amino acid sequence of the NY-ESO-1 comprises the amino acid sequence of SEQ ID NO: 15.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 variable heavy chain comprising the amino acid sequence of SEQ ID NO: 5. The nucleic acid sequence encoding the anti-OX40 variable heavy chain is SEQ ID NO: 6. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 variable light chain comprising the amino acid sequence of SEQ ID NO: 7. The nucleic acid sequence encoding the anti-OX40 variable light chain is SEQ ID NO: 8.
The Sindbis viral vector can comprise a nucleic acid encoding a mouse anti-OX40 heavy chain of amino acid sequence of SEQ ID NO; 5 and a nucleic acid encoding a mouse anti-OX40 light chain of amino acid sequence of SEQ ID NO: 7. The Sindbis viral vector can comprise a nucleic acid encoding a mouse anti-OX40 heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to amino acid sequence of SEQ ID NO: 5 and a nucleic acid encoding a mouse anti-OX40 light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to amino acid sequence of SEQ ID NO: 7.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain comprising the amino acid sequence of SEQ ID NO: 9. The nucleic acid sequence encoding the an anti-OX40 antibody heavy chain is SEQ ID NO: 10.
The Sindbis viral vector can comprise a nucleic acid encoding a mouse anti-OX40 antibody light chain comprising the amino acid sequence of SEQ ID No: 11. The nucleic acid sequence encoding the anti-OX40 antibody light chain is SEQ ID NO: 12.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence of SEQ ID NO: 9 and an anti-OX40 antibody light chain with an amino acid sequence of SEQ ID NO: 11. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain of SEQ ID NO: 10 and an anti-OX40 antibody light chain with an amino acid sequence of SEQ ID NO: 12.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 9 and an anti-OX40 antibody light chain with an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 11. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 10 and an anti-OX40 antibody light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 12.
The Sindbis viral vector can comprise the nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody variable heavy chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody variable light chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise the nucleic acid encoding an anti-OX40 antibody light chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody variable heavy chain amino acid sequence, and an anti-OX40 antibody variable light chain amino acid sequence, that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain amino acid sequence, and an anti-OX40 antibody light chain amino acid sequence, that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13.
The Sindbis viral vector and the anti-OX40 monoclonal antibody can be administered sequentially or concurrently. The Sindbis viral vector can be administered systemically. The anti-OX40 monoclonal antibody can be administered systemically. Both the Sindbis viral vector and the anti-OX40 monoclonal antibody, or a nucleic acid encoding same, can be administered systemically. The Sindbis viral vector can be administered parenterally. The anti-OX40 monoclonal antibody can be administered parenterally. Both the Sindbis viral vector and the anti-OX40 monoclonal antibody, or a nucleic acid encoding same, can be administered parenterally. The Sindbis viral vector can be administered intraperitoneally. The anti-OX40 monoclonal antibody can be administered intraperitoneally. Both the Sindbis viral vector and the anti-OX40 monoclonal antibody, or a nucleic acid encoding same, can be administered intraperitoneally.
A “monoclonal antibody” as disclosed herein, can be a full-length antibody or an antigen binding fragment thereof, wherein the “antigen binding fragment” is a fragment of the full length antibody that retains binding to the target antigen of the said monoclonal antibody. For example, a monoclonal antibody against OX-40 or an anti-OX40 monoclonal antibody, as described herein, can be a full length antibody against OX40 antibody or an antigen binding fragment thereof, wherein the fragment retains binding to the OX40 receptor on a cell surface. An “antigen-binding fragment” of an anti-OX-40 antibody, as described herein can include any fragment selected from the group consisting of Fv, Fav, F(ab′)2, Fab′, dsFv, scFv, sc(Fv)2, scFv-CH3, scFv-Fc, and diabody fragments.
The cancer can be a solid cancer or a liquid/hematologic cancer. The cancer can comprise metastatic cancer. The cancer can comprise a solid tumor. The cancer can be a carcinoma, a lymphoma, a blastoma, a sarcoma, a leukemia, a brain cancer, a breast cancer, a blood cancer, a bone cancer, a lung cancer, a skin cancer, a liver cancer, an ovarian cancer, a bladder cancer, a renal cancer, a gastric cancer, a thyroid cancer, a pancreatic cancer, an esophageal cancer, a prostate cancer, a cervical cancer or a colorectal cancer. In one preferred aspect, the cancer is colon cancer. In one preferred aspect, the cancer is prostate cancer. In one preferred aspect, the cancer is ovarian cancer.
The present disclosure further provides a Sindbis viral vector comprising a nucleic acid encoding NY-ESO-1 and a nucleic acid encoding an anti-OX40 monoclonal antibody. The present disclosure also provides a composition comprising (a) a Sindbis viral vector comprising a nucleic acid encoding NY-ESO-1 and (b) an anti-OX40 monoclonal antibody or a nucleic acid encoding same. The present disclosure also provides a composition comprising (a) a Sindbis viral vector comprising a nucleic acid encoding NY-ESO-1 and (b) a Sindbis viral vector comprising a nucleic acid encoding an anti-OX40 monoclonal antibody. The Sindbis viral vector can comprise the nucleic acid encoding NY-ESO-1 of NCBI Reference accession no. NM_001327.1. The Sindbis viral vector can comprise the nucleic acid encoding NY-ESO-1 of NCBI Reference accession no. NM_001327.1. The nucleic acid encoding NY-ESO-1 comprises the nucleic acid sequence of SEQ ID NO: 14. The Sindbis viral vector can comprise the nucleic acid encoding NY-ESO-1 of amino acid sequence of NCBI Reference accession no. NP_001318.1. The amino acid sequence of the NY-ESO-1 comprises the amino acid sequence of SEQ ID NO: 15.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 variable heavy chain comprising the amino acid sequence of SEQ ID NO: 5. The nucleic acid sequence encoding the anti-OX40 variable heavy chain is SEQ ID NO: 6. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 variable light chain comprising the amino acid sequence of SEQ ID NO: 7. The nucleic acid sequence encoding the anti-OX40 variable light chain is SEQ ID NO: 8.
The Sindbis viral vector can comprise a nucleic acid encoding a mouse anti-OX40 heavy chain of amino acid sequence of SEQ ID NO; 5 and a nucleic acid encoding a mouse anti-OX40 light chain of amino acid sequence of SEQ ID NO: 7. The Sindbis viral vector can comprise a nucleic acid encoding a mouse anti-OX40 heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to amino acid sequence of SEQ ID NO: 5 and a nucleic acid encoding a mouse anti-OX40 light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to amino acid sequence of SEQ ID NO: 7.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain comprising the amino acid sequence of SEQ ID NO: 9. The nucleic acid sequence encoding the an anti-OX40 antibody heavy chain is SEQ ID NO: 10.
The Sindbis viral vector can comprise a nucleic acid encoding a mouse anti-OX40 antibody light chain comprising the amino acid sequence of SEQ ID No: 11. The nucleic acid sequence encoding the anti-OX40 antibody light chain is SEQ ID NO: 12.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence of SEQ ID NO: 9 and an anti-OX40 antibody light chain with an amino acid sequence of SEQ ID NO: 11. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain of SEQ ID NO: 10 and an anti-OX40 antibody light chain with an amino acid sequence of SEQ ID NO: 12.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 9 and an anti-OX40 antibody light chain with an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 11. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 10 and an anti-OX40 antibody light chain that is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 12.
The Sindbis viral vector can comprise the nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody variable heavy chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody variable light chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise the nucleic acid encoding an anti-OX40 antibody light chain with an amino acid sequence that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13.
The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody variable heavy chain amino acid sequence, and an anti-OX40 antibody variable light chain amino acid sequence, that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13. The Sindbis viral vector can comprise a nucleic acid encoding an anti-OX40 antibody heavy chain amino acid sequence, and an anti-OX40 antibody light chain amino acid sequence, that binds to a target antigen of the amino acid sequence of SEQ ID NO: 13.
A “monoclonal antibody” as disclosed herein, can be a full-length antibody or an antigen binding fragment thereof, wherein the “antigen binding fragment” is a fragment of the full length antibody that retains binding to the target antigen of the said monoclonal antibody. For example, a monoclonal antibody against OX-40 or an anti-OX40 monoclonal antibody, as described herein, can be a full length antibody against OX40 antibody or an antigen binding fragment thereof, wherein the fragment retains binding to the OX40 receptor on a cell surface. An “antigen-binding fragment” of an anti-OX-40 antibody, as described herein can include any fragment selected from the group consisting of Fv, Fav, F(ab′)2, Fab′, dsFv, scFv, sc(Fv)2, scFv-CH3, scFv-Fc, and diabody fragments.
The results provided in the instant disclosure demonstrate that administration of a combination of IL-12 and NY-ESO-1, expressed by separate Sindbis virus vector synergistically enhances the survival rate of a subject bearing an established tumor. The results described herein show that mice transplanted with Alm5-2Fluc-17 ovarian cancer cells by reinjection to establish tumor as depicted in
The results provided in the instant disclosure demonstrate that administration of a combination of IL-12 and NY-ESO-1, expressed by the same Sindbis virus vector synergistically enhances the survival rate of a subject bearing an established tumor. The results show that mice bearing established tumors of Alm5-2Fluc-17 ovarian cancer cells, when treated with a Sindbis viral vector that expresses both IL-12 and NYESO (SV-NYESO_SGP2_IL12), demonstrated synergistically enhanced survival as compared to mice treated with the SV-IL-12 or SV-NYESO. The results described herein clearly show the possibility of using a single SV vectors expressing both IL-12 and NY-ESO-1, for treatment of cancers that may be resistant to treatment with a SV expressing a tumor associated antigen.
Treating cancer means treating at least one symptom of cancer. Treating at least one symptom of cancer can include any of the following, or any combination thereof: inhibiting tumor growth, reducing tumor size, reducing tumor number, reducing tumor burden, preventing cancer recurrence, preventing metastasis of a primary tumor.
The terms “cancer” and “cancerous” refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth. Included in this definition are benign and malignant cancers. Examples of cancer include but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, leukemia and germ cell tumors. More particular examples of such cancers include adrenocortical carcinoma, bladder urothelial carcinoma, breast invasive carcinoma, cervical squamous cell carcinoma, endocervical adenocarcinoma, cholangiocarcinoma, colon adenocarcinoma, lymphoid neoplasm diffuse large B-cell lymphoma, esophageal carcinoma, glioblastoma multiforme, head and neck squamous cell carcinoma, kidney chromophobe, kidney renal clear cell carcinoma, kidney renal papillary cell carcinoma, acute myeloid leukemia, brain lower grade glioma, liver hepatocellular carcinoma, lung adenocarcinoma, lung squamous cell carcinoma, mesothelioma, ovarian serous cystadenocarcinoma, pancreatic adenocarcinoma, pheochromocytoma, paraganglioma, prostate adenocarcinoma, rectum adenocarcinoma, sarcoma, skin cutaneous melanoma, stomach adenocarcinoma, testicular germ cell tumors, thyroid carcinoma, thymoma, uterine carcinosarcoma, uveal melanoma. Other examples include breast cancer, lung cancer, lymphoma, melanoma, liver cancer, colorectal cancer, ovarian cancer, bladder cancer, renal cancer or gastric cancer. Further examples of cancer include neuroendocrine cancer, non-small cell lung cancer (NSCLC), small cell lung cancer, thyroid cancer, endometrial cancer, biliary cancer, esophageal cancer, anal cancer, salivary, cancer, vulvar cancer, cervical cancer, Acute lymphoblastic leukemia (ALL), Acute myeloid leukemia (AML), Adrenal gland tumors, Anal cancer, Bile duct cancer, Bladder cancer, Bone cancer, Bowel cancer, Brain tumors, Breast cancer, Cancer of unknown primary (CUP), Cancer spread to bone, Cancer spread to brain, Cancer spread to liver, Cancer spread to lung, Carcinoid, Cervical cancer, Children's cancers, Chronic lymphocytic leukemia (CLL), Chrome myeloid leukemia (CML), Colorectal cancer, Ear cancer, Endometrial cancer, Eye cancer, Follicular dendritic cell sarcoma, Gallbladder cancer, Gastric cancer, Gastro esophageal junction cancers, Germ cell tumors, Gestational trophoblastic disease (GIT)), Hairy cell leukemia, Head and neck cancer, Hodgkin lymphoma, Kaposi's sarcoma, Kidney cancer, Laryngeal cancer, Leukemia, Gastric linitis plastica, Liver cancer, Lung cancer, Lymphoma, Malignant schwannoma, Mediastinal germ cell tumors, Melanoma skin cancer, Men's cancer, Merkel cell skin cancer, Mesothelioma, Molar pregnancy, Mouth and oropharyngeal cancer, Myeloma, Nasal and paranasal sinus cancer, Nasopharyngeal cancer, Neuroblastoma, Neuroendocrine tumors, Non-Hodgkin lymphoma (NHL), Esophageal cancer, Ovarian cancer, Pancreatic cancer, Penile cancer, Persistent trophoblastic disease and choriocarcinoma, Pheochromocytoma, Prostate cancer, Pseudomyxoma peritonei, Rectal cancer. Retinoblastoma, Salivary gland cancer, Secondary' cancer, Signet cell cancer, Skin cancer, Small bowel cancer, Soft tissue sarcoma, Stomach cancer, T cell childhood non Hodgkin lymphoma (NHL), Testicular cancer, Thymus gland cancer, Thyroid cancer, Tongue cancer, Tonsil cancer, Tumors of the adrenal gland, Uterine cancer. Vaginal cancer, Vulval cancer, Wilms' tumor, Womb cancer and Gynaecological cancer. Examples of cancer also include, but are not limited to, Hematologic malignancies, Lymphoma, Cutaneous T cell lymphoma, Peripheral T cell lymphoma, Hodgkin's lymphoma, Non-Hodgkin's lymphoma, Multiple myeloma, Chrome lymphocytic leukemia, chronic myeloid leukemia, acute myeloid leukemia, Myelodysplastic syndromes, Myelofibrosis, Biliary tract cancer, Hepatocellular cancer, Colorectal cancer, Breast cancer, Lung cancer, Non-small cell lung cancer, Ovarian cancer, Thyroid Carcinoma, Renal Cell Carcinoma, Pancreatic cancer, Bladder cancer, skin cancer, malignant melanoma, merkel cell carcinoma, Uveal Melanoma or Glioblastoma multiforme.
The nucleotide sequences encoding the TAAs to be expressed by a Sindbis viral vector as described herein are well known in the art and can be easily obtained from the literature. For example, the sequence of NY-ESO-1, a testicular antigen aberrantly expressed in human cancers was published in 1997 (http://www.pnas.org/content/94/5/1914.full, Yao-Tseng Chen, Matthew J. Scanlant, Ugur Sahin, Ozlem Tiireci, Ali O. Guret, Solam Tsangt, Barbara Williamsont, Elisabeth Stockertt, Michael Pfreundschuh, and Lloyd J. Old, PNAS 1997.), whereas the Carcinoembryonic antigen sequence was published in 1987 (http://mcb.asm.org/content/7/9/3221.short Isolation and characterization of full-length functional cDNA clones for human carcinoembryonic antigen. N Beauchemin, S. Benchimol, D Cournoyer, A Fuks and C P Stanners, Molecular and Cellular Biology.
Although in mice a single i.p. injection of the SV/TAA as described herein, is sufficient to elicit a detectable CD8+ mediated immune response directed against the tumor, other regimens may be necessary for achieving a maximal response. For example, between 1 and about 8 i.p. injections over a time period of between 1 week and many weeks, with the possibility of injecting one or more booster injections 1 or more years later, may be preferably administered for a maximum effect.
This study, investigates the therapeutic efficacy of a replication-deficient oncolytic viral vector called Sindbis Virus. Because Sindbis Virus (SV) is a blood-borne pathogen, vectors from this virus can be administered in the bloodstream via the intravenous (i.v.) and intraperitoneal (i.p.) routes, which greatly facilitates their delivery [Tseng, J C, et al., Nature Biotech., 2004]. Furthermore, SV was genetically modified to be replication-defective by splitting its genome and deleting the packaging signal to block viral assembly after viral replication [Bredenbeek P J, et al., J. Virol. 1993]. This study shows that SV expressing the pro-inflammatory cytokine IL-12 (SV.IL12) activates T cells as well as enhances the expression of OX40 on CD4 T effector cells and, therefore, potentiates efficacy of the agonistic anti-OX40 antibody therapy. The data indicates that combination of SV.IL12 and anti-OX40 activates tumor immunity against low immunogenic tumors through the metabolic rewiring of T cells into highly activated effector cells. Furthermore, SV.IL12 in combination with anti-OX40 induces a marked immune cell infiltration into the tumor microenvironment. Considering that tumors tend to quickly escape the immune response by mutating or losing the expression of drug targets or tumor antigens targeted by the immune response, the treatment approach disclosed herein reduces the risk of developing tumor resistances and offers an attractive and safe strategy to change the immunogenic phenotype of various cancers without prior knowledge of tumor antigens.
The studies presented herein describe several, non-limiting examples of anti-OX-40 antibody, Sindbis viral vector (SV), Sindbis viral vector expressing IL-12 (SV.IL-12), Sindbis viral vector expressing an anti-OX-40 antibody and Sindbis viral vector expressing both IL-12 and an anti-OX-40 antibody. These examples are provided below to further illustrate different features of the present invention. The examples also illustrate useful methodology for practicing the invention. These examples do not and are not intended to limit the claimed invention.
Materials and Methods
Cell Lines
Baby hamster kidney (BHK), BALB/c colon carcinoma [CT26.WT (ATCC® CRL-2638™)] and FVB prostate carcinoma [MyC-CaP (ATCC® CRL-3255™)] cell lines were obtained from the American Type Culture Collection (ATCC). Firefly luciferase (Fluc)-expressing CT26 and MyC-CaP cells (CT26.Fluc and MyC-CaP.Fluc) were generated by stable transfection of pGL4.20_Fluc plasmid.
BHK cells were maintained in minimum essential a-modified media (a-MEM) (Corning CellGro) with 5% fetal bovine serum (FCS, Gibco) and 100 mg/ml penicillin-streptomycin (Corning CellGro). CT26.Fluc and MyC-CaP.Fluc cells were maintained in Dulbecco's modified Eagles medium containing 4.5 g/l Glucose (DMEM, Corning CellGro) supplemented with 10% FCS, 100 mg/ml penicillin-streptomycin, 7.5 μg/ml Puromycin or 400 g/ml Gentamycin, respectively. All cell lines were cultured at 37° C. and 5% C02.
SV Production
SV-LacZ production and titering were done the same as previously described [Scherwitzl I, Mal Ther Oncolytics. 2018]. SV.IL12 and SY.Lacz vectors were produced as previously described [Subramanian A et al., Proc Natl Acad Sci US A. 2005; Leonard W J et al., F1000Res. 2016; Rowell J F et al., J. Immunol. 1999; Metcalf T U et al., J. Virol. 2013]. All SV viral vectors used in these studies are replication-defective. Vectors were produced as previously described. SV.IL12 plasmid used in this study has been published in 2002 [Tseng J C et al., J Natl Cancer Inst. 2002]. To construct a Sindbis viral vector containing genes for interleukin 12 (IL-12), the Sindbis viral vector SinRep/2PSG was first constructed, which contains a secondary subgenomic promoter that is responsive to the Sindbis replicase. Two DNA oligonucleotide primers (sequence 5′ CGCGTAAAGCATCTCTACGGTGGTCCTAATAGTGCATG-3′; SEQ ID NO: 29) and its complementary strand 5′CACTATTAGGACCACCGTCGAGATGCTTTA-3′; SEQ ID NO: 30) containing the subgenomic promoter sequence were annealed and ligated into the MluI and SphI sites of the SinRep plasmid. The murine IL-12 α subunit gene (mp35; ATCC 87596) and the IL-12 β subunit gene (mp40; ATCC 87595) were subcloned into the MluI and the StuI sites of SinRep/2PSG, respectively, to produce the Sin-Rep/IL12 plasmid.
SV empty is the same plasmid without an additional gene of interest (e.g.IL12). SV.Luc was generated as described [Tseng, J C et al., Nature Biotech., 2004]. SV.GFP was generated as published in 2012 [Suzme R et al., Cancer Gene Ther., 2012]. Briefly, plasmids carrying the replicon (e.g. SinRep-IL12 or SinRep-IL-12) or DHBB helper RNAs were linearized with XhoI. In vitro transcription was performed using the mMessage mMachine RNA transcription kit (Ambion). Helper and replicon RNAs were then electroporated into BHK cells and incubated at 37° C. in αMEM supplemented with 10% FCS. After 12 hours, the media was replaced with OPTI-MEM (GIBCO-BRL) supplemented with CaCl2) (100 mg/l) and cells were incubated at 37° C. After 24 hours, the supernatant was collected, centrifuged to remove cellular debris, and frozen at −80° C. Vectors were titrated as previously described [Tseng J C et al., J. Natl. Can. Inst., 2002].
In Vivo Experiments and Tumor Models
All experiments were performed in accordance with the Institutional Animal Care and Use Committee of New York University Health. Six to 12-week old female BALB/c mice were purchased from Taconic (Germantown, N.Y.) and age matched male FVB/NJ mice were purchased from Jackson Laboratory.
Tumor Inoculation and Animal Studies
Treatment started on day 4 after i.p. inoculation of 7×104 CT26.Fluc cells or 105 cells of MyC-CaP.Fluc in 500 μl OPTI-MEM. For treatments, mice were randomized and SV (107 TU/ml), in a total volume of 500 μl, was injected i.p. into the left side of the animal once for CT26.Fluc and 4 days a week (days 1, 2, 3, 4) for a total of 4 weeks for MyC-CaP.Fluc inoculated mice. The immune checkpoint inhibitor anti-OX40 (clone OX-86, BioXCell) was injected i.p. into the left side of the animal at a dose of 250 μg per injection (1×/week for the CT26.Fluc and 3×/week for MyC-CaP.Fluc tumor bearing mice). Therapeutic efficacy of the treatment was monitored in two ways: tumor luminescence and survival. Noninvasive bioluminescent imaging was performed using the IVIS Spectrum imaging system (Caliper Life Science) at the indicated time points and tumor growth was quantified using the Living Image 3.0 software (Caliper Life Science) as previously described 86. Relative tumor growth for each mouse was calculated dividing total body counts of a given day by total body counts of the first IVIS image. Survival was monitored and recorded daily.
Flow Cytometry
For flow cytometry analysis, spleens and tumors were harvested from mice and processed as previously described [Scherwitzl I, et al., Mol. Ther. Oncol, 2018]. The extracted tumors were chopped into small pieces and incubated with a digestive mix containing RPMI with collagenase IV (50 μg/ml) and DNAse I (20 U/ml) for 1 hour at 37° C. Tumor samples had additional hyaluronidase V (50 μg/ml) in the digestive mix.
Spleens and digested tumors were mashed through a 70-μm strainer before red blood cells were lysed using ammonium-chloride-potassium (ACK) lysis (Gibco). Cells were washed with PBS containing 1% FCS and surface receptors were stained using various antibodies. Fluorochrome-conjugated antibodies against mouse CD3, CD4, CD44, ICOS, OX40, CD69, Foxp3, Granzyme B and Tbet, were purchased from Biolegend. Fluorochrome-conjugated antibodies against mouse CD8a were purchased from BD Biosciences. Mitotracker Deep Red FM, Mitotracker Green and Fluorchrome-conjugated antibodies against CXCR3 and Ki67 were purchased from Thermofisher. Stained cells were fixed with PBS containing 4% Formaldehyde. For intracellular staining, the forkhead box P3 (FOXP3) staining buffer set was used (eBioscience). Flow cytometry analysis was performed on a LSR II machine (BD Bioscience) and data were analyzed using FlowJo (Tree Star).
T Cell Isolation
Total T cells were freshly isolated with the EasySep™ mouse T Cell Isolation Kit. Freshly isolated lymphocytes were depleted of either CD4 or CD8 specific T cells using EasySep™ mouse CD4 and CD8 Positive Selection Kits II. Isolation of T cells and depletions were performed according to the manufacturer's protocols (Stemcell Technologies).
Enzyme-Linked Immunospot (ELISPOT)
Enzyme-linked immunospot was performed as previously described [Scherwitzl I, et al., Mol. Ther. Oncol, 2018]. Splenocytes and T cells were prepared as described for flow cytometry. Mouse IFNγ ELISPOT was performed according to the manufacturer's protocol (BD Bioscience). Lymphocytes (4×105 cells) and isolated (8×104) T cells were plated per well overnight in RPMI supplemented with 10% FCS. No additional stimulus was used in the ELISPOT. As positive control, cells were stimulated with 5 ng/ml PMA+1 g/ml Ionomycin.
Ex Vivo Cytotoxic Assay
T cells were isolated on day 7 and day 14 during treatment. 8×105/ml T cells were co-cultured with CT26.Fluc cells (2×104/ml) or MyC-CaP.Fluc cells (2×104/ml) in a 24 well plate for 2 days in 1 ml RPMI supplemented with 10% FCS. Cells were washed with PBS and lysed with 100 l of M-PER Mammalian Protein Extraction Reagent (Promega) per well. Cytotoxicity was assessed based on the viability of CT26 cells, which was determined by measuring the luciferase activity in each well. Luciferase activity was measured by adding 100 l of Steady-Glo reagent (Promega) to each cell lysate and measuring the luminescence using a GLOMAX portable luminometer (Promega).
CD8+ and CD4+ T-Cell Depletion In Vivo
CD8+ T cells were depleted using anti-CD8 antibody (clone 2.43) (Bio X cell, Lebanon, N.H.). 0.1 mg antibody in 0.2 ml PBS was injected into each mouse, starting 1 day before the first SV treatment, and then every 4 days for 2 weeks. CD4+ T cells were depleted using anti-CD4 antibody (clone GK 1.5) (Bio X cell, Lebanon, N.H.). 0.4 mg were injected into each mouse, starting day 1 before the first treatment. Control mice were injected with PBS and isotype controls.
Quantitative Real-Time PCR
RNA was extracted from tumor samples using RNeasy Kit (Qiagen), followed by cDNA synthesis with the iScript II Kit (Bio-Rad). qRT-PCR was performed using iQ™ SYBR Green Supermix (Biorad) and an StepOne™ Real-Time PCR Detection System (Applied Biosystems). PCR conditions were as follows: 95° C. for 10 min, followed by 40 cycles (94° C. for 30 s, 58° C. for 30 s) of amplification. For quantitation, CT values were normalized to GAPDH and expression was analysed using the 2-ΔΔCT method. Primers for CXCL9, CXCL10 and GAPDH were used. CXCL9 (Forward: GAAGTCCGCTGTTCTTTTCC; SEQ ID NO: 25 Reverse: TTGACTTCCGTTCTTCAGTG; SEQ ID NO: 26), CXCL10 (Forward: GCTGCAACTGCATCCATATC; SEQ ID NO: 27; Reverse: AGGAGCCCTTTTAGACCTTT; SEQ ID NO: 28).
Transcriptome Analysis of T Cells
Total RNA was extracted from freshly isolated T cells on day 7 of treatment from spleens using RNeasy Kit (Qiagen). For each group, 3 BALB/C mice or 3 FVB/J mice were used for biological repeats. RNA-seq was done by NYUMC Genome Center. RNA quality and quantity was analyzed. RNAseq libraries were prepared and loaded on the automated HiSeq 4000 Sequencing System (Illumina) and run as single 50 nucleotide reads.
Alignment and Differential Expression Analysis
Sequences were aligned to the mm10 mouse genome using Bowtie software, Version 1.0.087 [Langmead R et al., Genome Biol. 2009] with two mismatches allowed. Uniquely mapped reads were further processed by removing PCR duplicates with Picard (“Picard Tools.” Broad Institute, GitHub repository. http://broadinstitute.github.io/picard/) MarkDuplicates and transcripts were counted using HTSeq88 and differential gene expression was performed between all groups using DESeq [Anders S et al., Genome Biol. 2010]. Differences in gene expression were considered significant if padj<0.05.
GSEA and Enrichment Map Analysis
The network-based method enrichment map 90 was used for gene-set enrichment visualization and interpretation of data. As a follow up analysis of Gene-Set Enrichment Analysis2 (GSEA) [Mootha V K et al., Nat. Genet., 2003] it reduces redundancy and helps in the interpretation of large gene sets and helps to quickly identify major enriched functional themes in the gene expression data. To perform this analysis, we first assigned a unique row identifier for each transcript and obtained differentially expressed genes through DESeq [Anders S et al., Genome Biol. 2010]. These genes were then ranked and GSEA was performed in Gene Pattern 92 server using GSEA pre-ranked module. We then obtained the gene identifiers corresponding to the gene names using the Bioconductor package ‘org.Mm.eg.db’ and the resulting positively and negatively regulated gene identifiers were used to generate enrichment maps in Cytoscape [Shannon P et al., Genome Res. 2003]. Expression heatmap is drawn by Morpheus (https://software.broadinstitute.org/morpheus/). Highest and lowest expression for each gene (row min. and row max.) were displayed as red or blue color, respectively.
Measurement of Oxygen Consumption and Extracellular Acidification Rates of T Cells
T cell metabolic output was measured by Seahorse technology as previously described [Scharping N E et al., Cancer Immunol. Res., 2017]. Purified T cells were plated at 6×105 cells/well in a Seahorse XF24 cell culture microplate. Oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) were measured using an Agilent Seahorse XFe24 metabolic analyzer following the procedure recommended by the manufacturer (Agilent). For the mitochondrial stress test, 1) oligomycin (1 μM), 2) FCCP (1.5 μM) and 3) rotenone (100 nM) and antimycin A (1 μM) were injected sequentially through ports A, B and C.
Immunoblot Analysis
Cells were lysed in M-PER© Mammalian Protein Extraction Reagent according to the manufacturer's protocol. Lysates were separated by SDS-PAGE on 4-12% Bio-Rad gels, transferred to polyvinylidene difluoride (PVDF) membranes, blocked in 5% Milk in TBS buffer with 0.1% Tween-20 (TBST). Primary antibodies to c-Myc (Santa Cruz Biotechnology) and GAPDH (Thermofisher) were added at room temperature or overnight at 4° C. Secondary fluorescent antibodies (IRDye®, LI-COR) were added in 5% Milk in TBST for 1 h at room temperature. Odyssey® Classic Infrared Imaging System was used for visualization.
Histochemistry and Multiplex immunofluorescence (MIF)
Tumors of mice were collected, fixed in 4% PFA for 2 days and embedded in paraffin, sectioned and H&E stained. For Multiplex immunofluorescence staining and imaging, five micron paraffin sections were stained with Akoya Biosciences® Opal™ multiplex automation kit on a Leica BondRX® autostainer, according to the manufacturers' instructions. Prior to incubation with the first primary antibody, sections underwent heat retrieval with Bond Epitope Retrieval Buffer 2 (Leica ER2, AR9640) and blocking. Primary antibodies in Panel 1 were against CD3 (1:200, Biorad, MCA1477T), CD8 (1:2000, Cell Signaling, 98941S), Ki67 (1:200, Abcam, AB16667). Primary antibodies in Panel 2 recognized iNos (1:1000, Genetex, GTX130246), Arg1 (1:750, Genetex, GTX109242), Granzyme B (1:1000, Abcam, AB4059), CD11b (1:10,000, Abcam, AB133357), F480 (1:250, Cell Signaling, 70076S). Each primary antibody was followed by a cocktail of horse radish peroxidase-conjugated secondary antibodies against mouse and rabbit IgG (RTU, Akoya/Perkin Elmer, Cat # ARH1001) and then tyramide mediated signal amplification (TSA) with covalent linkage of the individual Opal fluorophor (each 1:250, Opal 520 (FP1496001KT), 540 (FP1487001KT), 570 (FP1494001KT), 620 (FP1488001KT), 650 (FP1495001KT) or 690 (FP1497001KT), Akoya/Perkin Elmer Cat #'s) to the tissue antigen. Antibodies were subsequently stripped using either ER1 (Leica, AR9961) or ER2 (Leica, AR9640) heat retrieval buffer and the next round of immunostaining initiated. After completion of the sequential incubations and stripping, slides were counterstained with spectral DAPI (Akoya/PerkinElmer, FP1490). Monoplex controls were used to confirm appropriate staining for antibodies integrated into the multiplex panels. Multispectral imaging was performed on a Vectra3 imaging system (Akoya/PerkinElmer) at 20×. The fluorophore emission signatures were captured by a multispectral camera and then unmixed with InForm software (Akoya/PerkinElmer). Autofluorescence, obtained from an unstained slide, was removed from the composites and pseudo-colored images exported as tif files.
Statistical Analysis
Statistical analysis was performed using GraphPad Prism 7.0 as described in Figure legends. All data are shown as mean±s.e.m. Figures were prepared using GraphPad Prism 7, Adobe Photoshop and ImageJ Software. Treated groups were compared, using a one-way analysis using Prism7 (GraphPad Software), to naïve mice. Differences with a P value of <0.05 were considered significant: *P<0.05; **P<0.005; ***P<0.0001.
SV Expressing IL-12 Enhances the Expression of OX40 on CD4 T Cells
The study described herein investigated the therapeutic effect of SV.IL12 in immune-competent tumor bearing mice (colon cancer; CT26). To exploit SV.IL12 for cancer therapy, tumor cells were i.p. implanted and after tumor establishment (4 days after tumor cell injection [day 0]), SV, SV.IL12 or IL-12 were i.p. injected on 4 consecutive days (day 1, 2, 3 and 4) for a total of 4 weeks (
Intraperitoneal Delivery of SV.IL12 and Anti-OX40 Antibody Cures Established Cancers
To investigate whether the oncolytic activity of SV.IL12 in combination with anti-OX40 is required for successful anti-cancer therapy, SV non-susceptible (colon cancer; CT26) and susceptible (prostate cancer; MyC-CaP) tumor cell lines were used in this study (
Combination Therapy Markedly Changes the Transcriptome Signature of T Cells
The requirement of T cells during SV.IL12 with anti-OX40 treatment was assessed. The presence of both CD4 and CD8 T cells was required for eliciting the observed therapeutic efficacy as mice treated with the corresponding depleting antibodies were unable to control tumor growth (
Unbiased pathway enrichment and network analyses of DEG from control versus combination therapy was performed to determine biological processes in T cells that are influenced by this treatment (
Combination Therapy Enhances Systemic T Cell Responses, Favoring Th1 Like ICOS CD4 T Cells
As T cells from combination therapy express a marked change in their transcriptome signature compared with all other groups, markers for T cell differentiation and activation (e.g., PD-1, ICOS, OX40, TIM3, KLRG1, IL7R) as well as T cell lineage transcription factors (e.g., EOMES, TBET, GATA3, BCL6, RORC, FOXP3) were analyzed (
CD4 and CD8 T Cells are Metabolically Reprogrammed in Mice Treated with SV.IL12 and Anti-OX40
The tumor microenvironment can be a very challenging milieu for an effector T cell as it is characterized by hypoxia, acidosis and low levels of nutrient sources such as glucose and glutamine [Delgoffe, G M et al, Cancer Immunol. Res., 2016; Scharping, N. E, Vaccines, 2016; Chang, C H et al., Cell, 2015]. Even if T cell activation and initiation of effector function is allowed, T cells may be unable to generate the bioenergetic intermediates necessary to carry out effector function in the tumor microenvironment. Thus, providing a metabolic support for T cells is crucial for the success of cancer treatments as previously reported [Scharping, N. E. et al., Immunity, 2016; Ho, P C et al., Cell, 2015; Siska P J., et al, Trends immunol., 2015; Zhao et al., Nat. Immunol., 2016]. To test if SV.IL12 in combination with anti-OX40 influences the metabolic state of T cells, Gene Set Enrichment Analysis (GSEA) of the RNA sequencing data was performed between T cells from combined therapy and control. GSEA analysis showed significantly higher expression of genes involved in oxidative phosphorylation and glycolysis pathways during combination therapy (
Analysis of mitochondrial mass (
To determine the kinetics of peripheral T cell metabolism over the course of treatment with SV.IL12 and anti-OX40, OCR and ECAR were measured on day 7, 14 and 40 in CT26.Fluc bearing mice (
Metabolic Reprogrammed T Cells in SV.IL12 with Anti-OX40 Treated Mice Display Enhanced CD4 Mediated Cytokine Production and Anti-Tumor Activity
To test if metabolic reprogrammed T cells in combined therapy possess enhanced effector functions, cytokine production and cytotoxicity were analyzed in T cells isolated from spleens on day 7. Genes encoding pro-inflammatory cytokines ifng and il2 were upregulated in T cells from mice treated with SV in combination with anti-OX40 (
In addition, RNA levels of the cytotoxic proteases, granzyme A and B, were upregulated in mice treated with combination therapy compared with all other groups (
Having observed upregulation of granzymes and cytotoxic receptors in combination therapy, the function of T cells was investigated using an ex vivo tumor growth assay. Splenocytes obtained from all groups were co-cultured at an effector-to-target cell ratio of 10:1 with either CT26.FLUC (
Mice Treated with SV.IL12 in Combination with Anti-OX40 Display Enhanced T Cell Migration and Intratumoral T Cell Immunity
Only a minority of the total of treated patients respond to current immunotherapy and the presence of TILs has been shown to be one of the main factors that influence the responsiveness towards various therapies in multiple cancers [Galon, J et al., Science 2006; Hwang W T et al., Gynecol Oncol 2012]. Due to the fact that SV elicited anti-tumor responses do not necessarily require direct infection of the tumor or intratumoral injection, whether SV.IL12 therapy in combination with anti-OX40 could nevertheless alter the local tumor microenvironment and favor intratumoral immunity, was investigated. To assess whether SV.IL12 in combination with anti-OX40 induces T cell infiltration into the tumor, the chemokine receptor CXCR3 on peripheral T cells was analyzed after one week of treatment. In the CT26.Fluc model CXCR3 levels were significantly upregulated on CD4 T cells during combination therapy compared with all other groups and CXCR3 levels remained elevated over the course of treatment (
The study described herein provides a practical strategy for cancer immunotherapy using an OV and anti-OX40. This strategy takes advantage of the preexisting T cell immune repertoire in vivo, removing the need to know about present tumor antigens. The study described herein shows that the combination of replication-deficient SV.IL12 and anti-OX40 amplifies these antitumor T cells and induces their action throughout the body against two distinct solid tumors, reversing effectively local tumor-mediated immune suppression. This effect was specific for combination therapy and was not observed during SV.IL12 or anti-OX40 treatment alone.
The high metabolic activity of cancer cells together with the poor vasculature blood supply in the tumor microenvironment can induce nutrient deprivation [Delgoffe, G M et al., Cancer Immunol Res. 2016; Scharping, N E & Delgoffe, GM, Vaccines, 2016; Chang, C H et al., Cell, 2015]. These conditions can impair TCR signaling, glycolytic and mitochondrial metabolism, as well as decrease mitochondrial mass, all hallmarks of T effector cells, resulting in impaired anti-tumor effector functions of tumor-specific T cells. 39-42 Scharping, N. E. et al., Immunity 2016; Ho, P. C. et al., Cell 2015; Siska, P J & Rathmell, J C, Trends Immunol., 2015; Zhao, E. et al., Nat Immunol, 2016]. The data in two distinct models of cancer immunotherapy disclosed in the study described herein, shows that SV.IL12 in combination with OX40 signaling provides the necessary metabolic support to T cells to generate an efficient antitumor response. This metabolic support is characterized most prominently by elevated mitochondrial function and mass in CD8 T cells as well as a switch to aerobic glycolysis in CD4 T cells. T cells from mice treated with SV.IL12 in combination with anti-OX40 demonstrated enhanced protein expression of c-Myc compared with all other groups. Thus, the study described herein clearly shows that T cells are metabolically reprogrammed in the periphery during combination therapy.
The study described herein strongly shows that the therapeutic efficacy of SV.IL12 with anti-OX40 is driven by T cell modulation and reprogramming of its metabolic state, in order to enhance the anti-tumor response in the periphery and in the tumor microenvironment. Furthermore, the use of SV allows these metabolically reprogrammed T cells to better infiltrate the tumor microenvironment, which is crucial for an adequate immunotherapy. Anti-OX40 antibody is currently being studied in phase 1 and 2 clinical trials. SV will be tested as a single agent in its first clinical trial in the third quarter of 2020. The results from our current preclinical studies provide a strong rationale for combining SV.IL12 with agonistic anti-OX40 antibodies in a therapeutic format in patients with solid tumors. In summary, the studies described herein clearly show that even in absence of direct SV tumor targeting, SV.IL12 in combination with anti-OX40, or SV vector encoding IL-12 and anti-OX40, can alter the tumor microenvironment in distinct solid tumors through an indirect and immunity driven mechanism that enhances T cell infiltration and intratumoral T cell immunity.
The study described herein investigates the effect of administering IL-12 and anti-OX40 antibody, both expressed by Sindbis viral vectors, on established tumors. This strategy is particularly advantageous for treatment of cancers like ovarian cancer, wherein the combination of SV.IL-12 and anti-OX40 antibody is not found to be as effective, as observed in colon and prostate cancers. The administration of SV/IL-12 and an anti-OX40 antibody enhanced clearance of established tumor of colon and prostate cancer cell lines, CT26 and MyC-Cap respectively. C57/B16 albino (female) mice re-injected with Alm5-2Fluc-17 ovarian cancer cells to establish a tumor (
The study described herein, provides plasmid constructs for expressing IL-12, and anti-OX40 in a SV vector. The study described herein, provides plasmid constructs encoding IL-12 a and b subunits (
SV.IL12 plasmid used in this study has been published in 2002 [Tseng J C et al., J Natl Cancer Inst. 2002]. To construct a Sindbis viral vector containing genes for interleukin 12 (IL-12), the Sindbis viral vector SinRep/2PSG was first constructed, which contains a secondary subgenomic promoter that is responsive to the Sindbis replicase. Two DNA oligonucleotide primers (sequence 5′ CGCGTAAAGCATCTCTACGGTGGTCCTAATAGTGCATG-3′; SEQ ID NO: 29) and its complementary strand 5′CACTATTAGGACCACCGTCGAGATGCTTTA-3′; SEQ ID NO: 30) containing the subgenomic promoter sequence were annealed and ligated into the MluI and SphI sites of the SinRep plasmid. The murine IL-12 α subunit gene (mp35; ATCC 87596) and the IL-12 β subunit gene (mp40; ATCC 87595) were subcloned into the MluI and the StuI sites of SinRep/2PSG, respectively, to produce the Sin-Rep/IL12 plasmid.
The H and L chains of the OX40 Ab are expressed from a single SV using two subgenomic promoters. The synthesized sequences were designed to encode an IL-12 secretory signal peptide upstream of both H and L polypeptide sequences preceded by a ribosome binding site and the start codon. The variable Ab binding sequences that functionally bind to activate the OX40 Receptor contain complementarity determining regions that are not unique. The variable chain is linked to the respective L (GenBank accession BAR42292) and H chain (GenBank accession CAC20702) constant region sequences of mouse IgG2a; the murine IgG2a isotype is comparable to the BioXcell OX40 Ab used in parallel in vivo experiments.
In summary results described herein clearly show the possibility of using a combination of SV vectors expressing IL-12 and anti-OX40 antibody or a SV vector expressing both expressing IL-12 and anti-OX40 antibody, for treatment of cancers that may be resistant to treatment with anti-OX40 antibody administered directly.
The studies described herein use an antibody directed at 4-1BB (CD137, TNFRSF9), a T cell costimulatory molecule. 4-1BB agonist stimulation greatly enhances NK and cytotoxic T cell activity. There are preclinical studies showing that α4-1BB effectively treats lymphoma and that depletion of Treg cells enhances the therapeutic effect of α4-1BB [Houot R et al., Blood, 2009]. The A20 tumor cells uses in the study described herein were derived from a spontaneously arising reticulum cell sarcoma (a non-Hodgkin lymphoma) in a BALB/c mouse.
Previously, SV carrying NYESO-1 was used, which encodes the cancer testis TAA, NYESO-1, to cure CT26 tumors expressing NYESO-1 [Scherwitzl I et al., Mol. Ther. Oncolytics, 2018]. The studies described herein show that systemically disseminated A20 lymphoma can be completely cured by SV plus α4-1BB mAb combination therapy without the need to produce a SV that encodes a TAA known to be present in the A20 lymphoma cells. Further, neither intratumoral injection of the SV vectors nor infection of the tumors is required as the A20 B lymphoma cells used in the current model are resistant to SV infection.
One difference in the current study, compared with those previously published, is the use of SV vector combination therapy that involves an agonistic mAb for a costimulatory receptor versus targeting checkpoint blockade molecules such as CTLA4 and PD-1. The studies described herein show that agonistic mAbs in combination with SV vectors trigger a cascade of events that results in curative results. The findings disclosed herein reveal the potential of SV combination therapy to cure tumors for which TAAs are completely unknown.
Materials and Methods
Firefly Luciferase (Fluc)-Expressing A20 Cells Generation
A20 cells were transfected with pGL4-neo_Fluc plasmid (Promega) by electroporation via Nucleofector™ kit V (Lonza). Fluc-A20 cell clones were selected and maintained in RPMI1640 (Cellgro)+10% FBS (Gibco)+250 μg/ml G418 (Gibco). One A20 clone stably expressed fLuc and was used for tumor inoculation and consecutive experiments.
SV Production
SV-LacZ production and titering were done the same as previously described [Scherwitzl I et al., Mol. Ther. Oncolytics, 2018].
SV-GFP Infection
A20 cells and control BHK cells were infected by SV carrying GFP for 1 h. The GFP expression was observed the next day by fluorescence microscopy.
A20 Tumor Inoculation and In Vivo Imaging System (IVIS) Imaging
3×106 fLuc-A20 cells were inoculated to BALB/C mice by i.p injection. Tumor growth was monitored as previously described [Scherwitzl I et al., Mol. Ther. Oncolytics, 2018].
SV and α4-1BB Ab treatment
Treatment was started after successful tumor inoculation (4 days after tumor cell injection, confirmed by IVIS imaging). Tumor growth was measured every week by noninvasive bioluminescent imaging. SVLacZ was injected 4 times per week, for totally 3 weeks. The virus (107-108 TU/mL) in a total volume of 500 μL was i.p. injected. For 2 groups (4-1BB and SV plus 4-1BB), 350 μg/mouse 41BB Ab was injected 3 times/week for 2 weeks. InVivo MAb anti-mouse 4-1BB was ordered from BioXCell (Clone: LOB12.3, Cat. No. BE0169). In low dose treatment protocol, SVLacZ was injected i.p. 3 times per week, for totally 3 weeks. 41BB Ab (50 g/mouse) was injected once a week for 3 weeks.
Elispot
Mouse IFNγ ELISPOT was performed according to the manufacturer's protocol (BD Biosciences). 2×105 splenocytes or 1×105 T cells were plated per well O/N in RPMI supplemented with 10% FBS. For a positive control, splenocytes were stimulated with 5 ng/ml PMA+1 μg/ml Ionomycin.
Flow Cytometry
Fluorochrome-conjugated antibodies against mouse CD3, CD4, CD8, CD25, CD44, CD62L, ICOS, CD11 a, ICAM-1 were purchased from Biolegend (San Diego, Calif.). Fluorochrome-conjugated antibodies against mouse Foxp3, EOMES and CCR5 were purchased from Thermofisher. BUV395 conjugated antibody against mouse CD8a was purchased from BD Biosciences. For surface staining, cells were washed and stained with anti-mouse direct conjugated antibodies. Cells were analyzed using the LSRII flow cytometer (BD Biosciences) and data were analyzed using Flowjo software (Treestar, Ashland, Oreg.). For intracellular cytokines staining, stimulated cells were fixed with cytofix/cytoperm solution (BD Biosciences), permeablized with perm/wash buffer (BD Biosciences) and stained with anti-mouse IFNγ antibodies. For nuclear antigen, cells were fixed and permeabilized by Foxp3 fixation/permeabilization buffer (eBioscience) and stained with anti-Foxp3, T-bet, Ki67 and EOMES antibody.
RNA Isolation and Transcriptome Analysis
Total RNA was harvested by RNAeasy isolation kit (Qiagen, Valencia, Calif.). For each group, 3 BALB/C mice were used as biological repeats. RNA-seq was performed by NYUMC Genome Technology Center (GTC). To identify significant differences in expression between any pair of groups, differential expression analysis was performed using Deseq2 and an adjusted p value cutoff of 0.05 was applied [19, Love M I et al, Genome Biol. 2014] (q<0.05). To increase stringency, only genes with a Log 2 fold change ≥1 (upregulated) or ≤−1 (downregulated) were selected for further analysis. Gene cluster analysis was performed by DAVID analysis using the selected differentially expressed genes [Huang da W et al., Nucleic Acids Res 2009, Huang da W et al., Nat Protoc. 2009]. RNA-seq results (normalized counts) were used as input to perform with Gene Set Enrichment Analysis (GSEA) [Subramanian A, et al., Proc Natl Acad Sci USA. 2005]. Molecular Signatures Database (MSigDB)v4.0 were used as screening database. For each gene, the gene expression value is normalized by the relative log 2 fold change compared to the median value of this gene. Expression heatmap is drawn by Morpheus (https://software. broadinstitute.org/morpheus/). Cannonical pathway and disease and biological functional analysis were generated by ingenuity pathway analysis (IPA; Ingenuity Systems, Redwood City, Calif.) using the statistical differential expressed genes list. To increase the sample representativeness, for IPA, we choose nominal p<0.05 as cutoff value.
Tumor Infiltrating Lymphocyte (TIL) Harvest
To investigate the phenotype of TIL, all treatments were started 11 days after tumor inoculation, After 7 days treatment, tumor mass was harvested and the phenotype of TIL were analyzed as previously described [18, Scherwitzl I, Mol Ther Oncolytics. 2018].
T Cell Seahorse Assay
T cells were isolated from spleen by using pan T cell isolation kit (Stemcells). T cells were plated at 6×105 cells/well in 24 well plate. Oxygen consumption rate (OCR) and excellular acidification rate (ECAR) were measured by Agilent Seahorse XFe24.
Statistical Analysis
For the two group comparison, statistical difference was determined by unpaired two tail Student t-test. The multiple sample comparison was analyzed by one way ANOVA. P<0.05 was determined to be significant for all experiments. All values were calculated with Excel (Microsoft) and Prism software (GraphPad).
SV and α4-1BB mAb Combination Completely Cured A20 Lymphoma
To explore if SV has therapeutic effect on tumors not targeted or infected by SV vectors, the A20 B cell lymphoma was used, which is highly resistant to SV infection (
SV Alone and SV Plus α4-1BB mAb Stimulated Cell Cycle Progression, Cytokine Production, and Activation
In the study described herein, SV significantly inhibited tumor growth by day 7 (
NIH DAVID cluster analysis was performed using the upregulated gene list. Enriched clusters were ranked based on enrichment score. Cell cycle gene cluster achieved the highest enrichment score (
To understand why SV plus α4-1BB mAb achieves the best therapeutic effect, Deseq2 analysis was run for SV plus α4-1BB mAb vs. untreated samples. 1046 upregulated genes (q<0.05 and Log 2 Fold Change≥1) and 877 downregulated genes (q<0.05 and Log 2 Fold Change≤−1) in the SV plus α4-1BB mAb group were identified (
Next, NIH DAVID analysis using the upregulated gene list was run. In both comparisons, cell cycle genes upregulation is the highest enrichment cluster [although SV+α4-1BB mAb vs. SV has a lower enrichment score compared with SV plus α4-1BB mAb vs. untreated samples (
CD69 is the earliest marker of immune system activation. SV plus α4-1BB mAb treatment synergistically upregulated CD69 on day 2 (
SV Plus α4-1BB mAb Stimulated Cytotoxic T Cell Function
To investigate the antitumor cytotoxicity of SV/α4-1BB treated splenocytes, f-Luc A20 lymphoma cells were co-cultured with splenocytes on day 7. The ratios explored between splenocytes and tumor cell were 40:1, 20:1, 10:1. SV plus α4-1BB treated splenocytes demonstrated the highest cytotoxicity among all groups, as calculated by the reduction of f-Luc activity (
SV Plus α4-1BB mAb Induced IFNγ Production from T Cells
Other upregulated genes in the SV plus α4-1BB mAb combined treatment include STAT4 (
Next, to identify whether CD4 or CD8 T cells produce IFNγ, flow cytometric analysis was performed for cytokine analysis. Among splenocytes, 2-2.5% SV plus α4-1BB mAb treated CD4 T cells produced IFNγ, which is significantly higher than other groups. Very low percentages of CD8 T cells produced IFNγ in all groups (
SV and α4-1BB mAb Stimulated Chemotaxis, Adhesion and Enhanced T Cell Infiltration and Activation in Tumor
Through RNA-Seq, a series of chemokines and chemokine receptors have been identified to be upregulated in SV plus α4-1BB mAb (
TIL play a critical anti-tumor role and is an important marker for prognosis. Compared with untreated, the percentage of CD3 and CD8 T cells were increased about 2 fold after combination treatment (
SV and α4-1BB mAb Synergistically Enhanced Oxidative Phosphorylation
T cell activation requires a quick consumption of energy through both enhanced glycolysis and oxidative phosphorylation [Wahl D R et al., Immunol Rev., 2012]. Metabolic switch is a major feature of T cell activation and memory T cell development [van der Windt G J et al., Immunol Rev., 2012]. GSEA KEGG analysis identified that the glycolysis gene set is upregulated in SV plus α4-1BB vs. untreated samples (
Both oxygen consumption rate (OCR, represents oxidative phosphorylation) and extracellular acidification rate (ECAR, represents glycolysis) of all groups (
SV Plus Low Dose α4-1BB mAb Cured A20 Tumor Bearing Mice
To reduce the potential risk of cytotoxicity and expense of treatment with SV vectors plus α4-1BB, the study disclosed herein explored whether low doses of α4-1BB mAb and fewer injections would be as effective in curing tumor bearing mice as the higher doses and frequencies used in our initial studies. As demonstrated (
All Tumor Cured Mice Acquired Long Lasting Antitumor Immunity
To investigate the memory response to A20 lymphoma, naive and tumor cured mice were inoculated with 3×106 A20 tumor cells. Only mice that had survived more than 4 months after 1st time of tumor challenge were chosen. In all tumor cured mice, we found that A20 lymphoma was completely rejected whereas naive mice were susceptible to A20 inoculation (
To confirm anti-tumor specificity has been elicited, IFNγ production of purified T cells in the presence or absence of tumor cells was measured by Elispot assay. T cells were isolated from naive and cured mice under SV plus α4-1BB treatment (4 months after treatment finished). Isolated T cells were co-cultured with A20 and CT26 tumor cells respectively. Co-culturing with A20 cells dramatically enhanced IFNγ production, whereas co-culturing with CT26 cells only slightly enhanced IFNγ production (
Next, cytotoxicity to both naive and cured mice under SV plus α4-1BB treatment (the same method as
To better understand differences between this memory T cell response and the initial treatment responses as observed on day 7, RNA-Seq was performed by using purified splenic T cells from all re-challenged groups. In T cells of these re-challenged mice we found only a few differentially expressed genes among the three treated groups (Table 5), indicating that tumor cured mice develop a very similar T cell gene expression profile regardless of treatment method. Compared with untreated, KEGG analysis indicates that TCR signaling is the highest upregulated pathway in SV plus α4-1BB group (
The conventional view of oncolytic virus therapy against tumors is that it requires selective infection of cancer cells resulting in the induction of cancer cell lysis and apoptosis. TAAs, released from dead tumor cells, attract and further stimulate an antitumor immune response. The study described herein found that encoding a TAA is not necessary for SV vectors plus α4-1BB mAb therapy to be fully successful. SV vectors lacking an A20 lymphoma TAA were able to treat A20 lymphoma and, in combination with α4-1BB mAb, eradicated the growing tumors. This is particularly important when effective immune reactive TAAs are unknown. It is possible that the immunotherapeutic response of SV vectors plus α4-1BB mAb is independent of whether a tumor is “cold” (i.e., having few TAAs or mutation-specific neoantigens capable of promoting robust T cell activation) or “hot.”
The study describe herein showed that both NKG2D (KLRK1) and granzyme B are highly expressed under combination treatment. This massive nonspecific activation is critical for controlling tumor growth at an early time point (day 7). This step is also important for inducing anti-tumor specificity that is mediated by TAAs released from dead tumor cells due to nonspecific killing. After tumor regression, T cells from treated animals were able maintain the ability to produce IFNγ and acquired immunological memory to rapidly reject A20 lymphoma rechallenges. IFNγ production from purified T cells of cured mice was significantly enhanced after encountering A20 tumor cells. This demonstrates that anti-tumor specificity is fully established in cured mice. Upregulated molecular pathways of responsive T cells induced by SV vectors and a 4-1 in mAbs alone and in combination were identified and compared in the study described herein. The combination of SV and α4-1BB mAb has a synergistic effect and represents a potent and robust therapeutic treatment able to cure B lymphomas and provide long term protection in a preclinical model.
In conclusion, SV vectors in combination with α4-1BB mAb completely eradicated a B-cell lymphoma in a preclinical mouse model, a result that could not be achieved with either treatment alone. Tumor elimination involves a synergistic effect of the combination that significantly boosts T cell cytotoxicity, IFN-γ production, migration, tumor infiltration and oxidative phosphorylation. In addition, all mice that survived after treatment developed long lasting antitumor immunity. The studies disclosed herein provides a novel, alternative method for B cell lymphoma treatment and describes a rationale to help translate SV vectors plus agonistic mAbs into clinical applications.
The study described herein investigates the effect of administering a tumor associated antigen and an immunostimulatory molecule, as expressed by a Sindbis viral vector on anti-tumor response and survival in a subject with an established tumor. Previous studies, demonstrated vectors encoding TAAs, such as NY-ESO-I, could cure CT26-NY-ESO-1 tumors [Galon J, et al., Nature reviews Drug discovery 2019; Gupta S, et al., Frontiers in oncology 2017]. However, while this approach has been effective in enhancing the immune response to and clearance of established tumors of colon and prostate cancers, the efficacy in curing other cancers, e.g. ovarian cancer has been limited. Therefore, an approach of administering a combination of a SV expressed immunostimulatory molecule, IL-12 along with the SV-NY-ESO-1, to a subject with an established tumor was tested.
Combination of NY-ESO-1 and IL-12 Expressed by Separate Sindbis Viral Vectors Enhances Survival of Subjects with Established Tumors
The study described herein investigates the effect of administering IL-12 and NY-ESO-1, both expressed by separate Sindbis viral vectors, on established tumors. C57/B16 albino (female) mice re-injected with Alm5-2Fluc-17 ovarian cancer cells to establish a tumor (
A Sindbis replicon expressing NYESO-1 cDNA (SV-NYESO1) was made by PCR amplification of the NYESO-1 gene from the pReceiver-M02 plasmid. Expression of the NYESO-1 gene was confirmed by western blot. NYESO-1 was detected by western blot following standard protocol, using as a primary antibody the anti-NYESO-1 clone E978 (Upstate) at a dilution 1/5,000 in Tris-buffered saline-Tween (TBS-T) with 5% non-fat milk. SV.IL12 plasmid used in this study has been published in 2002 [Tseng J C et al., J Natl Cancer Inst. 2002]. To construct a Sindbis viral vector containing genes for interleukin 12 (IL-12), the Sindbis viral vector SinRep/2PSG was first constructed, which contains a secondary subgenomic promoter that is responsive to the Sindbis replicase. Two DNA oligonucleotide primers (sequence 5′ CGCGTAAAGCATCTCTACGGTGGTCCTAATAGTGCATG-3′; SEQ ID NO: 29) and its complementary strand 5′CACTATTAGGACCACCGTCGAGATGCTTTA-3′; SEQ ID NO: 30) containing the subgenomic promoter sequence were annealed and ligated into the MluI and SphI sites of the SinRep plasmid. The murine IL-12 α subunit gene (mp35; ATCC 87596) and the IL-12 β subunit gene (mp40; ATCC 87595) were subcloned into the MluI and the StuI sites of SinRep/2PSG, respectively, to produce the Sin-Rep/IL12 plasmid.
As expected the SV-IL-12 treatment group showed a better percentage survival of mice with tumor over the SVNYESO treatment group and the untreated (control) group. However, a synergistically higher showed enhanced percentage survival rate was observed in the SV-NYESO_SV-IL12 in comparison to the SV-IL-12 treatment group (
Combination of NY-ESO-1 and IL-12 Expressed by the Same Sindbis Viral Vectors Enhances Survival of Subjects with Established Tumors
The study described herein investigates the effect of administering IL-12 and NY-ESO-1, both expressed by the same Sindbis viral vector, on established tumors. C57/B16 albino (female) mice re-injected with Alm5-2Fluc-17 ovarian cancer cells to establish a tumor (
The study described herein, provides plasmid constructs for expressing NY-ESO-1, IL-12 and anti-OX40 in a SV vector. The study described herein, provides plasmid constructs encoding IL-12 α and b subunits (
In summary, the results of the study described herein clearly show the possibility of using a SV vectors expressing both IL-12 and NY-ESO-1, for treatment of cancers that may be resistant to treatment with a SV expressing a tumor associated antigen.
This application claims the benefit of U.S. Provisional Application No. 62/871,675, filed on Jul. 8, 2019, the contents of which is hereby incorporated by reference in their entirety.
Filing Document | Filing Date | Country | Kind |
---|---|---|---|
PCT/US2020/041116 | 7/8/2020 | WO |
Number | Date | Country | |
---|---|---|---|
62871675 | Jul 2019 | US |