CAR T-CELLS RECOGNIZING CANCER-SPECIFIC IL 13Ra2

Provided are specific binding molecules, or fragments thereof, that bind to an epitope of IL13Rα2, a receptor polypeptide preferentially found on the surface of cancer cells rather than healthy cells. Exemplary specific binding molecules are bispecific binding molecules that comprise a fragment of an IL13Rα2 binding molecule and a peptide providing a second function providing a signaling function of the signaling domain of a T cell signaling protein, a peptide modulator of T cell activation, or an enzymatic component of a labeling system. Also provided are polynucleotides encoding such a specific binding molecule (e.g., bispecific binding molecule), vectors, host cells, pharmaceutical compositions and methods of preventing, treating or ameliorating a symptom associated with a cancer disease such as a solid tumor disease (e.g., glioblastoma multiforme).

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a continuation of U.S. patent application Ser. No. 16/675,861, filed Nov. 6, 2019, now U.S. Pat. No. 11,673,935, which is a continuation of U.S. patent application Ser. No. 15/545,950 filed Jul. 24, 2017, which is a national phase application under 35 U.S.C. § 371 of International Application No. PCT/US2016/014985 filed Jan. 26, 2016, which claims the priority benefit under 35 U.S.C. § 119(e) of Provisional U.S. Patent Application No. 62/107,980, filed Jan. 26, 2015 and Provisional U.S. Patent Application No. 62/245,771, filed Oct. 23, 2015, the disclosures of which are incorporated herein by reference in their entireties.

INCORPORATION BY REFERENCE OF MATERIAL SUBMITTED ELECTRONICALLY

The instant application contains a Sequence Listing which has been submitted in XML format and is hereby incorporated by reference in its entirety. Said XML copy, created on Oct. 3, 2023, is named “ARCDP0696USC2V2” and is 145,714 bytes in size.

FIELD OF THE DISCLOSURE

The disclosure relates generally to the fields of cancer biology and to molecular antibody-receptor technology.

BACKGROUND

Cancer is a major threat to human and non-human animal health, leading to reduced quality of life and, in too many cases, death. The burden placed on national, regional and local healthcare organizations to treat and prevent the various forms of cancer is significant in terms of the resources and manpower required. One of the main weapons vertebrates, including humans, have to combat disease is a functioning immune system. A brief consideration of immunotherapies to treat or prevent cancer might lead one to conclude that the effort held out little hope of success because immune systems guard against foreign, or non-self, materials and cancer cells arise from within, i.e., they are self materials. Continued progress in our understanding of cancer and immunology is modifying that view, however.

Mutant antigens are powerful targets for tumor destruction, e.g., in mice, and tumor-infiltrating lymphocytes targeting these mutations cause durable tumor regression in patients. Nevertheless, non-mutant antigens have been presumed by many scientists to be cancer-specific or “relatively cancer-specific” and safe antigens for vaccine approaches. However, adoptively transferred T cells can be orders of magnitude more effective and destructive than vaccinations. As a result, targeting MAGE-A3, HER-2 or CEA with T cells has caused death or serious toxicity in clinical trials now halted (8-11). As was shown in 2002, cancer cells with extremely high or very low expression levels of a target antigen differ only in the induction of immune responses, but not at the effector phase (15).

The high affinity interleukin-13 receptor α2 (IL13Rα2) is selectively expressed at a high frequency by glioblastoma multiforme (GBM) as well as several other tumor types. One approach for targeting this tumor-specific receptor utilizes the cognate ligand, IL-13, conjugated to cytotoxic molecules. This approach, however, lacks specificity because the lower affinity receptor for IL-13, IL13Rα1, is widely expressed by normal tissues.

Most human cancers lack specific antigens that are predictably present and serve as effective targets for eradication by T cells. Every cancer cell type harbors a unique set of mutations causing different tumor-specific antigens. Identifying an effective unique antigen and isolating an appropriate TCR for transduction of autologous T cells for adoptive immunotherapy is still difficult despite the enormous technological progress being made. Adoptive immunotherapy using antibodies or T cells is clinically as well as experimentally the most effective immunotherapy, at least when clinically relevant cancers are considered (22). The remarkable success of adoptive immunotherapy with chimeric antibody receptors (CARs) and bispecific T cell engaging proteins (BiTEs) is, however, largely restricted to those specific for CD19/CD20-eradicating B cell malignancies and normal B cells in patients, i.e., hematopoietic cancers. Thus, there is a need to identify shared, yet tumor-specific, antigens on a wide range of solid tumors, and a concomitant need to develop prophylactics and therapeutics that can diagnose, prevent, treat or ameliorate a symptom of these cancers, along with methods for diagnosing, preventing and treating various cancers.

SUMMARY

Disclosed herein are T cells expressing a chimeric antigen receptor (i.e., CAR) that specifically recognizes and binds to the α2 Interleukin 13 Receptor (i.e., IL13Rα2). The IL13Rα2-specific CARs, generally referred to herein as 47-CARs, when expressed in T cells effectively target and kill IL13Rα2-positive target cells. Also disclosed is evidence establishing that 47-CARs with a short spacer region, or SSR (i.e., 47-CAR.SSR), exhibit greater capacity to induce IL2-production in an antigen-dependent manner. Further disclosed herein is experimental evidence that 47-CAR.SSR T cells have potent anti-tumor activity in vivo.

The disclosure provides (i) the sequences of heavy (SEQ ID NO:7) and light (SEQ ID NO:8) chain variable regions of a monoclonal antibody (i.e., the clone 47 antibody) specifically targeting human tumor-associated antigen, IL13Rα2, and (ii) data demonstrating the functionality of the protein encoded by the heavy and light chain cDNAs in the format of an scFv antibody or fusion to other functional moieties. The sequences of the heavy and light chain constant regions were also determined and were found to be identical to the corresponding sequences in Genbank Acc. No. DQ381544.1. In particular, the CH1 sequence of the clone 47 antibody is set forth in SEQ ID NO:104, CH2 in SEQ ID NO:105 and CH3 in SEQ ID NO:106; the light chain constant region sequence of the clone 47 antibody is set forth in SEQ ID NO:107; and the hinge region of the clone 47 antibody in SEQ ID NO:108. The heavy and light chain can be arranged in different formats, such as single-chain antibody, diabodies, bi- and tri-specific antibodies, fusions with therapeutic proteins and other moieties, human or humanized whole antibodies as well as human or humanized Fab fragments and other functional derivatives. The single-chain antibody or other arrangements of the protein encoded by the heavy and light chains, e.g., a bispecific binding molecule, may be expressed and conjugated to therapeutic carriers (e.g., viruses, cells, nanomaterials) for specific delivery of therapeutic to IL13Rα2-overexpressing tumors or for imaging tumor burden.

Proteins expressed by tumor cells but not by normal cells are attractive molecules for the selective delivery of cytotoxic molecules. Accordingly, interleukin-13 receptor α2 (IL13Rα2), the high affinity receptor for interleukin-13 (IL-13), is a promising candidate. IL13Rα2 is expressed at a high frequency in the aggressive and incurable form of primary brain tumor known as glioblastoma multiforme (GBM) (1-3), as well as by other solid tumors (4). In contrast, normal tissues express little to no IL13Rα2, with the exception of the testes (6). Notably, IL13Rα1, a different receptor with low affinity for IL-13, is expressed ubiquitously by many tissues (7-9), making it a poor candidate for selective targeting of tumor-specific immunotherapeutic applications.

Several studies have investigated the therapeutic properties of an IL-13 fusion protein conjugated to a recombinant cytotoxin derived from Pseudomonas exotoxin A (IL-13PE) that induces apoptosis in IL13Rα2-expressing glioma cells in vitro, in preclinical animal models, and in patients tested in clinical trials (17-22). Such agents, however, lack a high specificity of interaction with IL13Rα2 because they alternatively bind to the ubiquitously expressed IL13Rα1. Therefore, developing highly selective antibody fragments that can be combined with effectors (e.g., T-cells, toxins) for specificity to IL13Rα2-expressing cells is expected to yield therapeutically beneficial results.

The disclosure captures the tumor specificity of IL13Rα2 by providing protein binding partners specific for IL13Rα2, rather than mimicking IL13 itself, which would result in a molecule exhibiting a capacity to bind to both IL13Rα1 and IL13Rα2. In addition, the disclosure provides a polynucleotide encoding one of these cancer-specific IL13Rα2 binding partners, including polynucleotides comprising codon-optimized coding regions for binding partners specific for an epitope of one of these IL13Rα2 binding partners. Expressly contemplated are fusion proteins or chimeras that comprise an IL13Rα2 binding partner as defined above in operable linkage to a peptide providing a second function, such as a signaling function of the signaling domain of a T cell signaling protein, a peptide modulator of T cell activation or an enzymatic component of a labeling system. Exemplary T cell signaling proteins include 4-1BB (CD137), CD3ζ, and fusion proteins, e.g., CD28-CD3ζ and 4-1BB-CD3ζ. 4-1BB (CD137) and CD28 are co-stimulatory molecules of T cells; CD3ζ is a signal-transduction component of the T-cell antigen receptor. In certain embodiments, the IL13Rα2-specific CAR may be expressed in two fragments that are inactive without the addition of an exogenous substance. By way of non-limiting example, the CAR would consist of two molecules: 1) the first molecule would contain the IL13Rα2-specific scFc, a hinge, a transmembrane domain, a costimulatory domain, and a heterodimerizer domain (Exto-TM-HD), and 2) the first molecule would contain a transmembrane domain, a costimulatory domain, a heterodimerizer domain, a CD3ζ activating domain (Cyto-HD) (Wu et al; Science. 2015 Oct. 16; 350(6258):aab407). Expression of Exto-TM-HD and Cyto-HD in cells would result in an inactive IL13Rα2-CAR unless a small molecule, for example but not limited to, a rapalog A/C Heterodimerizer is added that links Exto-TM-HD and Cyto-HD, allowing for pharmacological control of IL13Rα2-CAR activity. The peptide or protein providing a second function may provide a modulator of T cell activation, such as IL15, IL15Rα, of an IL15/IL15Rα fusion, or it may encode a label or an enzymatic component of a labeling system useful in monitoring the extent and/or location of binding, in vivo or in vitro. Agent encoding these prophylactically and therapeutically active biomolecules placed in the context of T cells, such as autologous T cells, provide a powerful platform for recruiting adoptively transferred T cells to prevent or treat a variety of cancers in some embodiments of the disclosure. Codon optimization of the coding regions for binding partners specific for epitopes found on cancer cells provides an efficient approach to delivery of the diagnostic, prophylactic, and/or therapeutic proteins disclosed herein.

In one aspect, the disclosure provides an Interleukin 13 Receptor α2 (IL13Rα2) binding partner comprising the antibody heavy chain variable fragment (VH) complementarity determining region 1 (CDR1) of SEQ ID NO:1, the VH CDR2 of SEQ ID NO: 2, the VH CDR3 of SEQ ID NO: 3, the light chain (VL) complementarity determining region 1 (CDR1) of SEQ ID NO: 4, the VL CDR2 of SEQ ID NO: 5, and the VL CDR3 of SEQ ID NO: 6, wherein the IL13Rα2 binding partner specifically binds to an epitope of IL13Rα2. In some embodiments, the VH sequence is set forth as SEQ ID NO: 7 and in some of the same and some different embodiments, the VL sequence is set forth as SEQ ID NO: 8.

A related aspect of the disclosure provides a bispecific binding molecule comprising a fragment of the IL13Rα2 binding partner described herein that binds to the IL13Rα2 epitope covalently linked to a peptide providing a second function to form a bispecific binding molecule. In some embodiments, the second function of the peptide is selected from the group consisting of a signaling function of the signaling domain of a T cell signaling protein, a peptide modulator of T cell activation, and an enzymatic component of a labeling system. In some embodiments, the fragment is a single-chain variable fragment (scFv), which may be contained within a bi-specific T-cell engager (BiTE) or a chimeric antigen receptor (CAR). Some embodiments are provided wherein the bispecific binding molecule as described herein is conjugated to a therapeutic carrier.

Another aspect of the disclosure is drawn to a pharmaceutical composition comprising the IL13Rα2 binding partner as described herein and a pharmaceutically acceptable carrier, adjuvant or diluent.

A related aspect provides a kit comprising the pharmaceutical composition described herein and a protocol for administration of the composition. Also related is an aspect providing a polynucleotide encoding the IL13Rα2 binding partner as described herein and a vector comprising the polynucleotide as described herein. Yet another aspect is directed to a host cell comprising the polynucleotide described herein or the vector described herein.

Yet another aspect of the disclosure provides a method of preventing, treating or ameliorating a symptom of a cancer disease comprising administering a therapeutically effective amount of the pharmaceutical composition as described herein. In some embodiments, the cancer is a solid tumor, such as a glioblastoma multiforme (GBM). In some embodiments, the cancer is treated by inhibiting the growth rate of the solid tumor. In some embodiments, the symptom ameliorated is pain.

More particularly, one aspect of the disclosure is drawn to an IL13Rα2-specific chimeric antigen receptor (CAR) comprising: (A) each of the amino acid sequences of: NYLMN (SEQ ID NO: 1); RIDPYDGDIDYNQNFKD (SEQ ID NO: 2); GYGTAYGVDY (SEQ ID NO: 3); RASESVDNYGISFMN (SEQ ID NO: 4); AASRQGSG (SEQ ID NO: 5); and QQSKEVPWT (SEQ ID NO: 6), (B) a hinge region, (C) a transmembrane domain, and (D) an endodomain comprising a signaling domain a CD3 zeta chain and a signaling domain of CD28. In some embodiments, the endodomain further comprises a signaling domain of one or more of: CD137, CD134, CD27, CD40, ICOS, and Myd88, optionally, wherein the endodomain comprises one or more of the amino acid sequences of SEQ ID NOs: 68, 70, 72, 74, 76, and 78. In some embodiments, the hinge region comprises the amino acid sequence of SEQ ID NO: 35 or SEQ ID NO: 37. In some embodiments, the IL13Rα2-specific CAR comprises a transmembrane domain of CD28. In some embodiments, the IL13Rα2-specific CAR comprises the amino acid sequence of SEQ ID NO: 39. In some embodiments, the CD3 zeta chain signaling domain comprises the amino acid sequence of SEQ ID NO: 41. In some embodiments, the IL13Rα2-specific CAR comprises the amino acid sequence of SEQ ID NO: 47. In some embodiments, the IL13Rα2-specific CAR comprises the amino acid sequence of SEQ ID NO 49 or 51. In some embodiments, the IL13Rα2-specific CAR comprises one or both of the amino acid sequences of SEQ ID NO: 7 and/or SEQ ID NO: 8. In some embodiments, the IL13Rα2-specific CAR of claim 9, wherein the amino acid sequence of SEQ ID NO: 7 is fused to the amino acid sequence of SEQ ID NO: 8 through a linker. In some embodiments, the linker comprises the amino acid sequence of EEGEFSEAR (SEQ ID NO 10). In some embodiments, the IL13Rα2-specific CAR comprises the amino acid sequence of SEQ ID NO: 13. In some embodiments, the IL13Rα2-specific CAR comprises the amino acid sequence of SEQ ID NO: 53 or SEQ ID NO: 55.

In a related aspect, the disclosure provides a nucleic acid encoding any of the IL13Rα2-specific CARs disclosed or described herein. In some embodiments, the nucleic acid comprises the sequence of SEQ ID NO: 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, or 65.

In yet another aspect, the disclosure provides a vector comprising a nucleic acid disclosed or described herein. In some embodiments, the vector is a retroviral vector.

In another aspect, the disclosure provides a host cell comprising a vector disclosed or described herein. In some embodiments, the host cell is a human host cell. In some embodiments, the host cell is a T-lymphocyte. In some embodiments, the host cell is a natural killer cell.

In a related aspect, the disclosure provides a cell population comprising a host cell disclosed or described herein. In some embodiments, the cell population comprises at least 107 host cells.

Another aspect is drawn to a pharmaceutical composition comprising an IL13Rα2-specific CAR as disclosed or described herein, a nucleic acid as disclosed or described herein, a vector as disclosed or described herein, a host cell as disclosed or described herein, or a cell population as disclosed or described herein, and a pharmaceutically acceptable carrier.

Another aspect of the disclosure provides a method of treating a cancer in a subject, comprising administering to the subject a cell population as disclosed or described herein, in an amount effective to treat the cancer in the subject. In some embodiments, the cancer is colon cancer. In some embodiments, the host cells of the cell population are cells obtained from the subject. In some embodiments, the cells obtained from the subject are T-lymphocytes. In some embodiments, the cells obtained from the subject are natural killer cells.

Another aspect of the disclosure provides an IL13Rα2-specific chimeric antigen receptor (CAR) comprising: (A) an ectodomain comprising each of the amino acid sequences of: (i) NYLMN (SEQ ID NO: 1); (ii) RIDPYDGDIDYNQNFKD (SEQ ID NO: 2); (III) GYGTAYGVDY (SEQ ID NO: 3); (iv) RASESVDNYGISFMN (SEQ ID NO: 4); (v) AASRQGSG (SEQ ID NO: 5); and (vi) QQSKEVPWT (SEQ ID NO: 6); (B) a spacer region; (C) a transmembrane domain; and (D) an endodomain selected from the group consisting of CD3.ζ, CD28.ζ, CD28.OX40.ζ, CD28.41BB.ζ and 41BB.ζ. In some embodiments, the spacer region comprises no more than 100 amino acids, or no more than 50 amino acids, or no more than 25 amino acids, or the spacer region comprises SEQ ID NO:103 (PKSCDKTHTCPPCPAPEL) from the IgG1 hinge region. In some embodiments, the transmembrane domain comprises the transmembrane domain of CD28, such as a transmembrane domain comprising the amino acid sequence of SEQ ID NO:39, or CD8α. In some embodiments, the endodomain further comprises a signaling domain of one or more of: CD137, CD134, CD27, CD40, ICOS, and Myd88. In some embodiments, the endodomain comprises one or more of the amino acid sequences of SEQ ID NOs: 68, 70, 72, 74, 76, and 78. In some embodiments comprising the CD3 zeta chain signaling domain, the IL13Rα2-specific CAR comprises the amino acid sequence of SEQ ID NO: 41. In some embodiments, the IL13Rα2-specific CAR comprises the amino acid sequence of SEQ ID NO: 47. In some embodiments, the IL13Rα2-specific CAR comprises the amino acid sequence of SEQ ID NO 49 or 51. In some embodiments, the IL13Rα2-specific CAR comprises one or both of the amino acid sequences of SEQ ID NO: 7 and/or SEQ ID NO: 8.

The disclosure also contemplates embodiments wherein the amino acid sequence of SEQ ID NO: 7 is fused to the amino acid sequence of SEQ ID NO: 8 through a linker. In some embodiments, the linker comprises the amino acid sequence of EEGEFSEAR (SEQ ID NO 10). In some of these embodiments, the IL13Rα2-specific CAR comprises the amino acid sequence of SEQ ID NO: 13. In some embodiments, the IL13Rα2-specific CAR comprises the amino acid sequence of SEQ ID NO: 53 or SEQ ID NO: 55.

Another aspect of the disclosure is drawn to a nucleic acid encoding the IL13Rα2-specific CAR disclosed herein. In some embodiments, the nucleic acid comprises the sequence of SEQ ID NO: 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, and 65.

Still another aspect of the disclosure is drawn to a vector comprising the nucleic acid disclosed herein. In some embodiments, the vector is a retroviral vector.

Yet another aspect of the disclosure is a host cell comprising the vector disclosed herein. In some embodiments, the host cell is a human host cell. In some embodiments, the host cell is a T-lymphocyte or a natural killer cell. In some embodiments, the cells obtained from the subject are T cells, and/or other lymphocytes including, but not limited to, NKT cells, γδ T cells, mucosa associated invariant T cells or MAIT cells, and innate lymphoid cells. In addition, stem and/or progenitor cells may be obtained from the subject that are subsequently differentiated into the aforementioned immune cells.

Another aspect of the disclosure is a cell population comprising the host cell disclosed herein. In some embodiments, the cell population comprises at least 107 host cells.

In another aspect, the disclosure provides a pharmaceutical composition comprising an IL13Rα2-specific CAR as disclosed herein, a nucleic acid as disclosed herein, a vector as disclosed herein, a host cell as disclosed herein, or a cell population as disclosed herein, and a pharmaceutically acceptable carrier.

Yet another aspect of the disclosure is a method of treating a cancer in a subject, comprising administering to the subject a cell population as disclosed herein, in an amount effective to treat the cancer in the subject. In some embodiments, the cancer is colon cancer. In some embodiments, the host cells of the cell population are cells obtained from the subject. In some embodiments, the cells obtained from the subject are T cells, and/or other lymphocytes including, but not limited to, NKT cells, γδ T cells, mucosa associated invariant T cells or MAIT cells, and innate lymphoid cells. In addition, stem and/or progenitor cells may be obtained from the subject that are subsequently differentiated into the aforementioned immune cells.

In some embodiments, the immune or stem and/or progenitor cells that are genetically modified to be IL13Rα2-specific by expressing a CAR or BITE molecule may be further genetically modified to enhance their anti-tumor activity. Non-limiting examples of additional genetic modification include, but are not limited to: i) CARs or BITEs that are specific for other antigens expressed on tumor cells or within the tumor environment, ii) cytokines (e.g., various interleukins such as IL7, IL12, IL15, IL21), iii) chimeric cytokine receptors (e.g., IL7R, IL15R), iv) chemokine receptors (e.g., CCR2b, CXCR2), iv) chimeric activating receptors (e.g., IL4/IL7R, IL4/IL2R, TGFβ/TLR4R), v) silencing negative regulators (e.g., PD1, SHP1), vi) silencing endogenous TCR expression, and vii) inducible suicide genes (e.g., CD20, truncated EGFR, inducible caspase 9).

Other features and advantages of the disclosure will become apparent from the following detailed description, including the drawing. It should be understood, however, that the detailed description and the specific examples, while indicating preferred embodiments, are provided for illustration only, because various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from the detailed description.

BRIEF DESCRIPTION OF THE DRAWING

The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawing(s) will be provided by the Office upon request and payment of the necessary fee.

FIG. 1A-D. Characterization of antigen recognition and screening of hybridoma clones. A, binding of B-D13 mAb to ELISA plates coated with rhIL13Rα2hFc at 0.1 and 1 μg/ml. B, binding of IL13Rα2 mAb to native and denatured (at 95° C. in the presence of β-mercaptoethanol) rhIL13Rα2hFc in a plate-bound ELISA. A paired t test was used to evaluate the difference between control groups (n=4). *, p<0.1; ***, p<0.001. Error bars represent S.D. These data are representative of two independent experiments. C, screening of selected hybridoma populations against rhIL13Rα2hFc in a plate-bound ELISA. D, screening of selected hybridoma populations against rhIL13Rα2hFC using a Western blot.

FIG. 2A-D. The IL13Rα2 (clone 47) mAb specifically binds to rhIL13Rα2 and IL13Rα2 expressed on the cell surface of CHO cells. A, binding of IL13Rα2 (clone 47, 83807, and B-D13) mAbs to rhIL13Rα2 in a plate-bound ELISA. B, binding of the IL13Rα2 (clone 47) mAb to human IL13Rα2 expressed on the surface of CHO cells. C, cross-reactivity of the IL13Rα2 (clone 47) mAb with hrIL13Rα1. D, cross-reactivity of IL13Rα2 (clones 47, 83807, and B-D13) mAbs with mouse rIL13Rα2. Error bars represent S.D.

FIG. 3A-D. Binding of IL13Rα2 mAb to glioma cells. A, flow charts of IL13Rα2 (clones 47, 83807, and B-D13) mAbs binding to the surface of glioma cells, normal human primary astrocytes, and HEK cells transfected with IL13Rα2. B, data of the median fluorescence intensity of binding between the IL13Rα2 (clones 47, 83807, and B-D13) mAbs to various cell lines analyzed by flow cytometry. Numbers above the bars represent the difference in the binding of clone 47 when compared with clone B-D13 for each cell line. The color key is the same for A and B. C, mRNA expression for IL13Rα2 in glioma cells as well as normal human primary astrocytes. D, panels a-c, flow cytometry demonstrating the specific binding of the IL13Rα2 (clone 47) mAb to GFP-tagged U251 glioma cells from an intracranial xenograft (xeno). The curve with a clear area under the curve in sub-panel b depicts the binding of mAb IL13Rα2 (clone 47) to GFP negative cells; the curve with a clear area under the curve in sub-panel c depicts the binding of mAb IL13Rα2 (clone 47) to GFP positive cells. Curves in sub-panels b and c with gray areas under the curves show the results when exposing control IgG to GFP-negative (sub-panel b) or GFP-positive (sub-panel c) cells. neg, negative. A, area; SSC-A, side scatter area; APC-A, allophycocyanin area.

FIG. 4A-C. The affinity between the IL13Rα2 (clone 47) mAb and rhIL13Rα2. The kinetics of interaction of IL13Rα2 (clone 47) mAb (A) and the commercially available mAb clones 83807 (B) and B-D13 (C) with rhIL13Rα2 as visualized by SPR in a Biacore 3000 are shown. The rhIL13Rα2 was injected at concentrations ranging from 1 to 100 nM (1 nM, 2.5 nM, 5 nM, 7.5 nM, 10 nM, 15 nM, 20 nM, 25 nM concentrations shown, lower to upper curves) at a constant flow rate of 20 μl/minute over immobilized antibodies and over a control dextran surface (these values were subtracted from the signal). The association and dissociation phases were monitored for 300 s by following the change in SPR signal (colored curves) given in RU. Black curves represent the fit of the data to a one-site binding model. For derived kinetic parameters, see Table 1. Lower panels show residuals from a one-site binding model, indicating an excellent fit.

FIG. 5A-C. The IL13Rα2 (clone 47) mAb competes with rhIL-13 for the binding site of IL13Rα2. A, using a competitive binding plate assay, the IL13Rα2 (clone 47) mAb but not control mIgG or antibody clones 83807, B-D13, and YY-23Z significantly abolished the binding of rhIL-13 to the rhIL13Rα2Fc chimera absorbed to plastic. One-way analysis of variance followed by Dunnett's post hoc test was performed. Data from a single representative experiment are shown. B, recombinant human IL-13 competes with the IL13Rα2 (clone 47) mAb for the binding site of WT IL13Rα2 but not with the 4-amino acid (4aa) mutant IL13Rα2 expressed on the surface of HEK cells. C, the IL13Rα2 (clone 47) mAb competes with rhIL-13 for the binding site of the WT and 4-amino acid mutant form of IL13Rα2. A paired t test was performed. Data represent the summary of three independent experiments shown in B and C. *, p<0.05; **, p<0.01; ***, p<0.001. Error bars represent S.D.

FIG. 6A-B. The contribution of Tyr207, Asp271, Tyr315, and Asp318 residues of IL13Rα2 to the binding of the IL13Rα2 (clone 47) mAb. A, variants of cDNA encoding individual mutations to Ala or a combinatorial 4-amino acid mutant (4aa mut) of IL13Rα2 was generated. HEK cells were transfected with a control vector or a vector encoding the IL13Rα2 variants. After 48 hours, binding of the IL13Rα2 (clone 47) mAb to the surface of transfected cells was analyzed by flow cytometry. Anti-IL13Rα2 antibody clones 83807 and B-D13 were used as reference antibodies in this assay. Binding of antibodies was determined as the percentage of positive cells. The ratio of bound clones was determined for each IL13Rα2 mutant and compared with that of the wild-type receptor. One-way analysis of variance followed by Dunnett's post hoc test was performed. Data represent a summary of four independent experiments. Error bars represent S.D. B, representative graphs of flow cytometry data demonstrating the binding of clone 47 or rhIL-13 to the WT and 4-amino acid-mutated variant of the IL13Rα2 receptor expressed on the surface of HEK cells. Filled curves: negative control, staining with isotype control IgG+secondary antibody; Open curves: staining with the anti-IL13Rα2 (clone 47) monoclonal antibody+secondary antibody. A, area; APC-A, allophycocyanin area; FITC-A, fluorescein isothiocyanate area.

FIG. 7A-C. Effect of N-linked glycosylation on the binding of IL13Rα2 to recombinant IL13Rα2. A, binding of IL13Rα2 to control and Pngase F-treated rhIL13Rα2. Plates were coated with hrIL13Rα2 at 1 μg/ml and treated with native buffer or with 1 milliunit/well Pngase F in native buffer for 3 hours at 37° C. An ELISA for binding of the IL13Rα2 (clone 47) mAb in comparison with antibody clones B-D13, 83807, and YY-23Z and rhIL-13 was performed, and the data of one representative experiment from three independent experiments are shown. A paired t test was used to evaluate the difference between control and Pngase F-treated groups (n=4). *, p<0.5; **, p<0.01; ***, p<0.001. B, a Western blot shows the lower molecular weight of Pngase F-treated rhIL13Rα2 due to removal of N-linked glycosylation adducts from the molecule. C, flow cytometry shows the binding of IL13Rα2 mAbs to IL13Rα2-expressing U251 and HEK293 cells treated with 1 milliunit of Pngase F for 1 hour at 37° C. The data are representative of three independent experiments. A paired t test was used to evaluate the difference between control and Pngase F-treated groups. *, p<0.5. MFI, mean fluorescence intensity. Error bars represent S.D.

FIG. 8. The IL13Rα2 (clone 47) mAb recognizes IL13Rα2 in GBM tissues and in a human glioma xenograft. Immunohistochemistry on frozen tissue sections from three human GBM samples and a U251 xenograft was performed with the IL13Rα2 (clone 47) mAb or mIgG at a concentration of 3 μg/ml. Staining of GBM tissues demonstrates positive staining of the majority of cells in sample 1, positive reactivity in only a fraction of the cells in sample 2, and negative staining in sample 3. Staining in all three samples was performed in the same experiment. Positive staining was also detected in U251 xenograft tissue. Arrows point to individual positive cells. Scale bars=100 μm.

FIG. 9A-B. The IL13Rα2 (clone 47) mAb improves the survival of mice in an orthotopic human glioma xenograft model. A, the survival of animals injected with U251 glioma cells (2.5×104) alone or in combination with either control IgG or the IL13Rα2 (clone 47) mAb. B, a representative photomicrograph of 10-μm-thick tissue sections stained with H&E from mice injected with U251 cells alone (panels a and b) or in combination with mIgG (panels c and d) or mAbIL13Rα2 (clone 47) (panels e and f). Arrows point to the tumor and invading cells. Scale bars (panels a, c, and e)=100 μm. Scale bars (panels b, d, and f)=100 μm.

FIG. 10A-B. A competitive binding assay for the IL13Rα2 (clone 47) mAb to the surface of N10 glioma cells. A. The IL13Rα2 (clone 47) mAb was pre-incubated with 10× excess rhIL13Rα2 for 30 minutes on ice. N10 cells were subsequently incubated with isotype control mIgG or IL13Rα2 (clone 47) mAb alone or in the presence of rhIL13Rα2 and bound antibodies were analyzed by flow cytometry. B. N10 glioma cells were pre-incubated either with 10× excess rhIL13 (left panel) or with 10× excess of IL13Rα2 (clone 47) mAb for 30 minutes on ice (right panel). N10 cells were subsequently incubated with isotype control mIgG, IL13Rα2 (clone 47) mAb or rhIL13. Bound antibodies or rhIL13 were detected with secondary antibodies and analyzed by flow cytometry. Data are presented as % of positive cells.

FIG. 11. The effects of IL13Rα2 (clone 47) mAb on the survival of mice with an established human U251 glioma. Mice were intracranially-injected with 2.5×104 U251 glioma cells and treated three days later with a single injection of PBS (n=7) or 10 μg IL13Rα2 (clone 47 or B-D13) mAb (n=7). The analysis of the animal's survival was performed using the Log-rank test. Median survival was determined to be 27 days in the PBS group, versus 23 and 35 days in the groups treated with B-D13 and 47 IL13Rα2 mAb, respectively (p>0.05).

FIG. 12. Binding of IL13Rα2 clone 47 phages with IL13Rα2hFc in plate ELISA. These data demonstrate that phages presenting scFv IL13Rα2 (clone 47) are positively selected against IL13Rα2Fc chimeric protein after 3 rounds of biopanning.

FIG. 13. Specificity of binding scFv IL13Rα2 clone 47 with IL13Rα2hFc-competitive assay. These data show that binding of the scFvIL13Rα2 (clone 47) presented on the phage surface to recombinant IL13Rα2 is completely abolished by parental monoclonal antibody (clone 47), but not other antibodies against IL13Rα2. It indicates that scFvIL13Rα2 (clone 47) and parental monoclonal antibody (clone 47) share the epitope (i.e., recognition site) on the IL13Rα2 molecule. Each data point is an average of 3 independent replicates in all figures. Data presented as mean f SEM. ***p<0.001.

FIG. 14. Binding of soluble scFv IL13Rα2 (clone 47) with IL13Rα2hFc chimera. These data show that soluble scFvIL13Rα2 (clone 47) generated in a prokaryotic expression system (E. coli) binds specifically to IL13Rα2Fc recombinant protein. Parental antibody, mAb IL13Rα2 (clone 47), and control mouse IgG served as positive and negative controls, respectively

FIG. 15. The effect of mesenchymal stem cells secreting scFvIL13Rα2-sTRAIL fusion protein on the U87-IL13Rα2 glioma cell line. These data show that mesenchymal stem cells modified to secrete a genetic fusion of scFvIL13Rα2 (clone 47) with TRAIL protein exhibit a therapeutic effect in the IL13Rα2-expressing U87 glioma cell line. The results establish the efficacy of conjugating the scFV to a TRAIL cytokine. The amount of cancer cell killing is equivalent to the use of TRAIL alone without the scFV, but it is expected that the scFV-TRAIL would be less harmful to non-cancer tissues, given the specificity conferred by the scFv targeting IL13Rα2.

FIG. 16. Schematic maps of retroviral vector encoding IL13Rα2-specific scFv CARs. The CAR consists of the immunoglobulin heavy-chain leader peptide, the IL13Rα2-specific scFv clone 47 (M47), a short hinge (SH) or long hinge (LH), a transmembrane domain (TM) derived from CD28, and a CD28. ζ endodomain. LTR: long terminal repeat (retroviral backbone). Domains are identified as block structures. Maps are not to scale.

FIG. 17. IL13Rα2-scFv CAR T cell agent: Expression of αCD3.ζ relative to αGAPDH of CAR agent in T cells. SH: short hinge. LH: long hinge.

FIG. 18A-B. IL13Rα2-scFv CARs are expressed on the surface of T cells. IL13Rα2-CAR T cells were generated by retroviral transduction and CAR expression was determined by FACS analysis. Short hinge CARs were detected with an antibody specific for murine scFV. Long hinge CARs were detected with an antibody specific for the long hinge. Isotype antibody control: open curve; Specific Antibody: filled curve.

FIG. 19. Functional characterization of IL13Rα2-CAR T cells—Cytotoxicity. Standard 51Chromium cytotoxicity assays were performed with Raji (IL13Rα1−/IL13Rα2−), 293T (IL13Rα1+/IL13Rα2−), 293T genetically modified to express IL13Rα2 cells (293T-IL13Rα2; IL13Rα1+/IL13Rα2+), or U373 (IL13Rα1+/IL13Rα2+) cells as targets. As effectors nontransduced (NT) T cells, IL13Rα2-CAR.SH.CD28.ζ T cells, IL13Rα2-CAR.LH.CD28.ζ T cells, IL13Rα2-CAR.SH.Δ T cells, or IL13Rα2-CAR.LH.Δ T cells were used. Only IL13Rα2-CAR.SH.CD28.ζ T cells and IL13Rα2-CAR.LH.CD28.ζ T cells killed with IL13Rα2+ target cells (U373 and 293T-IL13Rα2; n=4). T cells expressing nonfunctional CARs (IL13Rα2-CAR.SH.Δ and IL13Rα2-CAR.LH.Δ) had not cytolytic activity, demonstrating that the killing activity depends on the expression of a functional IL13Rα2-CAR. NT T cells killed none of the targets, further confirming specificity.

FIG. 20. Functional characterization of IL13Rα2-CAR T cells—IFNγ and IL2 Cytokine secretions. A. NT T cells, IL13Rα2-CAR.SH.CD28.ζ T cells, IL13Rα2-CAR.LH.CD28.ζ T cells, IL13Rα2-CAR.SH.Δ T cells, or IL13Rα2-CAR.LH.Δ T cells were co-cultured with U373 cells for 24 to 48 hours (n=4). Only IL13Rα2-CAR.SH.CD28.ζ T cells and IL13Rα2-CAR.LH.CD28.ζ T cells secreted IFNγ demonstrating target cell recognition in contrast to IL13Rα2-CAR.SH.Δ T cells, IL13Rα2-CAR.LH.Δ T cells or NT T cells. B. NT T cells, IL13Rα2-CAR.SH.CD28.ζ T cells, IL13Rα2-CAR.LH.CD28.ζ T cells, IL13Rα2-CAR.SH.Δ T cells, or IL13Rα2-CAR.LH.Δ T cells were co-cultured with U373 cells for 24 to 48 hours (n=4). Only IL13Rα2-CAR.SH.CD28.ζ T cells secreted IL2, demonstrating that IL13Rα2-CAR.SH.CD28.ζ induces superior T cell activation in comparison to IL13Rα2-CAR.LH.CD28.ζ. IL13Rα2-CAR.SH.Δ T cells, IL13Rα2-CAR.LH.Δ T cells or NT T cells also did not induce IL2 production.

FIG. 21. IL13Rα2-SH CARs have anti-glioma activity in vivo. Severe combined immunodeficient (SCID) mice were injected with 1×105 firefly luciferase expressing U373 cells intracranially. On day 7 mice were treated either with 1×106 IL13Rα2-CAR.SH.CD28.ζ T cells, IL13Rα2-CAR.LH.CD28.ζ T cells, IL13Rα2-CAR.SH.Δ T cells, or IL13Rα2-CAR.LH.Δ T cells (5 mice per group). Tumor growth was monitored by bioluminescence imaging. Only IL13Rα2-CAR.SH.CD28.ζ T cells had significant anti-glioma effects with 4/5 mice having a complete response.

FIG. 22. Properties of m47 CAR T cell agent. The m47-CAR T cells recognize IL13Rα2+, but not IL13Rα1+ targets. The data show that the short hinge CD28z-CAR (SH2) T cells perform better in terms of effector function than CD28z-CAR (SH3), CD28z-CAR (LH2), CD28z-CAR (LH3), CD28z-CAR (SH2Δ), or CD28z-CAR (SH3Δ).

FIG. 23. Functional comparison of m47 CAR T cell agents. Open curve: secondary antibody; Filled curve: IL13Rα2Fc+secondary antibody.

FIG. 24. The m47 CAR T cell agent is highly expressed after transduction. Open curve: secondary antibody; Filled curve: IL13Rα2Fc+secondary antibody.

FIG. 25. The m47 CAR T cell produce interferon γ and interleukin 2, but only after IL13Rα2 stimulation.

FIG. 26. IL13Rα2- and IL13Rα1-positive cell lines are made by genetic modification of HEK 293T cells. Filled curve: isotype antibody control; Open curve: specific antibody.

FIG. 27. The m47 CAR T cells kill only IL13Rα2+ cell lines. The in vitro experiments provide data establishing that m47 CAR T cells present a recombinant CAR protein on the cell surface that does not recognize IL4R, IL13Rα1 or any receptor other than its specific recognition of IL13Rα2. The specificity of the recognition extends to a specificity for only those cell lines expressing IL13Rα2.

FIG. 28. In vivo data comparing effect of m47 CAR T cell agent, untreated and NT-treated glioblastoma multiforme xenografts in nude mice. The U373 glioblastoma multiforme xenograft mouse model was used. At day 0, 1×105 GFP-ffluc U373 cells were administered per mouse. On day 7, 2×106 m47 CAR T cells or NT cells were administered. Untreated samples did not receive treatment on day 7. No exogenous interleukin 2 was administered and results of the survival analysis were recorded by serial bioluminescence imaging. n=3.

FIG. 29. The m47 CAR T cell agent prolonged the survival of nude mice with glioblastoma multiforme.

FIG. 30A-C. Characterization of IL13Rα2-CAR T cells. (A, B) Co-culture assay with recombinant protein demonstrated interferon 7 and interleukin 2 production in an IL14Rα2-dependent fashion; (C) Cytolytic activity in standard chromium release assay.

FIG. 31A-D. Generation of 47 CAR T cells. (A) Scheme of M47 CARs. All CARs contained an N-terminal leader sequence, a codon-optimized synthetic gene encoding M47 in scFv format, a spacer region, a CD28 transmembrane domain, and signaling domains derived from CD28 and CD3-ζ. The spacer region was either the IgG1 hinge (16 amino acids; short spacer region; M47-CAR.SSR.CD28.ζ) or the IgG1-CH2CH3 domain. LSR.Δ and SSR.Δ M47-CARs without signaling domains were constructed and served as controls. (C,B) CAR expression was confirmed using FACS analysis. Representative plots (B) and summary data (C) are shown (mean 74.1%-93.3%, n=5-6 per CAR construct). Open curve: secondary antibody; Filled curve: IL13Rα2Fc+secondary antibody. (D) Expression of full-length 47-CAR.SSR.CD28.ζ and 47-CAR.LSR.CD28.ζ by Western blot analysis using a CD3-ζ antibody.

FIG. 32. Phenotypic analysis of 47-CAR T cell lines. CAR T cells were analyzed for CD4 and CD8 surface expression using CD4-PacBlue and CD8-PerCP antibodies (BD Biosciences). The four CAR T cell lines analyzed for surface expression of CD4 and CD8 were SSR.Δ, SSR.CD28.ζ, LSR.Δ, and LSR.CD28.ζ. The histogram provides the results of the analysis, with light gray bars indicating CD4 expression and black bars indicating CD8 expression.

FIG. 33A-D. 47-CAR T cells release cytokines after stimulation with recombinant IL13Rα2 protein or IL13Rα2-positive cells. 47-CAR or non-transduced (NT) T cells were stimulated with recombinant IL13Rα1, IL13Rα2, or IL4Rα proteins. After 24 hours, IFNγ (A) was measured by ELISA (n=4). T cells expressing 47-CAR constructs, but not controls, expressed significant levels of IFNγ (p<0.001) when stimulated with recombinant IL13Rα2 protein in comparison to IL13Rα1 and IL4Rα stimulated T cells. 47-CAR T cells were co-cultured with Raji, U373 cells, 293T-GFP, and 293T-GFP/IL13Rα2 at a 1:2 E:T ratio. NT and CAR.Δ T cells served as controls. (B,C) After 24 hours, cytokines (IFNγ, IL2) were measured by ELISA (n=3). (B) U373 and 293T-GFP-IL13Rα2 (IFNγ); SSR.Δ vs SSR.CD28.ζ: p<0.001; LSR.Δ vs LSR.CD28.ζ: p<0.05. (C); U373 and 293T-GFP-IL13Rα2 (IL2); SSR.Δ vs SSR.CD28.ζ: p<0.01; LSR.Δ vs LSR.CD28.ζ: NS. (D) 4-hour cytotoxicity assay at an E:T ratio of 10:1 (n=4).

FIG. 34. LSR.CD28.ζ T cells show a self-activation phenotype during ex vivo expansion. T cells were analyzed for phosphor-CD3-ζ expression using CD247 (pY142)-AF647 antibody (BD Biosciences).

FIG. 35. Cell surface expression of IL13Rα1 and IL13Rα2. Cell lines were analyzed for IL13Rα1 and IL13Rα expression using primary goat anti-IL13Rα1 and anti-IL13Rα2 antibodies (AF152 and AF146, respectively; R&D) followed by secondary rabbit anti-goat IgG Alexa647 antibody (Life Technologies). Filled curve: isotype antibody control; Open curve: specific antibody.

FIG. 36A-D. Generation of SSR 47-CARs with CD28.OX40.ζ, CD28.41BB.ζ or 41BB.ζ endodomains. (A) Scheme of SSR 47-CARs. (B, C) CAR expression was confirmed using FACS analysis. Representative plots (B) and summary data (C) are shown. 47-CAR.SSR.CD28.OX40.ζ and 47-CAR.SSR.CD28.41BB.ζ: mean: 74.6%-77.5% (n=4); 47-CAR.SSR.CD28.41BB.ζ: mean: 4.9% (n=3). Open curve: secondary antibody; Filled curve: IL13Rα2Fc+secondary antibody. (D) Expression of 47-CAR.SSR.41BB.ζ, M47-CAR.SSR.OX40.CD28.ζ and M47-CAR.SSR.41BB.CD28.ζ by Western blot analysis.

FIG. 37A-B. Comparison of 47-CAR.SSR.CD28.ζ, 47-CAR.SSR.41BB.ζ, and 47-CAR.SSR.CD28.OX40.ζ T cells. (A) 47-CAR T cells were co-cultured with U373 cells at a 1:2 E:T ratio. NT and CAR.Δ T cells served as controls. After 24 hours, IFNγ and IL2 were measured by ELISA (n=3); SSR.Δ vs SSR.CD28.ζ (U373; IFNγ): p<0.001; SSR.Δ vs SSR.41BB.ζ (U373; IFNγ): p<0.05; SSR.Δ vs SSR.CD28.OX40.ζ for (U373; IFNγ): p<0.001; SSR.Δ vs SSR.CD28.ζ (U373; IL2): p<0.001; SSR.Δ vs SSR.41BB.ζ (U373; IL2): p<0.001; SSR.Δ vs SSR.CD28.OX40.ζ (U373; IL2): p<0.01. (B) 4-hour cytotoxicity assay at an E:T ratio of 10:1 (n=4).

FIG. 38A-C. Treatment of glioma xenograft with T cells expressing 47-CARs results in tumor regression and improved overall survival. U373 glioma bearing mice were treated on day 7 with SSR.CD28.ζ (n=9), SSR.41BB.ζ (n=9) or SSR.OX40.CD28.ζ (n=9) T cells. SSR.Δ CAR T cells (n=7) served as controls. (A) Representative images for each group and (B) quantitative bioluminescence (radiance=photons/sec/cm2/sr) imaging data for all mice are shown (dotted lines: individual mice; solid lines: median). (C) Kaplan-Meier survival analysis (SSR.Δ vs SSR.CD28.ζ: p=0.0002; SSR.Δ vs SSR.41BB.ζ: p=0.0039; SSR.Δ vs SSR.OX40.CD28.ζ: p=0.0092; SSR.CD28.ζ vs SSR.41BB.ζ: p=0.4723; SSR.CD28.ζ vs SSR.OX40.CD28.ζ: p=0.3582; SSR.41BB.ζ vs SSR.OX40.CD28.ζ: p=0.8374).

FIG. 39A-B. Analysis of U373 cells isolated from recurrent tumors. U373 cells were isolated from recurrent tumors of mice that were treated with 47-CAR T cells. After short-term culture (2 to 7 days), FACS analysis and cytotoxicity assays were performed. (A) FACS analysis for IL13Rα2. (B) 47-CAR T cells killed U373 tumor cells isolated from recurrent tumors in contrast to Raji cells in a standard four-hour cytotoxicity assay for Cr release from labeled cells. Open curve: isotype antibody control; Filled curve: specific antibody.

FIG. 40A-C. Limited persistence of 47-CAR T cell in vivo. 47.SSR.CD28.ζ-CAR T cells were transduced to express eGFP.ffLuc. (A) FACS analysis confirmed the expression of the CAR and eGFP.ffLuc transgenes. (B, C) 1×105 unmodified U373 cells were injected intracranially into mice. On day 7, mice received 2×106 47.SSR.CD28.ζ.eGFP.ffLuc CAR T cells intracranially using the same tumor coordinates. Bioluminescence imaging was used to monitor T cell persistence.

FIG. 41A-C. Generation and characterization of LSR-CD28.41BB.ζ CAR T cells. (A) Scheme of LSR.CD28.41BB.ζ CAR construct. (B) CAR expression was confirmed using FACS analysis. Representative plot. Open curve: secondary antibody; Filled curve: IL13Rα2Fc+secondary antibody. (C) LSR.CD28.41BB.ζ CAR T cells were co-cultured with U373 cells at a 1:2 E:T ratio. NT T cells served as controls. After 24 hours, IFNγ or IL2 was measured by ELISA (n=3).

FIG. 42A-B. Generation of SSR.α.CD28.41BB.ζ CAR T cells. (A) Scheme of SSR.α.CD28.41BB.ζ CAR construct. (B) CAR expression was tested using FACS analysis (representative plot shown).

FIG. 43. FACS analysis of PD-L1 expression on U373 cell surface with and without IFNγ stimulation. U373 cells were cultured with or without IFNγ (100 units/ml). After 24 hours, U373 cells were analyzed for PD-L1 expression using a CD271 PE antibody (BD Biosciences).

FIG. 44A-B. Transgenic expression of IL15 in SSR.CD28.ζ T cells results in enhanced antigen-dependent IL15 secretion. T cells were stimulated with (A) recombinant proteins or (B) cell lines.

FIG. 45A-B. Transgenic expression of IL15 results in (A) enhanced in vivo persistence of SSR.CD28.ζ T cells resulting in improved (B) progression-free survival (PFS).

DETAILED DESCRIPTION

The disclosure provides binding agents, or partners, that specifically recognize interleukin 13 receptor α2 (IL13Rα2) for use in diagnosing, preventing, treating or ameliorating a symptom of any of a wide range of cancers characterized by cells presenting IL13Rα2. More particularly, the disclosure provides (i) the sequences of the six complementarity determining regions of a monoclonal antibody (m47) that specifically targets human tumor-associated antigen, i.e., interleukin 13 receptor α2 (IL13Rα2), and (ii) data demonstrating the functionality of the protein encoded by the heavy and light chain cDNAs in the format of an scFv antibody or conjugate (e.g., fusion) to other functional moieties. The six complementarity determining regions of the m47 monoclonal antibody confer binding specificity for IL13Rα2, consistent with the understanding in the immunological arts. In some embodiments, the scFv comprises the complete heavy and light chain variable regions of antibody m47, or the complete heavy and light chains of antibody m47. In some embodiments, the heavy and light chain fragments comprise, e.g., the m47 CDRs, or the m47 variable regions, and these domains can be arranged in different formats, such as a single-chain variable fragment of an antibody, i.e., a scFv, a diabody, a bi-specific antibody fragment, a tri-specific antibody fragment, a fusion protein with any of a wide variety of therapeutic proteins and/or other moieties, a humanized antibody fragment, a Fab fragment, a Fab′ fragment, a F(ab)2′ fragment and any other functional format for a bi-functional peptide providing a targeting function and an effector function. Moreover, the single-chain antibody or other arrangements of the protein encoded by the heavy and light chains could be expressed and conjugated to therapeutic carriers (e.g., viruses, cells, nanomaterials) for specific delivery of a therapeutic to an IL13Rα2-expressing tumor. The materials according to the disclosure are also useful in imaging tumor burden.

The technology addresses the most serious obstacle to progress in immunotherapy, i.e., the virtual absence of defined, tumor-specific antigens that can be predictably found on at least a larger subgroup of human cancers and that can serve as effective targets for cancer eradication. Finding such antigens would move the field beyond the methods for treating CD19/CD20-expressing B cell malignancies.

The terms used throughout this disclosure are given their ordinary and accustomed meanings in the art, unless a different meaning is made clear from the text when considered in the context of the disclosure as a whole.

The disclosure describes the development and characterization of a monoclonal antibody (mAb) fragment specific to IL13Rα2 for the therapeutic purpose of targeting IL13Rα2-expressing tumors. The high affinity IL13Rα2 is selectively expressed at a high frequency by glioblastoma multiforme (GBM) as well as several other tumor types. One approach for targeting this tumor-specific receptor utilizes the cognate ligand, IL-13, conjugated to cytotoxic molecules. This approach, however, lacks specificity because the lower affinity receptor for IL-13, IL13Rα1, is widely expressed by normal tissues. A monoclonal antibody (mAb) specific to IL13Rα2 was expected to overcome the lack of specificity afflicting methodologies that recognized both IL13 receptors, i.e., IL13Rα1 as well as IL13Rα2. Such a mAb would be therapeutically useful in targeting and treating IL13Rα2-expressing cancers, including tumors.

As disclosed herein, hybridoma cell lines were generated and compared for binding affinities to recombinant human IL13Rα2 (rhIL13Rα2). Clone 47 demonstrated binding to the native conformation of IL13Rα2 and was therefore chosen for further studies. Clone 47 bound specifically and with high affinity (KD=1.39×10−9 M) to rhIL13Rα2 but not to rhIL13Rα1 or murine IL13Rα2. Furthermore, clone 47 specifically recognized wild-type IL13Rα2 expressed on the surface of CHO and HEK cells as well as several glioma cell lines. Competitive binding assays revealed that clone 47 also significantly inhibited the interaction between human soluble IL-13 and IL13Rα2 receptor. Moreover, N-linked glycosylation of IL13Rα2 contributes in part to the interaction of the antibody to IL13Rα2. In vivo, the IL13Rα2 mAb improved the survival of nude mice intracranially implanted with a human U251 glioma xenograft.

The IL13Rα2-specific, scFv-based CAR, 47-CAR, constructed as disclosed herein, provided the material used in exploring the influence of long and short spacer regions, as well as endodomains, on its function. While 47-CAR.SSR.CD28.ζ (i.e., the 47-CAR binding region provided as an scFv joined to a short spacer region as defined herein, in turn joined to an unmodified or chimeric endodomain or T cell cytoplasmic domain) and 47-CAR.LSR.CD28.ζ (similar construct substituting a long spacer region (LSR)) recognized target cells as judged by IFNγ production, only 47-CAR.SSR.CD28.ζ induced IL2 production, indicating better T-cell activation. An additional LSR 47-CAR containing a CD28.41BB.ζ endodomain (FIG. 41) was shown to lack the ability to induce IL2 expression. These observations are consistent with knowledge that scFvs that bind to an epitope in close proximity to the cancer cell membrane, requiring long spacer regions for optimal CAR function, in contrast to scFvs that bind to epitopes distal to the cell membrane. The data disclosed herein indicates that the IL13Rα2 epitope recognized by 47-CARs is located distal to the cell membrane.

In greater particularity, four SSR 47-CARs were constructed, each with a different endodomain, i.e., CD28.ζ, 41BB.ζ CD28.OX40.ζ, and CD28.41BB.ζ. While all four CARs were expressed, as judged by Western blot analysis, no significant cell-surface expression was observed for 47-CAR.SSR.CD28.41BB.ζ. We explored if changing the transmembrane domain from CD28 to CD8α in 47-CAR.SSR.CD28.41BB.ζ would result in better cell-surface expression, but no increase in expression was observed. Because 47-CARs.LSR.CD28.41BB.ζ are expressed on the cell surface (FIG. 42), the result indicates that the interplay between spacer region and endodomain influences CAR cell-surface expression.

47-CAR.SSR.CD28.ζ, 47-CAR.SSR.41BB.ζ, and 47-CAR.SSR.CD28.OX40.ζ T cells had potent antitumor effect in vivo, resulting in a significant survival advantage. While mice treated with 47-CAR.SSR.CD28.ζ T cells had the longest median survival in comparison to 47-CAR.SSR.41BB.ζ or 47-CAR.SSR.CD28.OX40.ζ T-cell treated mice, this difference did not reach significance. The experimental results also showed that addition of a second costimulatory endodomain did not improve antitumor activity in vivo. Limited T-cell persistence in vivo was identified as the principal limitation on therapy. This limitation may be overcome by the transgenic expression of cytokines' or by blocking inhibitory molecules that are secreted or present on the surface of gliomal cells. For example, gliomas such as U373 express PD-L1, which is upregulated in the presence of IFNγ (FIG. 43), and could be targeted in future studies.

The experimental results disclosed herein establish that T cells redirected to IL13Rα2 with 47-CARs have potent anti-tumor activity against glioma cells in vitro, and induce the regression of established GBM xenografts in vivo. 47-CARs are expected to be of value in the treatment of not only IL13Rα2-positive GBMs but also other malignancies in which IL13Rα2 is expressed.

The experimental results disclosed herein establish that T cells redirected to IL13Rα2 with 47-CARs and that also express IL15 have enhanced anti-tumor activity in the GBM xenografts in vivo.

The disclosure is based, at least in part, on the discovery that IL13Rα2 is found preferentially on cancer cells such as tumor cells. This receptor functions as a cancer-, or tumor-, specific antigen that has been used to elicit the high-affinity monoclonal antibody m47, along with antigen binding fragments of that antibody. The VL and VH variable regions of the m47 antibody have been engineered into a single chain (sc) variable fragment (scFv) to generate conjugates, such as chimeric antigen receptors (i.e., CARs), for introduction into T cells for adoptive transfer. Thus, CAR-transduced T cells are expected to target a tumor-specific IL13Rα2 epitope, leading to eradication of cancer cells presenting the receptor. It is believed that CAR-transduced T cells recognizing IL13Rα2 will destroy large solid tumors. CAR-transduced T cells, however, target cancer cells only directly and antigen-negative cancer cells may escape. It is expected that CAR-transduced T cells also will be effective in eliminating antigen-negative cancer cells via the bystander effect.

Disclosed herein are experiments establishing the development of IL13Rα2-specific CARs with a scFv47-based antigen-binding domain (47-CARs). The data show that 47-CARs perform better with a short spacer region, which provides for optimal functionality, and that 47-CAR T cells are able to recognize and kill only IL13Rα2-positive and not IL13Rα1-positive target cells in vitro. In addition, 47-CAR T cells induce tumor regression in an orthotopic xenograft mouse model of GBM, which was associated with a significant survival advantage.

The protein conjugates according to the disclosure are specific for IL13Rα2, which is associated with cancers, e.g., tumors. In addition, the disclosure provides a polynucleotide encoding one of these cancer-specific binding partners, including polynucleotides comprising codon-optimized coding regions for binding partners specific for an epitope of IL13Rα2. The polynucleotides of the disclosure encode conjugates, or bi-functional polypeptides, useful in diagnosing, preventing, treating, or ameliorating a symptom of cancer, such as any of a variety of human cancers, including those forming solid tumors. Also contemplated are vectors comprising a polynucleotide as disclosed herein, a host cell comprising such a polynucleotide and/or a vector as described above, and methods of treating, preventing or ameliorating a symptom of, a cancer disease, e.g., a solid tumor, a primary cancer site or a metastasized cancer.

The various forms of conjugates known in the art are contemplated by the disclosure. These conjugates provide exquisitely cancer—as well as protein-specific antibody receptors that can be incorporated into a variety of backbones providing effector function, such as bispecific T cell Engagers (BiTEs) or chimeric antigen receptors (CARs), as noted below. Exemplary conjugates of the disclosure include CARs, fusion proteins, including fusions comprising single-chain variable (antibody) fragment (scFv) multimers or scFv fusions to coding regions encoding products useful in treating cancer, e.g., IL15, IL15Rα, or IL15/IL15Rα agent, diabodies, tribodies, tetrabodies, and bispecific bivalent scFvs, including bispecific tandem bivalent scFvs, also known as bispecific T cell engagers, or BiTEs. Any of these conjugate forms, moreover, may exhibit any of various relative structures, as it is known in the art that different domain orders (e.g., H2N—VH-linker-VL-CO2H and H2N-VL-linker-VH—CO2H) are compatible with specific binding. Higher order forms of the conjugates described herein are also contemplated, such as peptibodies comprising at least one form of the conjugates disclosed herein. The conjugates of the disclosure specifically bind to a cancer-specific epitope (e.g., an IL13Rα2) and the polynucleotides encoding them may be codon-optimized, e.g., for maximal translation, for expression in the targeted cells (e.g., human or mouse cells). Codon optimization in the context of expressing the conjugates of the disclosure, such as CARs, is important to ensuring that production of the protein is both efficient and robust enough to be useful as a source of therapeutic.

The disclosure also contemplates conjugates in which a targeting moiety (an anti-IL13Rα2 antibody or fragment thereof) is linked to a peptide providing a second function, e.g., an effector function, such as a T cell signaling domain involved in T cell activation, a peptide that affects or modulates an immunological response to cancer cells, or an enzymatic component of a labeling system that results in a CAR encoded by a polynucleotide according to the disclosure, if the coding region for the conjugate is codon-optimized for expression in a target cell. Exemplary conjugates include an anti-IL13Rα2 scFv linked to a hinge, a transmembrane domain, and an effector compound or domain, e.g., CD28, CD3ζ, CD134 (OX40), CD137 (41BB), ICOS, CD40, CD27, or Myd88, thereby yielding a CAR.

The polynucleotide aspect of the disclosure comprises embodiments in which an unexpected variation on codon optimization in slower-growing higher eukaryotes such as vertebrates, e.g., humans, is provided that is focused on translation optimization (maximizing high-fidelity translation rates) rather than the typical codon optimization used in such organisms, which is designed to accommodate mutational bias and thereby minimize mutation. Also disclosed are the methods of diagnosing, preventing, treating or ameliorating a symptom of a cancer. Schematically described, the polynucleotides comprise a codon-optimized coding region for an antigen receptor specifically recognizing an IL13Rα2 epitope linked to any one of the following: a coding region for a T cell signaling domain involved in T cell activation, a gene product that affects or modulates an immunological response to cancer cells such as an IL15/IL15Rα fusion, or a labeling component such as an enzymatic component of a labeling system. The linked coding regions result in polynucleotides encoding conjugates according to the disclosure, such as BiTEs or chimeric antigen receptors (CARs).

In methods of diagnosing, preventing, treating or ameliorating a symptom of a cancer, the compositions of the disclosure are typically administered in the form of a conjugate-transduced cell, such as a T cell, an NK cell, or a lymphocyte including, but not limited to, NKT cells, γδ T cells, mucosa associated invariant T cells or MAIT cells, or innate lymphoid cells, although administration of a vector comprising a polynucleotide of the disclosure or administration of a polynucleotide of the disclosure are also contemplated, depending on the functionalities of the conjugate. Combining a polynucleotide, vector or host cell of the disclosure with a physiologically suitable buffer, adjuvant or diluent yields a pharmaceutical composition according to the disclosure, and these pharmaceutical compositions are suitable for administration to diagnose, prevent, treat, or ameliorate a symptom of, a cancer.

In the course of experimental work described herein, hybridoma cell lines were generated and compared for binding affinities to recombinant human IL13Rα2 (rhIL13Rα2). Clone 47 demonstrated binding to the native conformation of IL13Rα2 and was therefore characterized further. Clone 47 bound specifically and with high affinity (KD 1.39×10−9 M) to rhIL13Rα2 but not to rhIL13Rα1 or murine IL13Rα2. Furthermore, clone 47 specifically recognized wild-type IL13Rα2 expressed on the surface of CHO and HEK cells as well as several glioma cell lines. Competitive binding assays revealed that clone 47 also significantly inhibited the interaction between human soluble IL-13 and IL13Rα2 receptor. Moreover, N-linked glycosylation of IL13Rα2 was found to contribute, in part, to the interaction of the antibody with IL13Rα2. In vivo, the IL13Rα2 monoclonal antibody improved the survival of nude mice intracranially implanted with a human U251 glioma xenograft. Collectively, these data establish the efficacy of the immunomodulatory treatment of cancer disclosed herein.

Overexpression of IL13Rα2 in glioblastoma multiforme (GBM) but not in normal brain tissue uniquely positions this receptor as a candidate for targeting tumor cells. GBM is a highly infiltrative tumor, often making complete surgical removal impossible. Moreover, GBM is highly resistant to radiation and chemotherapy (16), warranting further development of novel and targeted therapies for the treatment of patients.

A phage display library approach has been used to select small antibody fragments specific to human IL13Rα2, followed by their evaluation in vitro and in vivo (23). Despite the high specificity of interaction with IL13Rα2, conjugation with toxins has failed to increase cytotoxicity in IL13Rα2-expressing glioma and renal cell carcinoma cell lines when compared with the effects of IL-13PE38. The low affinity of generated antibody fragments is the most reasonable explanation for the lack of success. Antibody fragments derived from phage display libraries are known to be lower in affinity and avidity than antibodies generated by conventional hybridoma technology (24). Modifications of those small antibody fragments are often required to enhance their affinity and avidity to targeted proteins. In recent years, monoclonal antibodies have shown increasing success as targeted anticancer and diagnostic agents (25, 26), and a further search for high affinity reagents with restricted specificity to tumor-associated antigens is needed. The experiments disclosed herein were designed to discover, develop, and characterize a high affinity antibody that specifically recognizes IL13Rα2 expressed on the surface of cancer cells. Consistent with that design, disclosed herein are experiments establishing the generation of an antibody possessing the properties critical for immunotherapeutic targeting of IL13Rα2-expressing tumors in vivo, and potentially suitable for various other applications.

Monoclonal antibodies appear to be valuable research and diagnostic tools as well as therapeutic agents. Monoclonal antibodies specific for tumor-associated antigens have significant advantages over systemic chemotherapies due to the ability to specifically target cancer cells while avoiding interaction with untransformed tissue. Therefore, the search for novel “magic bullets” continues to grow, confirmed by a global market for therapeutic antibodies worth $48 billion as of 2010. Therapeutic antibodies are products of traditional hybridoma technology or screening of libraries for antibody fragments and their subsequent engineering into humanized fragments or full size molecules. Prior to this study, the hybridoma cell line secreting a high affinity antibody to the tumor-specific antigen IL13Rα2 was unavailable to the scientific community. Here, we describe the generation and characterization of a high affinity antibody to the tumor-specific antigen IL13Rα2 and discuss its potential use in different applications.

The specificity of interaction of newly discovered antibodies to human IL13Rα2 was analyzed by ELISA using the rhIL13Rα2hFc fusion protein, recombinant human IL13Rα2 expressed on the surface of CHO and HEK cells, and several glioma cell lines expressing IL13Rα2 at various levels by flow cytometry. The antibody identified herein, and agent using the binding domain thereof, demonstrated a specificity of interaction to human IL13Rα2 and did not cross-react with human IL13Rα1 or mouse IL13Rα2. Moreover, the specificity of binding to IL13Rα2 was confirmed in competitive binding assays using rhIL13Rα2hFc fusion protein by ELISA or by flow cytometry for detection of IL13Rα2 expressed on the surface of HEK cells. In these assays, IL13Rα2 (clone 47) mAb competed with recombinant human IL-13 for its epitope and was able to block about 80% of the binding between IL-13 and IL13Rα2. Conversely, human recombinant IL-13 was able to block about 50% of antibody binding to IL13Rα2. Similarly, a significant decrease in the binding of IL13Rα2 (clone 47) mAb to N10 glioma cells was observed when rhIL13R2hFc chimera and rhIL-13 were used as competitors. The binding of rhIL-13 to N10 cells was also abolished by IL13Rα2 (clone 47) mAb. These data indicate that the two molecules have significant overlap in their recognition sites for IL13Rα2.

IL-13 is a small 10-kDa molecule (31), whereas an antibody is about 15 times greater in molecular mass. The ability of rhIL-13 to compete with an antibody for a binding site suggests that the inhibitory property of the antibody is likely due to the specific interaction with amino acid residues contributing to the binding of IL-13 to the cognate receptor rather than to steric hindrance, which can also prevent the interaction of IL-13 with its receptor. Previously, Tyr207, Asp271, Tyr315, and Asp318 were identified as critical residues of IL13Rα2 necessary for interaction with IL-13 (28). In the assays disclosed herein, the binding of IL-13 to a mutant IL13Rα2 carrying a combination of all 4 amino acid mutations to alanine was significantly abolished when compared with the wild-type receptor. Binding of the IL13Rα2 mAb to either the individual or the 4-amino acid mutant form of IL13Rα2, however, was not significantly affected. These findings indicate that Tyr207, Asp271, Tyr315, and Asp318 residues are not critical for the recognition of IL13Rα2 by the IL13Rα2 mAb. The human IL13Rα2 and murine IL13Rα2 are structurally conserved and share 59% amino acid identity (32). Moreover, Tyr207, Asp271, Tyr315, and Asp318 residues are conserved in human and murine IL13Rα2. Absence of binding of the IL13Rα2 mAb to murine IL3Rα2hFc fusion further supports the expectation that these amino acid residues contribute to the binding of IL-13 to IL13Rα2 and are not critical for the interaction of this antibody with the receptor.

To further characterize the interaction of IL13Rα2 with the antibody and antibody agent disclosed herein, the affinity of the IL13Rα2 mAb was measured and compared with the binding properties of two commercially available antibodies using the surface plasmon resonance method. The affinity of the IL13Rα2 mAb was determined to be equal to 1.39×10−9 M, greatly exceeding the affinity of comparable commercially available antibodies by up to 75-fold. In agreement with the affinity studies, the IL13Rα2 mAb (clone 47) demonstrated superiority to two commercial antibodies in binding to the IL13Rα2 expressed on the surface of various glioma cells and in ELISA. Although many properties of antibodies, including the affinity and avidity, in vivo stability, rate of clearance and internalization, tumor penetration, and retention, should be considered prior to specific usage, it has been reported that higher affinity antibodies are better for immunotherapeutic tumor-targeting applications (33). The single chain antibody fragment (scFv) MR1-1 against epidermal growth factor receptor variant III demonstrates about 15-fold higher affinity than the parental scFvMR1 and also showed on average a 244% higher tumor uptake than that for the scFvMR1 (34). It is likely that the high affinity properties of the IL13Rα2 mAb and agent thereof that are disclosed herein will be advantageous for applications utilizing antibodies or associated derivatives for targeting tumor cells expressing IL13Rα2.

The N-linked glycosylation of IL13Rα2 has been identified as a necessary requirement for efficient binding to IL-13 (30). Taking into consideration that the IL13Rα2 mAb disclosed herein inhibits about 80% of IL-13 binding to the cognate receptor, IL13Rα2, it is reasonable to expect that the binding of this antibody, or an agent containing its binding domain, with the deglycosylated form of IL13Rα2 could also be affected. The IL13Rα2 molecule has four potential sites of N-linked glycosylation. The binding of the antibody to rhIL13Rα2 or to IL13Rα2 expressed on the surface of HEK or U251 cells treated with Pngase F was decreased by 35 and 30%, respectively, when compared with non-treated control. A partial change in binding activity for the clone 47 when compared with clones 83807 and B-D13 suggests that removal of carbohydrate adducts from IL13Rα2 with Pngase F causes conformational changes of the receptor, indirectly affecting the binding of both IL-13 (30) and the IL13Rα2 mAb to IL13Rα2. This also supports the expectation that the antibody binds directly to the IL13Rα2 amino acid backbone rather than interacting with carbohydrate moieties added post-translationally. Supporting this expectation, several studies have previously demonstrated that the conformational profile and structural rigidity of proteins depends on N-linked glycosylation (22, 35-38).

To investigate the therapeutic properties of the IL13Rα2 mAb and its agent, an in vivo study was performed whereby glioma cells and the IL13Rα2 (clone 47) mAb were intracranially co-injected into brain, or antibody was injected into established tumor-bearing mice. Interestingly, the IL13Rα2 mAb was able to delay tumor progression and improve survival of animals with intracranial U251 glioma xenografts most significantly in the co-injected model, demonstrating a trend in the improvement of median survival in animals with established glioma. Although the underlying mechanism for this antitumor effect remains unclear, the result establishes the therapeutic applicability of this antibody, or its agent (containing the IL13Rα2 binding domain in the form of the six CDR regions, or in the form of the two variable domains of the clone 47 anti-IL13Rα2), alone or in combination with a pharmaceutical carrier, thereby providing therapies for the treatment of IL13Rα2-expressing glial and other lineage tumors. Several antibodies have been shown to mediate a cytotoxic effect in tumors through Fc-mediated activation of complement (39). Antibody-dependent cell-mediated cytotoxicity-induced activation of effector cells can also contribute to the cytotoxic effect of antibodies against targeted cells (40, 41). Anti-IL13Rα2 activity derived from the sera of animals challenged with D5 melanoma cells expressing human IL13Rα2 demonstrates the ability to inhibit cellular growth in vitro (4).

Cancers amenable to the described treatments include cancers in which IL13Rα2 has been found to be expressed, including glioblastoma; medulloblastoma; Kaposi sarcoma; and head and neck, ovarian, pancreatic, kidney, and colorectal cancers (2, 43-47). Although the role of IL13Rα2 in some cancers is not yet defined, recent reports have demonstrated that IL13Rα2 contributes to the invasive phenotype of ovarian, pancreatic, and colorectal cancers (5, 13). Moreover, Minn et al. (42) have suggested a relationship between IL13Rα2 expression and breast cancer metastasis to the lung. Additionally, Fichtner-Feigl et al. (11) demonstrated that the interaction of IL-13 with IL13Rα2 upregulates TGF-β1, mediating fibrosis in a bleomycin-induced model of lung fibrosis. In light of this finding, it is expected that the anti-IL13Rα2 antibody (clone 47) and binding agents thereof, will be able to attenuate TGF-β1-induced pulmonary fibrosis.

As disclosed herein, the described experiments led to the generation of an anti-IL13Rα2 antibody and binding agents thereof, all of which are specific to human IL13Rα2. The antibody and its agent possess a high affinity for IL13Rα2 and compete with IL-13 for the binding site on IL13Rα2. The antibody recognizes antigen expressed on the cell surface of glioma cells as well as other IL13Rα2-expressing cells, establishing the suitability for targeting IL13Rα2-expressing tumor cells in vivo. The anti-IL13Rα2 antibody and binding agents thereof are also expected to be efficacious and cost effective in diagnostic imaging, delivery of antibody radionuclide conjugates, bioassays for the detection of IL13Rα2, and as a carrier for therapeutic agents in various types of IL13Rα2-overexpressing tumors.

In methods of diagnosing, preventing, treating or ameliorating a symptom of a cancer, the compositions of the disclosure are typically administered in the form of conjugate-transduced T cells, although administration of a vector comprising a polynucleotide of the disclosure or administration of a polynucleotide of the disclosure are also contemplated, depending on the functionalities of the conjugate. Combining a polynucleotide, vector or host cell of the disclosure with a physiologically suitable buffer, adjuvant or diluent yields a pharmaceutical composition according to the disclosure, and these pharmaceutical compositions are suitable for administration to diagnose, prevent, treat, or ameliorate a symptom of, a cancer.

A conjugate according to the disclosure, such as a fusion protein composed of an scFv-receptor for an IL13Rα2 epitope fused to IL15/IL15Rα, is also contemplated. It is expected that the fusion protein will eliminate clinical size tumors or only incipient and microdisseminated cancer cells. The disclosure further contemplates the simultaneous targeting of two independent IL13Rα2 epitopes on a human cancer, which may be essential for preventing escape from treatment, such as CAR treatment.

Simultaneous targeting of different epitopes of IL13Rα2 by CARs should reduce the chance of escape of a cancer subpopulation, which provides a strong reason for identifying additional IL13Rα2 antibody products and/or epitopes.

The disclosure provides materials and methods that are adaptable and can serve as the basis for a platform technology with considerable growth potential. The cancer-specific nature of IL13Rα2 is expected to provide targets for cancer diagnostics, prophylactics and therapeutics that offer major advantages over previously and presently used targets.

Consistent with the spirit of the foregoing, the following provides a description of the materials and methods provided herein.

Disclosed herein are IL13Rα2 binding agents comprising each of the amino acid sequences of NYLMN (SEQ ID NO: 1); RIDPYDGDIDYNQNFKD (SEQ ID NO: 2); GYGTAYGVDY (SEQ ID NO: 3); RASESVDNYGISFMN (SEQ ID NO: 4); AASRQGSG (SEQ ID NO: 5); and QQSKEVPWT (SEQ ID NO: 6). In exemplary aspects, the binding agent comprises each of the foregoing six amino acid sequences in addition to further sequences which provide a framework to support a three-dimensional conformation that binds to IL13Rα2. In exemplary aspects, the IL13Rα2 binding agent comprises one or both of the amino acid sequences of SEQ ID NO: 7 and/or SEQ ID NO: 8. In exemplary aspects, the IL13Rα2 binding agent comprises the amino acid sequence of SEQ ID NO: 7. In exemplary aspects, the IL13Rα2 binding agent comprises the amino acid sequence of SEQ ID NO: 8. In exemplary aspects, the IL13Rα2 binding agent comprises both the amino acid sequences of SEQ ID NO: 7 and SEQ ID NO: 8. In exemplary aspects wherein both the amino acid sequences of SEQ ID NO: 7 and SEQ ID NO: 8 are present in the binding agent, the amino acid sequence of SEQ ID NO: 7 is fused to the amino acid sequence of SEQ ID NO: 8 through a linker. Suitable linkers are known in the art. In exemplary aspects, the linker comprises a short amino acid sequence of about 5 to about 25 amino acids, e.g., about 10 to about 20 amino acids. In exemplary aspects, the linker comprises the amino acid sequence of EEGEFSEAR (SEQ ID NO 10). In exemplary aspects, the linker comprises the amino acid sequence of AKTTPPKLEEGEFSEARV (SEQ ID NO: 80). In exemplary aspects, IL13Rα2 binding agent comprises the amino acid sequence of SEQ ID NO: 13.

In exemplary embodiments, the binding agent provided herein further comprises additional amino acid sequences. In exemplary aspects, the binding agent further comprises a constant region of a heavy chain and/or a constant region of a light chain. Sequences for heavy and light chain constant regions are publically available. For example, the National Center of Biotechnology Information (NCBI) nucleotide database provides a sequence of the constant region of the IgG1 kappa light chain. See GenBank Accession No. DQ381549.1, incorporated herein by reference. In exemplary aspects, the binding agent comprises an amino acid sequence of SEQ ID NO: 28. In exemplary aspects, the binding agent comprises a modified amino acid sequence of SEQ ID NO: 28. In exemplary aspects, the binding agent comprises an amino acid sequence which is at least 90%, at least 93%, at least 95%, or at least 98% identical to SEQ ID NO: 28. Also, for example, the NCBI nucleotide database provides a sequence of the constant region of the Mus musculus IgG1. See GenBank Accession No. DQ381544.1. In exemplary aspects, the binding agent comprises an amino acid sequence of SEQ ID NO: 29. In exemplary aspects, the binding agent comprises a modified amino acid sequence of SEQ ID NO: 29. In exemplary aspects, the binding agent comprises an amino acid sequence which is at least 90%, at least 93%, at least 95%, or at least 98% identical to SEQ ID NO: 29.

In exemplary aspects, the IL13Rα2 binding agent is an antibody, or an antigen-binding fragment thereof. In exemplary aspects, the antibody comprises each of the amino acid sequences of SEQ ID NOs: 1-6. In exemplary aspects, the antibody comprises the amino acid sequence of SEQ ID NO: 7 and/or SEQ ID NO: 8. In exemplary aspects, the antibody comprises the amino acid sequences of SEQ ID NO: 7 and SEQ ID NO: 8. In exemplary aspects, the antibody comprises the amino acid sequences of SEQ ID NO: 7 and SEQ ID NO: 8 and the amino acid sequence of SEQ ID NO: 7 is fused to the amino acid sequence of SEQ ID NO: 8 through a linker. In exemplary aspects, the linker comprises a short amino acid sequence of about 5 to about 25 amino acids, e.g., about 10 to about 20 amino acids. In exemplary aspects, the linker comprises the amino acid sequence of EEGEFSEAR (SEQ ID NO 10). In exemplary aspects, the linker comprises the amino acid sequence of AKTTPPKLEEGEFSEARV (SEQ ID NO: 80). In exemplary aspects, the antibody comprises the amino acid sequence of SEQ ID NO: 13.

In exemplary aspects, the antibody can be any type of immunoglobulin that is known in the art. For instance, the antibody can be of any isotype, e.g., IgA, IgD, IgE, IgG, or IgM. The antibody can be monoclonal or polyclonal. The antibody can be a naturally-occurring antibody, i.e., an antibody isolated and/or purified from a mammal, e.g., mouse, rabbit, goat, horse, chicken, hamster, human, and the like. In this regard, the antibody may be considered to be a mammalian antibody, e.g., a mouse antibody, rabbit antibody, goat antibody, horse antibody, chicken antibody, hamster antibody, human antibody, and the like. The term “isolated” as used herein means having been removed from its natural environment. The term “purified,” as used herein relates to the isolation of a molecule or compound in a form that is substantially free of contaminants normally associated with the molecule or compound in a native or natural environment and means having been increased in purity as a result of being separated from other components of the original composition. It is recognized that “purity” is a relative term, and not to be necessarily construed as absolute purity or absolute enrichment or absolute selection. In some aspects, the purity is at least or about 50%, is at least or about 60%, at least or about 70%, at least or about 80%, or at least or about 90% (e.g., at least or about 91%, at least or about 92%, at least or about 93%, at least or about 94%, at least or about 95%, at least or about 96%, at least or about 97%, at least or about 98%, at least or about 99% or is approximately 100%.

In exemplary aspects, the antibody comprises a constant region of an IgG. In exemplary aspects, the antibody comprises a constant region of an IgG1. In exemplary aspects, the antibody comprises a constant region of an IgG kappa light chain. For instance, the antibody may comprise the amino acid sequence of SEQ ID NO: 28. In exemplary aspects, the antibody comprises an amino acid sequence that is highly similar to SEQ ID NO: 28. For instance, the antibody may comprise an amino acid sequence having at least 85% sequence identity to SEQ ID NO: 28, or an amino acid sequence having at least 90% sequence identity to SEQ ID NO: 28, or an amino acid sequence having at least 93% sequence identity to SEQ ID NO: 28, or an amino acid sequence having at least 95% sequence identity to SEQ ID NO: 28, or an amino acid sequence having at least 98% sequence identity to SEQ ID NO: 28.

In exemplary aspects, the antibody comprises a constant region of a Mus musculus IgG1. For instance, the antibody may comprise the amino acid sequence of SEQ ID NO: 30. In exemplary aspects, the antibody comprises an amino acid sequence which is highly similar to SEQ ID NO: 30. For instance, the antibody may comprise an amino acid sequence having at least 85% sequence identity to SEQ ID NO: 30, or an amino acid sequence having at least 90% sequence identity to SEQ ID NO: 30, or an amino acid sequence having at least 93% sequence identity to SEQ ID NO: 30, or an amino acid sequence having at least 95% sequence identity to SEQ ID NO: 30, or an amino acid sequence having at least 98% sequence identity to SEQ ID NO: 30.

The anti-IL13Rα2 antibodies and fragments thereof of the disclosure can have any level of affinity or avidity for IL13Rα2. The dissociation constant (KD) may be any of those exemplary dissociation constants described herein with regard to binding units. Binding constants, including dissociation constants, are determined by methods known in the art, including, for example, methods that utilize the principles of surface plasmon resonance, e.g., methods utilizing a Biacore™ system. In accordance with the foregoing, in some embodiments, the antibody is in monomeric form, while in other embodiments, the antibody is in polymeric form. In certain embodiments in which the antibody comprises two or more distinct antigen binding regions or fragments, the antibody is considered bispecific, trispecific, or multi-specific, or bivalent, trivalent, or multivalent, depending on the number of distinct epitopes that are recognized and bound by the binding agent.

Because the binding agent of the disclosures can compete with IL13 for binding to IL13Rα2, the antibody in exemplary aspects is considered to be a blocking antibody or neutralizing antibody. In some aspects, the KD of the binding agent is about the same as the KD of the native ligand, IL13, for IL13Rα2. In some aspects, the KD of the binding agent is lower (e.g., at least 0.5-fold lower, at least 1-fold lower, at least 2-fold lower, at least 5-fold lower, at least 10-fold lower, at least 25-fold lower, at least 50-fold lower, at least 75-fold lower, at least 100-fold lower) than the KD of IL13 for IL13Rα2. In exemplary aspects, the KD is between about 0.0001 nM and about 100 nM. In some embodiments, the KD is at least or about 0.0001 nM, at least or about 0.001 nM, at least or about 0.01 nM, at least or about 0.1 nM, at least or about 1 nM, or at least or about 10 nM. In some embodiments, the KD is no more than or about 100 nM, no more than or about 75 nM, no more than or about 50 nM, or no more than or about 25 nM. In exemplary aspects, the antibody has a KD for human IL13Rα2 that is no greater than about 1.39×10−9 M.

In exemplary aspects, the binding agent, e.g., antibody, or antigen binding fragment thereof, does not bind to human IL13Rα1.

In exemplary embodiments, the antibody is a genetically engineered antibody, e.g., a single chain antibody, a humanized antibody, a chimeric antibody, a CDR-grafted antibody, an antibody that includes portions of CDR sequences specific for IL13Rα2 (e.g., an antibody that includes CDR sequences of SEQ ID NOs: 1-6), a humaneered or humanized antibody, a bispecific antibody, a trispecific antibody, and the like, as defined in greater detail herein. Genetic engineering techniques also provide the ability to make fully human antibodies in a non-human.

In some aspects, the antibody is a chimeric antibody. The term “chimeric antibody” is used herein to refer to an antibody containing constant domains from one species and the variable domains from a second, or more generally, containing stretches of amino acid sequence from at least two species.

In some aspects, the antibody is a humanized antibody. The term “humanized” when used in relation to antibodies is used to refer to antibodies having at least CDR regions from a nonhuman source that are engineered to have a structure and immunological function more similar to true human antibodies than the original source antibodies. For example, humanizing can involve grafting CDR from a non-human antibody, such as a mouse antibody, into a human antibody. Humanizing also can involve select amino acid substitutions to make a non-human sequence look more like a human sequence, as would be known in the art.

Use of the terms “chimeric or humanized” herein is not meant to be mutually exclusive; rather, is meant to encompass chimeric antibodies, humanized antibodies, and chimeric antibodies that have been further humanized. Except where context otherwise indicates, statements about (properties of, uses of, testing, and so on) chimeric antibodies apply to humanized antibodies, and statements about humanized antibodies pertain also to chimeric antibodies. Likewise, except where context dictates, such statements also should be understood to be applicable to antibodies and antigen binding fragments of such antibodies.

In some aspects of the disclosure, the binding agent is an antigen binding fragment of an antibody that specifically binds to an IL13Rα2 in accordance with the disclosure. The antigen binding fragment (also referred to herein as “antigen binding portion”) may be an antigen binding fragment of any of the antibodies described herein. The antigen binding fragment can be any part of an antibody that has at least one antigen binding site, including, but not limited to, Fab, F(ab′)2, dsFv, sFv, scFv, diabodies, triabodies, bis-scFvs, fragments expressed by a Fab expression library, domain antibodies, VhH domains, V-NAR domains, VH domains, VL domains, and the like. Antibody fragments of the invention, however, are not limited to these exemplary types of antibody fragments.

In exemplary aspects, the IL13Rα2 binding agent is an antigen binding fragment. In exemplary aspects, the antigen binding fragment comprises each of the amino acid sequences of SEQ ID NOs: 1-6. In exemplary aspects, the antigen binding fragment comprises the amino acid sequence of SEQ ID NO: 7 and/or SEQ ID NO: 8. In exemplary aspects, the antigen binding fragment comprises the amino acid sequences of SEQ ID NO: 7 and SEQ ID NO: 8. In exemplary aspects, the antigen binding fragment comprises the amino acid sequences of SEQ ID NO: 7 and SEQ ID NO: 8 and the amino acid sequence of SEQ ID NO: 7 is fused to the amino acid sequence of SEQ ID NO: 8 through a linker. In exemplary aspects, the linker comprises a short amino acid sequence of about 5 to about 25 amino acids, e.g., about 10 to about 20 amino acids. In exemplary aspects, the linker comprises the amino acid sequence of EEGEFSEAR (SEQ ID NO 10). In exemplary aspects, the linker comprises the amino acid sequence of AKTTPPKLEEGEFSEARV (SEQ ID NO: 80). In exemplary aspects, the antigen binding fragment provided herein comprises the amino acid sequence of SEQ ID NO: 13.

In exemplary aspects, the antigen binding fragment comprises a leader sequence. Optionally, the leader sequence, in some aspects, is located N-terminal to the heavy chain variable region. In exemplary aspects, the antigen binding fragment comprises an Ig kappa leader sequence. Suitable leader sequences are known in the art, and include, for example, an Ig kappa leader sequence of METDTLLLWVLLLWVPGSTGD (SEQ ID NO: 9).

In exemplary aspects, an antigen binding fragment comprises one more tag sequences. Tag sequences may assist in the production and characterization of the manufactured antigen binding fragment. In exemplary aspects, the antigen binding fragment comprises one or more tag sequences C-terminal to the light chain variable region. Suitable tag sequences are known in the art and include, but are not limited to, Myc tags, His tags, and the like. In exemplary aspects, an antigen binding fragment comprises a Myc tag of GGPEQKLISEEDLN (SEQ ID NO: 11). In exemplary aspects, an antigen binding fragment comprises a His tag sequence of HHHHHH (SEQ ID NO: 12).

In exemplary aspects, the antigen binding fragment of the disclosures comprises, from the N- to the C-terminus, a leader sequence, a heavy chain variable region, a linker sequence, a light chain variable region, a Myc tag (e.g., SEQ ID NO: 11), and a His tag (e.g., SEQ ID NO: 12). In exemplary aspects, the antigen binding fragment of the disclosure comprises the amino acid sequence of SEQ ID NO: 14.

In exemplary aspects, the antigen binding fragment is a domain antibody. A domain antibody comprises a functional binding unit of an antibody, and can correspond to the variable regions of either the heavy (VH) or light (VL) chains of antibodies. A domain antibody can have a molecular weight of approximately 13 kDa, or approximately one-tenth the weight of a full antibody. Domain antibodies may be derived from full antibodies, such as those described herein. The antigen binding fragments in some embodiments are monomeric or polymeric, bispecific or trispecific, and bivalent or trivalent.

Antibody fragments that contain the antigen binding, or idiotype, of the antibody molecule share a common idiotype and are contemplated by the disclosure. Such antibody fragments may be generated by techniques known in the art and include, but are not limited to, the F(ab′)2 fragment which may be produced by pepsin digestion of the antibody molecule; the Fab′ fragments which may be generated by reducing the disulfide bridges of the F(ab′)2 fragment, and the two Fab′ fragments which may be generated by treating the antibody molecule with papain and a reducing agent.

In exemplary aspects, the binding agent provided herein is a single-chain variable region fragment (scFv) antibody fragment. An scFv may consist of a truncated Fab fragment comprising the variable (V) domain of an antibody heavy chain linked to a V domain of an antibody light chain via a synthetic peptide, and it can be generated using routine recombinant DNA technology techniques (see, e.g., Janeway et al., Immunobiology, 2nd Edition, Garland Publishing, New York, (1996)). Similarly, disulfide-stabilized variable region fragments (dsFv) can be prepared by recombinant DNA technology (see, e.g., Reiter et al., Protein Engineering, 7, 697-704 (1994)).

In exemplary aspects, the IL13Rα2 binding agent provided herein is an scFv. In exemplary aspects, the scFv comprises each of the amino acid sequences of SEQ ID NOs: 1-6. In exemplary aspects, the scFv comprises the amino acid sequence of SEQ ID NO: 7 or SEQ ID NO: 8. In exemplary aspects, the scFv comprises the amino acid sequences of SEQ ID NO: 7 and SEQ ID NO: 8. In exemplary aspects, the scFv comprises the amino acid sequences of SEQ ID NO: 7 and SEQ ID NO: 8 and the amino acid sequence of SEQ ID NO: 7 is fused to the amino acid sequence of SEQ ID NO: 8 through a linker. In exemplary aspects, the linker comprises a short amino acid sequence of about 5 to about 25 amino acids, e.g., about 10 to about 20 amino acids. In exemplary aspects, the linker comprises the amino acid sequence of EEGEFSEAR (SEQ ID NO 10). In exemplary aspects, the linker comprises the amino acid sequence of AKTTPPKLEEGEFSEARV (SEQ ID NO: 80). In exemplary aspects, the scFv provided herein comprises the amino acid sequence of SEQ ID NO: 13.

Recombinant antibody fragments, e.g., scFvs of the disclosure, can also be engineered to assemble into stable multimeric oligomers of high binding avidity and specificity to different target antigens. Such diabodies (dimers), triabodies (trimers) or tetrabodies (tetramers) are well known in the art. See e.g., Kortt et al., Biomol Eng. 2001 18:95-108, (2001) and Todorovska et al., J Immunol Methods. 248:47-66, (2001).

In exemplary aspects, the binding agent is a bispecific antibody (bscAb). Bispecific antibodies are molecules comprising two single-chain Fv fragments joined via a glycine-serine linker using recombinant methods. The V light-chain (VL) and V heavy-chain (VH) domains of two antibodies of interest in exemplary embodiments are isolated using standard PCR methods. The VL and VH cDNAs obtained from each hybridoma are then joined to form a single-chain fragment in a two-step fusion PCR. Bispecific fusion proteins are prepared in a similar manner. Bispecific single-chain antibodies and bispecific fusion proteins are antibody substances included within the scope of the present invention. Exemplary bispecific antibodies are taught in U.S. Patent Application Publication No. 2005-0282233A1 and International Patent Application Publication No. WO 2005/087812, both applications of which are incorporated herein by reference in their entireties.

In exemplary aspects, the binding agent is a bispecific T-cell engaging antibody (BiTE) containing two scFvs produced as a single polypeptide chain. In exemplary aspects, the binding agent is a BiTE comprising two scFvs, wherein at least one comprises each of the amino acid sequences of SEQ ID NOs: 1-6 or comprises SEQ ID NO: 7 and/or SEQ ID NO: 8. Methods of making and using BiTE antibodies are described in the art. See, e.g., Cioffi et al., Clin Cancer Res 18: 465, Brischwein et al., Mol Immunol 43:1129-43 (2006); Amann M et al., Cancer Res 68:143-51 (2008); Schlereth et al., Cancer Res 65: 2882-2889 (2005); and Schlereth et al., Cancer Immunol Immunother 55:785-796 (2006).

In exemplary aspects, the binding agent is a dual affinity re-targeting antibody (DART). DARTs are produced as separate polypeptides joined by a stabilizing interchain disulphide bond. In exemplary aspects, the binding agent is a DART comprising an scFv comprising each of the amino acid sequences of SEQ ID NOs: 1-6 or comprises SEQ ID NO: 7 and/or SEQ ID NO: 8. Methods of making and using DART antibodies are described in the art. See, e.g., Rossi et al., MAbs 6: 381-91 (2014); Fournier and Schirrmacher, BioDrugs 27:35-53 (2013); Johnson et al., J Mol Biol 399:436-449 (2010); Brien et al., J Virol 87: 7747-7753 (2013); and Moore et al., Blood 117:4542 (2011).

In exemplary aspects, the binding agent is a tetravalent tandem diabody (TandAbs) in which an antibody fragment is produced as a non-covalent homodimer folder in a head-to-tail arrangement. In exemplary aspects, the binding agent is a TandAbs comprising an scFv comprising each of the amino acid sequences of SEQ ID NOs: 1-6 or comprises SEQ ID NO: 7 and/or SEQ ID NO: 8. TandAbs are known in the art. See, e.g., McAleese et al., Future Oncol 8: 687-695 (2012); Portner et al., Cancer Immunol Immunother 61:1869-1875 (2012); and Reusch et al., MAbs 6:728 (2014).

In exemplary aspects, the BiTE, DART, or TandAbs comprises the CDRs of SEQ ID NOs: 1-6. In exemplary aspects, the BiTE, DART, or TandAbs comprises the amino acid sequence of SEQ ID NOs: 7 and 8. In exemplary aspects, the BiTE, DART, or TandAbs comprises SEQ ID NOs: 13.

Suitable methods of making antibodies are known in the art. For instance, standard hybridoma methods are described in, e.g., Harlow and Lane (eds.), Antibodies: A Laboratory Manual, CSH Press (1988), and CA. Janeway et al. (eds.), Immunobiology, 5th Ed., Garland Publishing, New York, NY (2001)).

Monoclonal antibodies for use in the invention may be prepared using any technique that provides for the production of antibody molecules by continuous cell lines in culture. These include, but are not limited to, the hybridoma technique originally described by Koehler and Milstein (Nature 256: 495-497, 1975), the human B-cell hybridoma technique (Kosbor et al., Immunol Today 4:72, 1983; Cote et al., Proc Natl Acad Sci 80: 2026-2030, 1983) and the EBV-hybridoma technique (Cole et al., Monoclonal Antibodies and Cancer Therapy, Alan R Liss Inc, New York N.Y., pp 77-96, (1985).

Briefly, a polyclonal antibody is prepared by immunizing an animal with an immunogen comprising a polypeptide of the present invention and collecting antisera from that immunized animal. A wide range of animal species can be used for the production of antisera. In some aspects, an animal used for production of anti-antisera is a non-human animal including rabbits, mice, rats, hamsters, goat, sheep, pigs or horses. Because of the relatively large blood volume of rabbits, a rabbit, in some exemplary aspects, is a preferred choice for production of polyclonal antibodies. In an exemplary method for generating a polyclonal antisera immunoreactive with the chosen IL13Rα2 epitope, 50 μg of IL13Rα2 antigen is emulsified in Freund's Complete Adjuvant for immunization of rabbits. At intervals of, for example, 21 days, 50 μg of epitope are emulsified in Freund's Incomplete Adjuvant for boosts. Polyclonal antisera may be obtained, after allowing time for antibody generation, simply by bleeding the animal and preparing serum samples from the whole blood.

Briefly, in exemplary embodiments, to generate monoclonal antibodies, a mouse is injected periodically with recombinant IL13Rα2 against which the antibody is to be raised (e.g., 10-20 μg IL13Rα2 emulsified in Freund's Complete Adjuvant). The mouse is given a final pre-fusion boost of a IL13Rα2 polypeptide containing the epitope that allows specific recognition of lymphatic endothelial cells in PBS, and four days later the mouse is sacrificed and its spleen removed. The spleen is placed in 10 ml serum-free RPMI 1640, and a single cell suspension is formed by grinding the spleen between the frosted ends of two glass microscope slides submerged in serum-free RPMI 1640, supplemented with 2 mM L-glutamine, 1 mM sodium pyruvate, 100 units/ml penicillin, and 100 μg/ml streptomycin (RPMI) (Gibco, Canada). The cell suspension is filtered through sterile 70-mesh Nitex cell strainer (Becton Dickinson, Parsippany, N.J.), and is washed twice by centrifuging at 200 g for 5 minutes and resuspending the pellet in 20 ml serum-free RPMI. Splenocytes taken from three naive Balb/c mice are prepared in a similar manner and used as a control. NS-1 myeloma cells, kept in log phase in RPMI with 11% fetal bovine serum (FBS) (Hyclone Laboratories, Inc., Logan, Utah) for three days prior to fusion, are centrifuged at 200 g for 5 minutes, and the pellet is washed twice.

Spleen cells (1×108) are combined with 2.0×107 NS-1 cells and centrifuged, and the supernatant is aspirated. The cell pellet is dislodged by tapping the tube, and 1 ml of 37° C. PEG 1500 (50% in 75 mM Hepes, pH 8.0) (Boehringer Mannheim) is added with stirring over the course of 1 minute, followed by the addition of 7 ml of serum-free RPMI over 7 minutes. An additional 8 ml RPMI is added and the cells are centrifuged at 200 g for 10 minutes. After discarding the supernatant, the pellet is resuspended in 200 ml RPMI containing 15% FBS, 100 μM sodium hypoxanthine, 0.4 μM aminopterin, 16 μM thymidine (HAT) (Gibco), 25 units/ml IL-6 (Boehringer Mannheim) and 1.5×106 splenocytes/ml and plated into 10 Corning flat-bottom 96-well tissue culture plates (Corning, Corning N.Y.).

On days 2, 4, and 6, after the fusion, 100 μl of medium is removed from the wells of the fusion plates and replaced with fresh medium. On day 8, the fusion is screened by ELISA, testing for the presence of mouse IgG binding to IL13Rα2 as follows. Immulon 4 plates (Dynatech, Cambridge, Mass.) are coated for 2 hours at 37° C. with 100 ng/well of IL13Rα2 diluted in 25 mM Tris, pH 7.5. The coating solution is aspirated and 200 μl/well of blocking solution (0.5% fish skin gelatin (Sigma) diluted in CMF-PBS) is added and incubated for 30 minutes at 37° C. Plates are washed three times with PBS containing 0.05% Tween 20 (PBST) and 50 μl culture supernatant is added. After incubation at 37° C. for 30 minutes, and washing as above, 50 μl of horseradish peroxidase-conjugated goat anti-mouse IgG(Fc) (Jackson ImmunoResearch, West Grove, Pa.) diluted 1:3500 in PBST is added. Plates are incubated as above, washed four times with PBST, and 100 μl substrate, consisting of 1 mg/ml o-phenylene diamine (Sigma) and 0.1 μl/ml 30% H2O2 in 100 mM citrate, pH 4.5, are added. The color reaction is stopped after 5 minutes with the addition of 50 μl of 15% H2SO4. The A490 absorbance is determined using a plate reader (Dynatech).

Selected fusion wells are cloned twice by dilution into 96-well plates and visual scoring of the number of colonies/well after 5 days. The monoclonal antibodies produced by hybridomas are isotyped using the Isostrip system (Boehringer Mannheim, Indianapolis, Ind.).

When the hybridoma technique is employed, myeloma cell lines may be used. Such cell lines suited for use in hybridoma-producing fusion procedures preferably are non-antibody-producing, have high fusion efficiency, and enzyme deficiencies that render them incapable of growing in certain selective media that support the growth of only the desired fused cells (hybridomas). For example, where the immunized animal is a mouse, one may use P3-X63/Ag8, P3-X63-Ag8.653, NS1/1.Ag 4 1, Sp210-Ag14, FO, NSO/U, MPC-11, MPC11-X45-GTG 1.7 and S194/15XX0 Bul; for rats, one may use R210.RCY3, Y3-Ag 1.2.3, IR983F and 4B210; and U-266, GM1500-GRG2, LICR-LON-HMy2 and UC729-6 are all useful in connection with cell fusions. It should be noted that the hybridomas and cell lines produced by such techniques for producing the monoclonal antibodies are contemplated to be compositions of the disclosure.

Depending on the host species, various adjuvants may be used to increase an immunological response. Such adjuvants include, but are not limited to, Freund's, mineral gels such as aluminum hydroxide, and surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanin, and dinitrophenol. BCG (bacilli Calmette-Guerin) and Corynebacterium parvum are potentially useful human adjuvants.

Alternatively, other methods, such as EBV-hybridoma methods (Haskard and Archer, J. Immunol. Methods, 74(2), 361-67 (1984), and Roder et al.5 Methods Enzymol., 121, 140-67 (1986)), and bacteriophage vector expression systems (see, e.g., Huse et al., Science, 246, 1275-81 (1989)) that are known in the art may be used. Further, methods of producing antibodies in non-human animals are described in, e.g., U.S. Pat. Nos. 5,545,806, 5,569,825, and 5,714,352, and U.S. Patent Application Publication No. 2002/0197266 A1).

Antibodies may also be produced by inducing in vivo production in the lymphocyte population or by screening recombinant immunoglobulin libraries or panels of highly specific binding reagents as disclosed in Orlandi et al. (Proc. Natl. Acad. Sci. 86: 3833-3837; 1989), and Winter and Milstein (Nature 349: 293-299, 1991).

Furthermore, phage display can be used to generate an antibody of the disclosure. In this regard, phage libraries encoding antigen-binding variable (V) domains of antibodies can be generated using standard molecular biology and recombinant DNA techniques (see, e.g., Sambrook et al. (eds.), Molecular Cloning, A Laboratory Manual, 3rd Edition, Cold Spring Harbor Laboratory Press, New York (2001)). Phage encoding a variable region with the desired specificity are selected for specific binding to the desired antigen, and a complete or partial antibody is reconstituted comprising the selected variable domain. Nucleic acid sequences encoding the reconstituted antibody are introduced into a suitable cell line, such as a myeloma cell used for hybridoma production, such that antibodies having the characteristics of monoclonal antibodies are secreted by the cell (see, e.g., Janeway et al., supra, Huse et al., supra, and U.S. Pat. No. 6,265,150). Related methods also are described in U.S. Pat. Nos. 5,403,484; 5,571,698; 5,837,500; and 5,702,892. The techniques described in U.S. Pat. Nos. 5,780,279; 5,821,047; 5,824,520; 5,855,885; 5,858,657; 5,871,907; 5,969,108; 6,057,098; and 6,225,447, are also contemplated as useful in preparing antibodies according to the disclosure.

Antibodies can be produced by transgenic mice that are transgenic for specific heavy and light chain immunoglobulin genes. Such methods are known in the art and described in, for example U.S. Pat. Nos. 5,545,806 and 5,569,825, and Janeway et al., supra.

Methods for generating humanized antibodies are well known in the art and are described in detail in, for example, Janeway et al., supra, U.S. Pat. Nos. 5,225,539; 5,585,089; and 5,693,761; European Patent No. 0239400 B1; and United Kingdom Patent No. 2188638. Humanized antibodies can also be generated using the antibody resurfacing technology described in U.S. Pat. No. 5,639,641 and Pedersen et al., J. Mol. Biol., 235:959-973 (1994).

Techniques developed for the production of “chimeric antibodies,” the splicing of mouse antibody genes to human antibody genes to obtain a molecule with appropriate antigen specificity and biological activity, can be used (Morrison et al., Proc. Natl. Acad. Sci. 81: 6851-6855, 1984; Neuberger et al., Nature 312: 604-608, 1984; and Takeda et al., Nature 314: 452-454; 1985). Alternatively, techniques described for the production of single-chain antibodies (U.S. Pat. No. 4,946,778) can be adapted to produce IL13Rα2-specific single chain antibodies.

A preferred chimeric or humanized antibody has a human constant region, while the variable region, or at least a CDR, of the antibody is derived from a non-human species. Methods for humanizing non-human antibodies are well known in the art. (see U.S. Pat. Nos. 5,585,089, and 5,693,762). Generally, a humanized antibody has one or more amino acid residues introduced into a CDR region and/or into its framework region from a source which is non-human. Humanization can be performed, for example, using methods described in Jones et al. (Nature 321: 522-525, 1986), Riechmann et al., (Nature, 332: 323-327, 1988) and Verhoeyen et al. (Science 239:1534-1536, 1988), by substituting at least a portion of a rodent complementarity-determining region (CDR) for the corresponding region of a human antibody. Numerous techniques for preparing engineered antibodies are described, e.g., in Owens and Young, J. Immunol. Meth., 168:149-165 (1994). Further changes can then be introduced into the antibody framework to modulate affinity or immunogenicity.

Consistent with the foregoing description, compositions comprising CDRs may be generated using, at least in part, techniques known in the art to isolate CDRs. Complementarity-determining regions are characterized by six polypeptide loops, three loops for each of the heavy or light chain variable regions. The amino acid position in a CDR is defined by Kabat et al., “Sequences of Proteins of Immunological Interest,” U.S. Department of Health and Human Services, (1983), which is incorporated herein by reference. For example, hypervariable regions of human antibodies are roughly defined to be found at residues 28 to 35, from 49-59 and from residues 92-103 of the heavy and light chain variable regions [Janeway et al., supra]. The murine CDRs also are found at approximately these amino acid residues. It is understood in the art that CDR regions may be found within several amino acids of the approximated amino acid positions set forth above. An immunoglobulin variable region also consists of four “framework” regions surrounding the CDRs (FR1-4). The sequences of the framework regions of different light or heavy chains are highly conserved within a species, and are also conserved between human and murine sequences.

Compositions comprising one, two, and/or three CDRs of a heavy chain variable region or a light chain variable region of a monoclonal antibody are generated. For example, using antibody of hybridoma clone 47 comprising the CDRs having the sequences of SEQ ID NOs: 1-6, polypeptide compositions comprising these CDRs are generated. Polypeptide compositions comprising one, two, three, four, five and/or six complementarity-determining regions of an antibody are also contemplated. Using the conserved framework sequences surrounding the CDRs, PCR primers complementary to these consensus framework sequences are generated to amplify the CDR sequence located between the primer regions. Techniques for cloning and expressing nucleotide and polypeptide sequences are well-established in the art [see e.g., Sambrook et al., Molecular Cloning. A Laboratory Manual, 2nd Edition, Cold Spring Harbor, New York (1989)]. The amplified CDR sequences are ligated into an appropriate plasmid. The plasmid comprising one, two, three, four, five and/or six cloned CDRs optionally contains additional polypeptide encoding regions linked to the CDR.

It is contemplated that modified polypeptide compositions comprising one, two, three, four, five, or six CDRs of a heavy or light chain of SEQ ID NOs: 1-6 are generated, wherein a CDR is altered to provide increased specificity or affinity or avidity to the target IL13Rα2. Sites at locations in the CDRs are typically modified in series, e.g., by substituting first with conservative choices (e.g., hydrophobic amino acid substituted for a non-identical hydrophobic amino acid) and then with more dissimilar choices (e.g., hydrophobic amino acid substituted for a charged amino acid), and then deletions or insertions may be made at the target site.

Framework regions (FR) of a murine antibody are humanized by substituting compatible human framework regions chosen from a large database of human antibody variable sequences, including over twelve hundred human VH sequences and over one thousand VL sequences. The database of antibody sequences used for comparison is downloaded from Andrew C. R. Martin's KabatMan web page (http://www.rubic.rdg.ac.uk/abs/). The Kabat method for identifying CDRs provides a means for delineating the approximate CDR and framework regions of any human antibody and comparing the sequence of a murine antibody for similarity to determine the CDRs and FRs. Best matched human VH and VL sequences are chosen on the basis of high overall framework matching, similar CDR length, and minimal mismatching of canonical and VH/VL contact residues. Human framework regions most similar to the murine sequence are inserted between the murine CDRs. Alternatively, the murine framework region may be modified by making amino acid substitutions of all or part of the native framework region that more closely resemble a framework region of a human antibody.

“Conservative” amino acid substitutions are made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the residues involved. For example, nonpolar (hydrophobic) amino acids include alanine (Ala, A), leucine (Leu, L), isoleucine (Ile, I), valine (Val, V), proline (Pro, P), phenylalanine (Phe, F), tryptophan (Trp, W), and methionine (Met, M); polar neutral amino acids include glycine (Gly, G), serine (Ser, S), threonine (Thr, T), cysteine (Cys, C), tyrosine (Tyr, Y), asparagine (Asn, N), and glutamine (Gln, Q); positively charged (basic) amino acids include arginine (Arg, R), lysine (Lys, K), and histidine (His, H); and negatively charged (acidic) amino acids include aspartic acid (Asp, D) and glutamic acid (Glu, E). “Insertions” or “deletions” are preferably in the range of about 1 to 20 amino acids, more preferably 1 to 10 amino acids. The variation may be introduced by systematically making substitutions of amino acids in a polypeptide molecule using recombinant DNA techniques and assaying the resulting recombinant variants for activity. Nucleic acid alterations can be made at sites that differ in the nucleic acids from different species (variable positions) or in highly conserved regions (constant regions). Methods for expressing polypeptide compositions useful in the invention are described in greater detail below.

Additionally, another useful technique for generating antibodies for use in the methods of the disclosure may be one which uses a rational design-type approach. The goal of rational design is to produce structural analogs of biologically active polypeptides or compounds with which they interact (agonists, antagonists, inhibitors, peptidomimetics, binding partners, and the like). In this case, the active polypeptides comprise the sequences of SEQ ID NOs: 1-6 disclosed herein. By creating such analogs, it is possible to fashion additional antibodies which are more immunoreactive than the native or natural molecule. In one approach, one would generate a three-dimensional structure for the antibodies or an epitope binding fragment thereof. This could be accomplished by x-ray crystallography, computer modeling or by a combination of both approaches. An alternative approach, “alanine scan,” involves the random replacement of residues throughout a molecule with alanine, and the resulting effect on function is determined.

It also is possible to solve the crystal structure of the specific antibodies. In principle, this approach yields a pharmacore upon which subsequent drug design can be based. It is possible to bypass protein crystallography altogether by generating anti-idiotypic antibodies to a functional, pharmacologically active antibody. As a mirror image of a mirror image, the binding site of anti-idiotype antibody is expected to be an analog of the original antigen. The anti-idiotype antibody is then be used to identify and isolate additional antibodies from banks of chemically- or biologically-produced peptides.

Chemically synthesized bispecific antibodies may be prepared by chemically crosslinking heterologous Fab or F(ab′)2 fragments by means of chemicals such as heterobifunctional reagent succinimidyl-3-(2-pyridyldithiol)-propionate (SPDP, Pierce Chemicals, Rockford, Ill.). The Fab and F(ab′)2 fragments can be obtained from intact antibody by digesting it with papain or pepsin, respectively (Karpovsky et al., J. Exp. Med. 160:1686-701, 1984; Titus et al., J. Immunol., 138:4018-22, 1987).

Methods of testing antibodies for the ability to bind to the epitope of the IL13Rα2, regardless of how the antibodies are produced, are known in the art and include any antibody-antigen binding assay such as, for example, radioimmunoassay (RIA), ELISA, Western blot, immunoprecipitation, and competitive inhibition assays (see, e.g., Janeway et al., infra, and U.S. Patent Application Publication No. 2002/0197266 A1).

Selection of antibodies from an antibody population for purposes herein also include using blood vessel endothelial cells to “subtract” those antibodies that cross-react with epitopes on such cells other than IL13Rα2 epitopes. The remaining antibody population is enriched in antibodies preferential for IL13Rα2 epitopes.

Aptamers

Recent advances in the field of combinatorial sciences have identified short polymer sequences (e.g., oligonucleic acid or peptide molecules) with high affinity and specificity to a given target. For example, SELEX technology has been used to identify DNA and RNA aptamers with binding properties that rival mammalian antibodies, the field of immunology has generated and isolated antibodies or antibody fragments which bind to a myriad of compounds, and phage display has been utilized to discover new peptide sequences with very favorable binding properties. Based on the success of these molecular evolution techniques, it is certain that molecules can be created which bind to any target molecule. A loop structure is often involved with providing the desired binding attributes as in the case of aptamers, which often utilize hairpin loops created from short regions without complementary base pairing, naturally derived antibodies that utilize combinatorial arrangement of looped hyper-variable regions and new phage-display libraries utilizing cyclic peptides that have shown improved results when compared to linear peptide phage display results. Thus, sufficient evidence has been generated to indicate that high affinity ligands can be created and identified by combinatorial molecular evolution techniques. For the present disclosure, molecular evolution techniques can be used to isolate binding agents specific for the IL13Rα2 disclosed herein. For more on aptamers, see generally, Gold, L., Singer, B., He, Y. Y., Brody. E., “Aptamers As Therapeutic And Diagnostic Agents,” J. Biotechnol. 74:5-13 (2000). Relevant techniques for generating aptamers are found in U.S. Pat. No. 6,699,843, which is incorporated herein by reference in its entirety.

In some embodiments, the aptamer is generated by preparing a library of nucleic acids; contacting the library of nucleic acids with a growth factor, wherein nucleic acids having greater binding affinity for the growth factor (relative to other library nucleic acids) are selected and amplified to yield a mixture of nucleic acids enriched for nucleic acids with relatively higher affinity and specificity for binding to the growth factor. The processes may be repeated, and the selected nucleic acids mutated and rescreened, whereby a growth factor aptamer is identified. Nucleic acids may be screened to select for molecules that bind to more than target. Binding more than one target can refer to binding more than one simultaneously or competitively. In some embodiments, a binding agent comprises at least one aptamer, wherein a first binding unit binds a first epitope of an IL13Rα2 and a second binding unit binds a second epitope of the IL13Rα2.

With regard to the binding agents of the compositions of the disclosure, ligand-induced activation of the IL13Rα2 is reduced upon binding of the binding agent to the IL13Rα2. As used herein, the term “reduce” as well as like terms, e.g., “inhibit,” do not necessarily imply 100% or a complete reduction or inhibition. Rather, there are varying degrees of reduction or inhibition of which one of ordinary skill in the art recognizes as having a potential benefit or therapeutic effect. Accordingly, in some embodiments, ligand-induced activation of the IL13Rα2 is completely abolished. In some embodiments, ligand-induced activation is substantially reduced, e.g., reduced by about 10% (e.g., by about 20%, by about 30%, by about 40%, by about 50%, by about 60%, by about 70%, by about 80%, by about 90%) or more, as compared to ligand-induced activation of the IL13Rα2 when the binding agent is absent or not bound to the IL13Rα2. Methods of measuring ligand-induced activation of an IL13Rα2 are known in the art, and include, for example, the assays described in the Examples, below.

Conjugates

Conjugates comprising a targeting domain and an effector domain are disclosed herein. In exemplary embodiments, the conjugate comprises any one of the binding agents disclosed herein as the targeting domain to localize the conjugate to a cell expressing IL13Rα2, e.g., a tumor cell expressing the same, and an effector domain. In exemplary aspects, the conjugate is a fusion protein. In exemplary aspects, the conjugate is a chimeric protein. As used herein, the term “chimeric” refers to a molecule composed of parts of different origins. A chimeric molecule, as a whole, is non-naturally occurring, e.g., synthetic or recombinant, although the parts which comprise the chimeric molecule may be naturally occurring.

Exemplary Effector Domains

As used herein, the term “effector domain” refers to a portion of a conjugate that effects a desired biological function. In exemplary aspects, the effector domain identifies or locates IL13Rα2-expressing cells. For example, the effector domain may be a diagnostic agent, e.g., a radiolabel, a fluorescent label, an enzyme (e.g., that catalyzes a calorimetric or fluorometric reaction), a substrate, a solid matrix, or a carrier (e.g., biotin or avidin). The diagnostic agent in some aspects is an imaging agent. Many appropriate imaging agents are known in the art, as are methods of attaching the labeling agents to the peptides of the invention (see, e.g., U.S. Pat. Nos. 4,965,392; 4,472,509; 5,021,236; and 5,037,630; each incorporated herein by reference). The imaging agents are administered to a subject in a pharmaceutically acceptable carrier, and allowed to accumulate at a target site having the lymphatic endothelial cells. This imaging agent then serves as a contrast reagent for X-ray, magnetic resonance, positron emission tomography, single photon emission computed tomography (SPECT), or sonographic or scintigraphic imaging of the target site. Of course, it should be understood that the imaging may be performed in vitro where tissue from the subject is obtained through a biopsy, and the presence of lymphatic endothelial cells is determined with the aid of the imaging agents described herein in combination with histochemical techniques for preparing and fixing tissues. Paramagnetic ions useful in the imaging agents of the invention include for example chromium (III), manganese (II), iron (III), iron (II), cobalt (II), nickel (II) copper (II), neodymium (III), samarium (III), ytterbium(III), gadolinium (III), vanadium (II), terbium (III), dysprosium (III), holmium (III) and erbium (III). Ions useful for X-ray imaging include, but are not limited to, lanthanum (III), gold (III), lead (II) and particularly bismuth (III). Radioisotopes for diagnostic applications include for example, 211astatine, 14carbon, 51chromium, 36chlorine, 57cobalt, 67copper, 152europium, 67gallium, 3hydrogen, 123iodine, 125iodine, 111indium, 59iron, 32phosphorus, 186rhenium, 75selenium, 35sulphur, 99mtechnicium, 90yttrium, and 99zirconium.

The effector domain may be one which alters the physico-chemical characteristics of the conjugate, e.g., an effector which confers increased solubility and/or stability and/or half-life, resistance to proteolytic cleavage, modulation of clearance. In exemplary aspects, the effector domain is a polymer, a carbohydrate, or a lipid.

The polymer may be branched or unbranched. The polymer may be of any molecular weight. The polymer in some embodiments has an average molecular weight of between about 2 kDa to about 100 kDa (the term “about” indicating that in preparations of a water-soluble polymer, some molecules will weigh more, some less, than the stated molecular weight). The average molecular weight of the polymer is in some aspects between about 5 kDa and about 50 kDa, between about 12 kDa to about 40 kDa or between about 20 kDa to about 35 kDa. In some embodiments, the polymer is modified to have a single reactive group, such as an active ester for acylation or an aldehyde for alkylation, so that the degree of polymerization may be controlled. The polymer in some embodiments is water soluble so that the protein to which it is attached does not precipitate in an aqueous environment, such as a physiological environment. In some embodiments when, for example, the composition is used for therapeutic use, the polymer is pharmaceutically acceptable. Additionally, in some aspects, the polymer is a mixture of polymers, e.g., a co-polymer, a block co-polymer. In some embodiments, the polymer is selected from the group consisting of: polyamides, polycarbonates, polyalkylenes and derivatives thereof, including polyalkylene glycols, polyalkylene oxides, polyalkylene terepthalates, polymers of acrylic and methacrylic esters, including poly(methyl methacrylate), poly(ethyl methacrylate), poly(butylmethacrylate), poly(isobutyl methacrylate), poly(hexylmethacrylate), poly(isodecyl methacrylate), poly(lauryl methacrylate), poly(phenyl methacrylate), poly(methyl acrylate), poly(isopropyl acrylate), poly(isobutyl acrylate), and poly(octadecyl acrylate), polyvinyl polymers including polyvinyl alcohols, polyvinyl ethers, polyvinyl esters, polyvinyl halides, poly(vinyl acetate), and polyvinylpyrrolidone, polyglycolides, polysiloxanes, polyurethanes and co-polymers thereof, celluloses including alkyl cellulose, hydroxyalkyl celluloses, cellulose ethers, cellulose esters, nitro celluloses, methyl cellulose, ethyl cellulose, hydroxypropyl cellulose, hydroxy-propyl methyl cellulose, hydroxybutyl methyl cellulose, cellulose acetate, cellulose propionate, cellulose acetate butyrate, cellulose acetate phthalate, carboxylethyl cellulose, cellulose triacetate, and cellulose sulphate sodium salt, polypropylene, polyethylenes including poly(ethylene glycol), poly(ethylene oxide), and poly(ethylene terephthalate), and polystyrene. In some aspects, the polymer is a biodegradable polymer, including a synthetic biodegradable polymer (e.g., polymers of lactic acid and glycolic acid, polyanhydrides, poly(ortho)esters, polyurethanes, poly(butic acid), poly(valeric acid), and poly(lactide-cocaprolactone)), and a natural biodegradable polymer (e.g., alginate and other polysaccharides including dextran and cellulose, collagen, chemical derivatives thereof (substitutions, additions of chemical groups, for example, alkyl, alkylene, hydroxylations, oxidations, and other modifications routinely made by those skilled in the art), albumin and other hydrophilic proteins (e.g., zein and other prolamines and hydrophobic proteins)), as well as any copolymer or mixture thereof. In general, these materials degrade either by enzymatic hydrolysis or exposure to water in vivo, by surface or bulk erosion. In some aspects, the polymer is a bioadhesive polymer, such as a bioerodible hydrogel described by H. S. Sawhney, C. P. Pathak and J. A. Hubbell in Macromolecules, 1993, 26, 581-587, the teachings of which are incorporated herein, polyhyaluronic acids, casein, gelatin, glutin, polyanhydrides, polyacrylic acid, alginate, chitosan, poly(methyl methacrylates), poly(ethyl methacrylates), poly(butylmethacrylate), poly(isobutyl methacrylate), poly(hexylmethacrylate), poly(isodecyl methacrylate), poly(lauryl methacrylate), poly(phenyl methacrylate), poly(methyl acrylate), poly(isopropyl acrylate), poly(isobutyl acrylate), and poly(octadecyl acrylate). In some embodiments, the polymer is a water-soluble polymer or a hydrophilic polymer. Suitable water-soluble polymers are known in the art and include, for example, polyvinylpyrrolidone, hydroxypropyl cellulose (HPC; Klucel), hydroxypropyl methylcellulose (HPMC; Methocel), nitrocellulose, hydroxypropyl ethylcellulose, hydroxypropyl butylcellulose, hydroxypropyl pentylcellulose, methyl cellulose, ethylcellulose (Ethocel), hydroxyethyl cellulose, various alkyl celluloses and hydroxyalkyl celluloses, various cellulose ethers, cellulose acetate, carboxymethyl cellulose, sodium carboxymethyl cellulose, calcium carboxymethyl cellulose, vinyl acetate/crotonic acid copolymers, poly-hydroxyalkyl methacrylate, hydroxymethyl methacrylate, methacrylic acid copolymers, polymethacrylic acid, polymethylmethacrylate, maleic anhydride/methyl vinyl ether copolymers, poly vinyl alcohol, sodium and calcium polyacrylic acid, polyacrylic acid, acidic carboxy polymers, carboxypolymethylene, carboxyvinyl polymers, polyoxyethylene polyoxypropylene copolymer, polymethylvinylether co-maleic anhydride, carboxymethylamide, potassium methacrylate divinylbenzene co-polymer, polyoxyethyleneglycols, polyethylene oxide, and derivatives, salts, and combinations thereof. In some aspects, the water-soluble polymers or mixtures thereof include, but are not limited to, N-linked or O-linked carbohydrates, sugars, phosphates, carbohydrates; sugars; phosphates; polyethylene glycol (PEG) (including the forms of PEG that have been used to derivatize proteins, including mono-(C1-C 10) alkoxy- or aryloxy-polyethylene glycol); monomethoxy-polyethylene glycol; dextran (such as low molecular weight dextran of, for example, about 6 kD), cellulose; other carbohydrate-based polymers, poly-(N-vinyl pyrrolidone), polyethylene glycol, propylene glycol homopolymers, a polypropylene oxide/ethylene oxide co-polymer, polyoxyethylated polyols (e.g., glycerol) and polyvinyl alcohol. Also encompassed by the disclosure are bifunctional crosslinking molecules which may be used to prepare covalently attached multimers. A particularly preferred water-soluble polymer for use herein is polyethylene glycol (PEG). As used herein, polyethylene glycol is meant to encompass any of the forms of PEG that can be used to derivatize other proteins, such as mono-(C1-C10) alkoxy- or aryloxy-polyethylene glycol. PEG is a linear or branched neutral polyether, available in a broad range of molecular weights, and is soluble in water and most organic solvents. PEG is effective at excluding other polymers or peptides when present in water, primarily through its high dynamic chain mobility and hydrophobic nature, thus creating a water shell or hydration sphere when attached to other proteins or polymer surfaces. PEG is nontoxic, non-immunogenic, and approved by the Food and Drug Administration for internal consumption. Proteins or enzymes when conjugated to PEG have demonstrated bioactivity, non-antigenic properties, and decreased clearance rates when administered in animals. F. M. Veronese et al., Preparation and Properties of Monomethoxypoly(ethylene glycol)-modified Enzymes for Therapeutic Applications, in J. M. Harris ed., Poly(Ethylene Glycol) Chemistry-Biotechnical and Biomedical Applications, 127-36, 1992, incorporated herein by reference. Without wishing to be bound by theory, these phenomena may be due to the exclusion properties of PEG in preventing recognition by the immune system. In addition, PEG has been widely used in surface modification procedures to decrease protein adsorption and improve blood compatibility. S. W. Kim et al., Ann. N.Y. Acad. Sci. 516: 116-30 1987; Jacobs et al., Artif. Organs 12: 500-501, 1988; Park et al., J. Poly. Sci, Part A 29:1725-31, 1991, each incorporated herein by reference in its entirety. Hydrophobic polymer surfaces, such as polyurethanes and polystyrene, can be modified by the grafting of PEG (MW 3,400) and employed as nonthrombogenic surfaces. Surface properties (contact angle) can be more consistent with hydrophilic surfaces, due to the hydrating effect of PEG. More importantly, protein (albumin and other plasma proteins) adsorption can be greatly reduced, resulting from the high chain motility, hydration sphere, and protein exclusion properties of PEG. PEG (MW 3,400) was determined as an optimal size in surface immobilization studies, Park et al., J. Biomed. Mat. Res. 26:739-45, 1992, while PEG (MW 5,000) was most beneficial in decreasing protein antigenicity. F. M. Veronese et al., In J. M. Harris, et al., Poly(Ethylene Glycol) Chemistry-Biotechnical and Biomedical Applications, 127-36. Methods for preparing pegylated binding agent polypeptides may comprise the steps of (a) reacting the polypeptide with polyethylene glycol (such as a reactive ester or aldehyde derivative of PEG) under conditions whereby the binding agent polypeptide becomes attached to one or more PEG groups, and (b) obtaining the reaction product(s). In general, the optimal reaction conditions for the acylation reactions will be determined based on known parameters and the desired result. For example, the larger the ratio of PEG:protein, the greater the percentage of poly-pegylated product. In some embodiments, the binding agent will have a single PEG moiety at the N-terminus. See U.S. Pat. No. 8,234,784, incorporated by reference herein.

In some embodiments, the effector domain is a carbohydrate. In some embodiments, the carbohydrate is a monosaccharide (e.g., glucose, galactose, fructose), a disaccharide (e.g., sucrose, lactose, maltose), an oligosaccharide (e.g., raffinose, stachyose), a polysaccharide (e.g., a starch, amylase, amylopectin, cellulose, chitin, callose, laminarin, xylan, mannan, fucoidan, or galactomannan).

In some embodiments, the effector domain is a lipid. The lipid, in some embodiments, is a fatty acid, eicosanoid, prostaglandin, leukotriene, thromboxane, N-acyl ethanolamine, glycerolipid (e.g., mono-, di-, tri-substituted glycerols), glycerophospholipid (e.g., phosphatidylcholine, phosphatidylinositol, phosphatidylethanolamine, phosphatidylserine), sphingolipid (e.g., sphingosine, ceramide), sterol lipid (e.g., steroid, cholesterol), prenol lipid, saccharolipid, or a polyketide, oil, wax, cholesterol, sterol, fat-soluble vitamin, monoglyceride, diglyceride, triglyceride, or a phospholipid.

Lethal Domains

In exemplary aspects, the effector domain is a lethal domain that confers lethality, such that when the conjugate is localized to a cell expressing IL13Rα2, e.g., a tumor cell expressing the same. The effector domain confers upon the conjugate the ability to kill an IL13Rα2-expressing cell once the binding agent has found and bound to its IL13Rα2 target.

In exemplary aspects, the effector domain is a cytotoxin (also referred to herein as a “cytotoxic agent”). The cytotoxic agent is any molecule (chemical or biochemical) which is toxic to a cell. In some embodiments, the cytotoxic agent is a chemotherapeutic agent. Chemotherapeutic agents are known in the art and include, but are not limited to, platinum coordination compounds, topoisomerase inhibitors, antibiotics, antimitotic alkaloids and difluoronucleosides, as described in U.S. Pat. No. 6,630,124. In some embodiments, the chemotherapeutic agent is a platinum coordination compound. The term “platinum coordination compound” refers to any tumor cell growth-inhibiting platinum coordination compound that provides the platinum in the form of an ion. In some embodiments, the platinum coordination compound is cis-diamminediaquoplatinum (II)-ion; chloro(diethylenetriamine)-platinum(II)chloride; dichloro(ethylenediamine)-platinum(II), diammine(1,1-cyclobutanedicarboxylato) platinum(II) (carboplatin); spiroplatin; iproplatin; diammine(2-ethylmalonato)-platinum(II); ethylenediaminemalonatoplatinum(II); aqua(1,2-diaminodyclohexane)-sulfatoplatinum(II); (1,2-diaminocyclohexane)malonatoplatinum(II); (4-caroxyphthalatoX1,2-diaminocyclohexane)platinum(II); (1,2-diaminocyclohexane)-(isocitrato)platinum(II); (1,2-diaminocyclohexane)cis(pyruvato)platinum(II); (1,2-diaminocyclohexane)oxalatoplatinum(II); ormaplatin; or tetraplatin. In some embodiments, cisplatin is the platinum coordination compound employed in the compositions and methods of the present invention. Cisplatin is commercially available under the name PLATINOL™ from Bristol Myers-Squibb Corporation and is available as a powder for constitution with water, sterile saline or other suitable vehicle. Other platinum coordination compounds suitable for use in the present invention are known and are available commercially and/or can be prepared by conventional techniques. Cisplatin, or cis-dichlorodiammineplatinum II, has been used successfully for many years as a chemotherapeutic agent in the treatment of various human solid malignant tumors. More recently, other diamino-platinum complexes have also shown efficacy as chemotherapeutic agents in the treatment of various human, solid, malignant tumors. Such diamino-platinum complexes include, but are not limited to, spiroplatinum and carboplatinum. Although cisplatin and other diamino-platinum complexes have been widely used as chemotherapeutic agents in humans, they have had to be delivered at high dosage levels that can lead to toxicity problems such as kidney damage.

In some embodiments, the chemotherapeutic agent is a topoisomerase inhibitor. Topoisomerases are enzymes that are capable of altering DNA topology in eukaryotic cells. They are critical for cellular functions and cell proliferation. Generally, there are two classes of topoisomerases in eukaryotic cells, type I and type II. Topoisomerase I is a monomeric enzyme of approximately 100,000 molecular weight. The enzyme binds to DNA and introduces a transient single-strand break, unwinds the double helix (or allows it to unwind), and subsequently reseals the break before dissociating from the DNA strand. Various topoisomerase inhibitors have recently shown clinical efficacy in the treatment of humans afflicted with ovarian cancer, esophageal cancer or non-small cell lung carcinoma. In some aspects, the topoisomerase inhibitor is camptothecin or a camptothecin analog. Camptothecin is a water-insoluble, cytotoxic alkaloid produced by Camptotheca accuminata trees indigenous to China and Nothapodytes foetida trees indigenous to India. Camptothecin exhibits tumor cell growth-inhibiting activity against a number of tumor cells. Compounds of the camptothecin analog class are typically specific inhibitors of DNA topoisomerase I. By the term “inhibitor of topoisomerase” is meant any tumor cell growth-inhibiting compound that is structurally related to camptothecin. Compounds of the camptothecin analog class include, but are not limited to; topotecan, irinotecan and 9-amino-camptothecin. In additional embodiments, the cytotoxic agent is any tumor cell growth-inhibiting camptothecin analog claimed or described in U.S. Pat. No. 5,004,758; European Patent Application Number 88311366.4 (Publication Number EP 0 321 122); U.S. Pat. No. 4,604,463; European Patent Application Publication Number EP 0 137 145; U.S. Pat. No. 4,473,692; European Patent Application Publication Number EP 0 074 256; U.S. Pat. No. 4,545,880; European Patent Application Publication Number EP 0 074 256; European Patent Application Publication Number EP 0 088 642; Wani et al., J. Med. Chem., 29, 2358-2363 (1986); and Nitta et al., Proc. 14th International Congr. Chemotherapy, Kyoto, 1985, Tokyo Press, Anticancer Section 1, p. 28-30. In particular, the disclosure contemplates a compound called CPT-11. CPT-11 is a camptothecin analog with a 4-(piperidino)-piperidine side chain joined through a carbamate linkage at C-10 of 10-hydroxy-7-ethyl camptothecin. CPT-11 is currently undergoing human clinical trials and is also referred to as irinotecan; Wani et al, J. Med. Chem., 23, 554 (1980); Wani et al., J. Med. Chem., 30, 1774 (1987); U.S. Pat. No. 4,342,776; European Patent Application Publication Number EP 418 099; U.S. Pat. No. 4,513,138; European Patent Application Publication Number EP 0 074 770; U.S. Pat. No. 4,399,276; European Patent Application Publication Number 0 056 692; the entire disclosure of each of which is hereby incorporated by reference. All of the above-listed compounds of the camptothecin analog class are available commercially and/or can be prepared by conventional techniques including those described in the above-listed references. The topoisomerase inhibitor may be selected from the group consisting of topotecan, irinotecan and 9-aminocamptothecin.

The preparation of numerous compounds of the camptothecin analog class (including pharmaceutically acceptable salts, hydrates and solvates thereof) as well as the preparation of oral and parenteral pharmaceutical compositions comprising such a compound of the camptothecin analog class and an inert, pharmaceutically acceptable carrier or diluent, is extensively described in U.S. Pat. No. 5,004,758; and European Patent Application Number 88311366.4 (Publication Number EP 0 321 122), the teachings of each of which are incorporated herein by reference in its entirety.

In still another embodiment of the invention, the chemotherapeutic agent is an antibiotic compound. Suitable antibiotics include, but are not limited to, doxorubicin, mitomycin, bleomycin, daunorubicin and streptozocin. In some embodiments, the chemotherapeutic agent is an antimitotic alkaloid. In general, antimitotic alkaloids can be extracted from Catharanthus roseus, and have been shown to be efficacious as anticancer chemotherapy agents. A great number of semi-synthetic derivatives have been studied both chemically and pharmacologically (see, O. Van Tellingen et al, Anticancer Research, 12, 1699-1716 (1992)). The antimitotic alkaloids of the present invention include, but are not limited to, vinblastine, vincristine, vindesine, Taxol and vinorelbine. The latter two antimitotic alkaloids are commercially available from Eli Lilly and Company, and Pierre Fabre Laboratories, respectively (see, U.S. Pat. No. 5,620,985). In one aspect of the disclosure, the antimitotic alkaloid is vinorelbine.

In another embodiment of the invention, the chemotherapeutic agent is a difluoronucleoside. 2′-deoxy-2′,2′-difluoronucleosides are known in the art as having antiviral activity. Such compounds are disclosed and taught in U.S. Pat. Nos. 4,526,988 and 4,808,614. European Patent Application Publication 184,365 discloses that these same difluoronucleosides have oncolytic activity. In certain specific aspects, the 2′-deoxy-2′,2′-difluoronucleoside used in the compositions and methods of the disclosure is 2′-deoxy-2′,2′-difluorocytidine hydrochloride, also known as gemcitabine hydrochloride. Gemcitabine is commercially available or can be synthesized in a multi-step process as disclosed in U.S. Pat. Nos. 4,526,988, 4,808,614 and 5,223,608, the teachings of each of which are incorporated herein by reference in its entirety.

In exemplary aspects, the effector domain is an apoptosis tag which causes the IL13Rα2-expressing cell to apoptose. In exemplary aspects, the apoptosis tag is a TRAIL protein, or a portion thereof. In exemplary aspects, the apoptosis tag comprises the amino acid sequence of SEQ ID NO: 27. In exemplary aspects, the conjugate comprises the amino acid sequence of SEQ ID NO: 25.

In exemplary embodiments, the effector domain is an Fc domain of IgG or other immunoglobulin. For substituents such as an Fc region of human IgG, the fusion can be fused directly to a binding agent or fused through an intervening sequence. For example, a human IgG hinge, CH2 and CH3 region may be fused at either the N-terminus or C-terminus of a binding agent to attach the Fc region. The resulting Fc-fusion agent enables purification via a Protein A affinity column (Pierce, Rockford, Ill.). Peptide and proteins fused to an Fc region can exhibit a substantially greater half-life in vivo than the unfused counterpart. A fusion to an Fc region allows for dimerization/multimerization of the fusion polypeptide. The Fc region may be a naturally occurring Fc region, or may be modified for superior characteristics, e.g., therapeutic qualities, circulation time, reduced aggregation. As noted above, in some embodiments, the binding agent are conjugated, e.g., fused to an immunoglobulin or portion thereof (e.g., variable region, CDR, or Fc region). Known types of immunoglobulins (Ig) include IgG, IgA, IgE, IgD or IgM. The Fc region is a C-terminal region of an Ig heavy chain, which is responsible for binding to Fc receptors that carry out activities such as recycling (which results in prolonged half-life), antibody dependent cell-mediated cytotoxicity (ADCC), and complement dependent cytotoxicity (CDC).

For example, according to some definitions the human IgG heavy chain Fc region stretches from Cys226 to the C-terminus of the heavy chain. The “hinge region” generally extends from Glu216 to Pro230 of human IgG1 (hinge regions of other IgG isotypes may be aligned with the IgG1 sequence by aligning the cysteines involved in cysteine bonding). The Fc region of an IgG includes two constant domains, CH2 and CH3. The CH2 domain of a human IgG Fc region usually extends from amino acids 231 to amino acid 341. The CH3 domain of a human IgG Fc region usually extends from amino acids 342 to 447. References made to amino acid numbering of immunoglobulins or immunoglobulin fragments, or regions, are all based on Kabat et al. 1991, Sequences of Proteins of Immunological Interest, U.S. Department of Public Health, Bethesda, Md., incorporated herein by reference. In related embodiments, the Fc region may comprise one or more native or modified constant regions from an immunoglobulin heavy chain, other than CH1, for example, the CH2 and CH3 regions of IgG and IgA, or the CH3 and CH4 regions of IgE.

Suitable conjugate moieties include portions of immunoglobulin sequence that include the FcRn binding site. FcRn, a salvage receptor, is responsible for recycling immunoglobulins and returning them to circulation in the blood. The region of the Fc portion of IgG that binds to the FcRn receptor has been described based on X-ray crystallography (Burmeister et al. 1994, Nature 372:379). The major contact area of the Fc with the FcRn is near the junction of the CH2 and CH3 domains. Fc-FcRn contacts are all within a single Ig heavy chain. The major contact sites include amino acid residues 248, 250-257, 272, 285, 288, 290-291, 308-311, and 314 of the CH2 domain and amino acid residues 385-387, 428, and 433-436 of the CH3 domain.

Some conjugate moieties may or may not include FcγR binding site(s). FcγR are responsible for antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). Examples of positions within the Fc region that make a direct contact with FcγR are amino acids 234-239 (lower hinge region), amino acids 265-269 (B/C loop), amino acids 297-299 (C′/E loop), and amino acids 327-332 (F/G) loop (Sondermann et al., Nature 406: 267-273, 2000). The lower hinge region of IgE has also been implicated in the FcRI binding (Henry, et al., Biochemistry 36, 15568-15578, 1997). Residues involved in IgA receptor binding are described in Lewis et al., (J Immunol. 175:6694-701, 2005). Amino acid residues involved in IgE receptor binding are described in Sayers et al. (J Biol Chem. 279(34):35320-5, 2004).

Amino acid modifications may be made to the Fc region of an immunoglobulin. Such variant Fc regions comprise at least one amino acid modification in the CH3 domain of the Fc region (residues 342-447) and/or at least one amino acid modification in the CH2 domain of the Fc region (residues 231-341). Mutations believed to impart an increased affinity for FcRn include T256A, T307A, E380A, and N434A (Shields et al. 2001, J. Biol. Chem. 276:6591). Other mutations may reduce binding of the Fc region to FcγRI, FcγRIIA, FcγRIIB, and/or FcγRIIIA without significantly reducing affinity for FcRn. For example, substitution of the Asn at position 297 of the Fc region with Ala or another amino acid removes a highly conserved N-glycosylation site and may result in reduced immunogenicity with concomitant prolonged half-life of the Fc region, as well as reduced binding to FcγRs (Routledge et al. 1995, Transplantation 60:847; Friend et al. 1999, Transplantation 68:1632; Shields et al. 1995, J. Biol. Chem. 276:6591). Amino acid modifications at positions 233-236 of IgG1 have been made that reduce binding to FcγRs (Ward and Ghetie 1995, Therapeutic Immunology 2:77 and Armour et al. 1999, Eur. J. Immunol. 29:2613). Some exemplary amino acid substitutions are described in U.S. Pat. Nos. 7,355,008 and 7,381,408, each of which is incorporated by reference herein in its entirety.

In some embodiments, the binding agent is fused to alkaline phosphatase (AP). Methods for making Fc or AP fusion agents are provided in WO 02/060950.

Chimeric Antigen Receptors (CARs)

In exemplary aspects, the effector domain is a T-cell signaling domain. In exemplary aspects, the conjugate is a chimeric antigen receptor (CAR). Chimeric antigen receptors (CARs) are engineered transmembrane proteins that combine the specificity of an antigen-specific antibody with a T-cell receptor's function. In general, CARs comprise an ectodomain, a spacer region, a transmembrane domain, and an endodomain. The ectodomain of a CAR in exemplary aspects comprises an antigen recognition region, which may be an scFV of an antigen-specific antibody. The ectodomain also in some embodiments comprises a signal peptide which directs the nascent protein into the endoplasmic reticulum. In exemplary aspects, the ectodomain comprises a spacer which links the antigen recognition region to the transmembrane domain. The transmembrane (TM) domain is the portion of the CAR which traverses the cell membrane. In exemplary aspects, the TM domain comprises a hydrophobic alpha helix. In exemplary aspects, the TM domain comprises all or a portion of the TM domain of CD28. In exemplary aspects, the TM domain comprises all or a portion of the TM domain of CD8α. The endodomain of a CAR comprises one or more signaling domains. In exemplary aspects, the endodomain comprises the zeta chain of CD3, which comprises three copies of the Immunoreceptor Tyrosine-based Activation Motif (ITAM). An ITAM generally comprises a Tyr residue separated by two amino acids from a Leu or Ile. In the case of immune cell receptors, e.g., the T cell receptor and the B cell receptor, the ITAMs occur in multiples (at least two) and each ITAM is separated from another by 6-8 amino acids. The endodomain of CARs may also comprises additional signaling domains, e.g., portions of proteins that are important for downstream signal transduction. In exemplary aspects, the endodomain comprises signaling domains from one or more of CD28, 41BB or 4-1BB (CD137), ICOS, CD27, CD40, OX40 (CD134), or Myd88. Sequences encoding signaling domains of such proteins are provided herein as SEQ ID NOs: 39-42, 68-79, 81, and 83. Methods of making CARs, expressing them in cells, e.g., T-cells, and utilizing the CAR-expressing T-cells in therapy, are known in the art. See, e.g., International Patent Application Publication Nos. WO2014/208760, WO2014/190273, WO2014/186469, WO2014/184143, WO2014180306, WO2014/179759, WO2014/153270, U.S. Application Publication Nos. US20140369977, US20140322212, US20140322275, US20140322183, US20140301993, US20140286973, US20140271582, US20140271635, US20140274909, European Application Publication No. 2814846, each of which are incorporated by reference in their entirety.

In exemplary aspects, the conjugate of the disclosure is an IL13Rα2-specific chimeric antigen receptor (CAR) comprising a binding agent described herein, a hinge region, and an endodomain comprising a signaling domain of a CD3 zeta chain and a signaling domain of CD28, CD134, and/or CD137. In exemplary aspects, the CAR comprises (A) each of the amino acid sequence of: NYLMN (SEQ ID NO: 1); RIDPYDGDIDYNQNFKD (SEQ ID NO: 2); GYGTAYGVDY (SEQ ID NO: 3); RASESVDNYGISFMN (SEQ ID NO: 4); AASRQGSG (SEQ ID NO: 5); and QQSKEVPWT (SEQ ID NO: 6), (B) a hinge region; and (C) an endodomain comprising a signaling domain of a CD3 zeta chain and a signaling domain of CD28, CD134, and/or CD137. In exemplary aspects, the CD3 zeta chain signaling domain comprises the amino acid sequence of SEQ ID NO: 41. In exemplary aspects, the CAR further comprises a transmembrane (TM) domain based on the TM domain of CD28. In exemplary aspects, the CAR comprises the amino acid sequence of SEQ ID NO: 47. In exemplary aspects, the CAR further comprises a transmembrane (TM) domain based on the TM domain of CD8a. In exemplary aspects, the CAR comprises the amino acid sequence of SEQ ID NO: 85. In exemplary aspects, the hinge region comprises the amino acid sequence of SEQ ID NO: 35 or SEQ ID NO: 37. In exemplary aspects, the CAR comprises the amino acid sequence of SEQ ID NO: 49 or SEQ ID NO: 51. In exemplary aspects, the endodomain of the CAR of the disclosures comprises the amino acid sequence of SEQ ID NO: 53 or SEQ ID NO: 55. In exemplary aspects, the endodomain of the CAR of the disclosure comprises the amino acid sequence of SEQ ID NO: 87 or SEQ ID NO: 89. In exemplary aspects, the endodomain of the CAR of the disclosure comprises the amino acid sequence of SEQ ID NO: 91, SEQ ID NO: 93 or SEQ ID NO: 95. In exemplary aspects, the endodomain of the CAR of the disclosure comprises the amino acid sequence of SEQ ID NO: 97, SEQ ID NO: 99 or SEQ ID NO: 101.

In exemplary aspects, the endodomain further comprises a signaling domain of one or more of: CD137, CD134, CD27, CD40, ICOS, and Myd88. In exemplary aspects, the endodomain comprises one or more of the amino acid sequences of SEQ ID NOs: 68, 70, 72, 74, 76, and 78, which provide a sequence comprising a CD27 signaling domain, a sequence comprising a CD40 signaling domain, a sequence comprising a CD134 signaling domain, a sequence comprising a CD137 signaling domain, a sequence comprising an ICOS signaling domain, and a sequence comprising a Myd88 signaling domain, respectively.

In exemplary aspects, the CAR comprises (A) each of the amino acid sequence of: NYLMN (SEQ ID NO: 1); RIDPYDGDIDYNQNFKD (SEQ ID NO: 2); GYGTAYGVDY (SEQ ID NO: 3); RASESVDNYGISFMN (SEQ ID NO: 4); AASRQGSG (SEQ ID NO: 5); and QQSKEVPWT (SEQ ID NO: 6), (B) a hinge region; (C) an endodomain comprising a signaling domain of a CD3 zeta chain and a signaling domain of CD28 and at least one other signaling domain. In exemplary aspects, the CAR comprises an endodomain comprising a signaling domain of 41BB (CD137). In exemplary aspects the CAR comprises an endodomain comprising an amino acid sequence of SEQ ID NO: 81. In exemplary aspects, the CD137 signaling is N-terminal to a CD3 zeta chain signaling chain. In exemplary aspects, the endodomain comprises the amino acid sequence of SEQ ID NO: 87. In exemplary aspects, the CAR comprises the amino acid sequence of SEQ ID NO: 91. In exemplary aspects, the CAR comprises the amino acid sequence of SEQ ID NO: 97.

In exemplary aspects, the CAR comprises an endodomain comprising a signaling domain of OX40 (CD134). In exemplary aspects the CAR comprises an endodomain comprising an amino acid sequence of SEQ ID NO: 83. In exemplary aspects, the CD137 signaling is N-terminal to a CD3 zeta chain signaling chain. In exemplary aspects, the endodomain comprises the amino acid sequence of SEQ ID NO: 89. In exemplary aspects, the CAR comprises the amino acid sequence of SEQ ID NO: 95. In exemplary aspects, the CAR comprises the amino acid sequence of SEQ ID NO: 99.

In exemplary aspects, the CAR comprises (A) each of the amino acid sequence of: NYLMN (SEQ ID NO: 1); RIDPYDGDIDYNQNFKD (SEQ ID NO: 2); GYGTAYGVDY (SEQ ID NO: 3); RASESVDNYGISFMN (SEQ ID NO: 4); AASRQGSG (SEQ ID NO: 5); and QQSKEVPWT (SEQ ID NO: 6), (B) a hinge region; (C) a transmembrane domain of CD8α chain, and (D) an endodomain comprising a signaling domain of a CD3 zeta chain, and, optionally, at least one other signaling domain. In exemplary aspects, the transmembrane domain comprises the amino acid sequence of SEQ ID NO: 85. In exemplary aspects, the CAR further comprises a CD137 signaling domain and a CD3 zeta chain signaling domain. In exemplary aspects, the CAR comprises the amino acid sequence of SEQ ID NO: 93. In exemplary aspects, the CAR comprises the amino acid sequence of SEQ ID NO: 101.

As an example, sequences of three additional IL13Rα2-specific CARs are provided. One CAR contains a CD8α TM domain, and a 41BB.zeta signaling domain (SEQ ID NO:93 encoded by SEQ ID NO:94). The other two CARs contain a CD28 TM domain and either a CD28.CD134.zeta (SEQ ID NO:99 encoded by SEQ ID NO:100) or CD28.CD137.zeta (SEQ ID NO:101 encoded by SEQ ID NO:102) signaling domain.

Nucleic Acids, Vectors, Host Cells

Further provided by the disclosures is a nucleic acid comprising a nucleotide sequence encoding any of the binding agents and conjugates (e.g., chimeric proteins, fusion proteins, CARs) described herein. The nucleic acid may comprise any nucleotide sequence which encodes any of the binding agents and conjugates described herein. In exemplary aspects, the nucleic acid comprises a nucleotide sequence encoding each of the CDRs of SEQ ID NOs: 1-6. In exemplary aspects, the nucleic acid of the disclosures comprises a nucleic acid sequence which encodes a SEQ ID NO: 7 and/or SEQ ID NO: 8. In exemplary aspects, the nucleic acid of the disclosures comprises a nucleic acid sequence which encodes SEQ ID NO: 13 or SEQ ID NO: 14. In exemplary aspects, the nucleic acid provided herein comprises the sequence of SEQ ID NO: 15 and/or SEQ ID NO: 16. In exemplary aspects, the nucleic acid comprises a nucleotide sequence of SEQ ID NO: 66 or 67. In exemplary aspects, the nucleic acid comprises a nucleotide sequence which encodes the sequence of SEQ ID NO: 25. In exemplary aspects, the nucleic acid comprises a nucleotide sequence of SEQ ID NO: 26. In exemplary aspects, the nucleic acid comprises a nucleotide sequence which encodes each of SEQ ID NOs: 1-6 and encodes SEQ ID NO: 28 or 30. In exemplary aspects, the nucleic acid comprises a nucleotide sequence which encodes each of SEQ ID NOs: 1-6 and encodes an amino acid sequence which is at least 90% identical to SEQ ID NO: 28 or 30. In exemplary aspects, the nucleic acid comprises a nucleotide sequence which encodes each of SEQ ID NOs: 1-6 and comprises SEQ ID NO: 29 or 31. In exemplary aspects, the nucleic acid comprises a nucleotide sequence which encodes SEQ ID NO: 33. In exemplary aspects, the nucleic acid comprises a nucleotide sequence of SEQ ID NO: 34. In exemplary aspects, the nucleic acid comprises a nucleotide sequence which encodes each of SEQ ID NOs: 1-6 and encodes SEQ ID NO: 35 or 37. In exemplary aspects, the nucleic acid comprises a nucleotide sequence which encodes each of SEQ ID NOs: 1-6 and comprises SEQ ID NO: 36 or 38. In exemplary aspects, the nucleic acid comprises a nucleotide sequence which encodes each of SEQ ID NOs: 1-6 and encodes SEQ ID NO: 39 or 41. In exemplary aspects, the nucleic acid comprises a nucleotide sequence which encodes each of SEQ ID NOs: 1-6 and comprises SEQ ID NO: 40 or 42. In exemplary aspects, the nucleic acid comprises a nucleotide sequence which encodes each of SEQ ID NOs: 1-6 and encodes SEQ ID NO: 47. In exemplary aspects, the nucleic acid comprises a nucleotide sequence which encodes each of SEQ ID NOs: 1-6 and comprises SEQ ID NO: 48. In exemplary aspects, the nucleic acid comprises a nucleotide sequence which encodes each of SEQ ID NOs: 1-6 and encodes SEQ ID NO: 49 or 51. In exemplary aspects, the nucleic acid comprises a nucleotide sequence which encodes each of SEQ ID NOs: 1-6 and comprises SEQ ID NO: 50 or 52. In exemplary aspects, the nucleic acid comprises a nucleotide sequence which encodes SEQ ID NO: 53 or 55. In exemplary aspects, the nucleic acid comprises a nucleotide sequence which encodes each of SEQ ID NOs: 1-6 and comprises SEQ ID NO: 54 or 56. In exemplary aspects, the nucleic acid comprises a nucleotide sequence which encodes each of SEQ ID NOs: 1-6 and encodes one or more of SEQ ID NOs: 68, 70, 72, 74, 76, and 78. In exemplary aspects, the nucleic acid comprises a nucleotide sequence which encodes each of SEQ ID NOs: 1-6 and comprises one or more of SEQ ID NOs: 69, 71, 73, 75, 77, and 79. In exemplary aspects, the nucleic acid comprises a nucleotide sequence which encodes each of SEQ ID NOs: 1-6 and comprises one or more of SEQ ID NOs: 82, 84, 86, 88, 90, 92, 94, 96. In exemplary aspects, the nucleic acid comprises a nucleotide sequence comprising one of SEQ ID NOs: 98, 100, and 102.

By “nucleic acid” as used herein includes “polynucleotide,” “oligonucleotide,” and “nucleic acid molecule,” and generally means a polymer of DNA or RNA, which may be single-stranded or double-stranded, synthesized or obtained (e.g., isolated and/or purified) from natural sources, which may contain natural, non-natural or altered nucleotides, and which may contain a natural, non-natural or altered internucleotide linkage, such as a phosphoroamidate linkage or a phosphorothioate linkage, instead of the phosphodiester found between the nucleotides of an unmodified oligonucleotide. It is generally preferred that the nucleic acid does not comprise any insertions, deletions, inversions, and/or substitutions. However, it may be suitable in some instances, as discussed herein, for the nucleic acid to comprise one or more insertions, deletions, inversions, and/or substitutions.

In exemplary aspects, the nucleic acids of the disclosures are recombinant. As used herein, the term “recombinant” refers to (i) molecules that are constructed outside living cells by joining natural or synthetic nucleic acid segments to nucleic acid molecules that may replicate in a living cell, or (ii) molecules that result from the replication of those described in (i) above. For purposes herein, the replication may be in vitro replication or in vivo replication.

The nucleic acids in exemplary aspects are constructed based on chemical synthesis and/or enzymatic ligation reactions using procedures known in the art. See, for example, Sambrook et al., supra, and Ausubel et al., supra. For example, a nucleic acid may be chemically synthesized using naturally occurring nucleotides or variously modified nucleotides designed to increase the biological stability of the molecules or to increase the physical stability of the duplex formed upon hybridization (e.g., phosphorothioate derivatives and acridine substituted nucleotides). Examples of modified nucleotides that may be used to generate the nucleic acids include, but are not limited to, 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5-(carboxyhydroxymethyl) uracil, 5-carboxymethylaminomethyl-2-thiouridine, 5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N-substituted adenine, 7-methylguanine, 5-methylammomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta-D-mannosylqueosine, 5′-methoxycarboxymethyluracil, 5-methoxyuracil, 2-methylthio-N6-isopentenyladenine, uracil-5-oxyacetic acid (v), wybutoxosine, pseudouracil, queosine, 2-thiocytosine, 5-methyl-2-thiouracil, 2-thiouracil, 4-thiouracil, 5-methyluracil, uracil-5-oxyacetic acid methylester, 3-(3-amino-3-N-2-carboxypropyl) uracil, and 2,6-diaminopurine. Alternatively, one or more of the nucleic acids of the disclosures may be purchased from companies, such as Macromolecular Resources (Fort Collins, CO) and Synthegen (Houston, TX).

The nucleic acids of the disclosures in exemplary aspects are incorporated into a recombinant expression vector. In this regard, the disclosures provides recombinant expression vectors comprising any of the nucleic acids of the disclosures. For purposes herein, the term “recombinant expression vector” means a genetically-modified oligonucleotide or polynucleotide construct that permits the expression of an mRNA, protein, polypeptide, or peptide by a host cell, when the construct comprises a nucleotide sequence encoding the mRNA, protein, polypeptide, or peptide, and the vector is contacted with the cell under conditions sufficient to have the mRNA, protein, polypeptide, or peptide expressed within the cell. The vectors of the disclosures are not naturally-occurring as a whole. However, parts of the vectors may be naturally-occurring. The inventive recombinant expression vectors may comprise any type of nucleotides, including, but not limited to DNA and RNA, which may be single-stranded or double-stranded, synthesized or obtained in part from natural sources, and which may contain natural, non-natural or altered nucleotides. The recombinant expression vectors may comprise naturally-occurring or non-naturally occurring internucleotide linkages, or both types of linkages. In exemplary aspects, the altered nucleotides or non-naturally occurring internucleotide linkages do not hinder the transcription or replication of the vector.

The recombinant expression vector of the disclosures may be any suitable recombinant expression vector, and may be used to transform or transfect any suitable host. Suitable vectors include those designed for propagation and expansion or for expression or both, such as plasmids and viruses. The vector may be selected from the group consisting of the pUC series (Fermentas Life Sciences), the pBluescript series (Stratagene, LaJolla, CA), the pET series (Novagen, Madison, WI), the pGEX series (Pharmacia Biotech, Uppsala, Sweden), and the pEX series (Clontech, Palo Alto, CA). Bacteriophage vectors, such as λGTIO, λGTl 1, λZapII (Stratagene), λEMBL4, and λNMl 149, also may be used. Examples of plant expression vectors include pBIOl, pBI101.2, pBI101.3, pBI121 and pBIN19 (Clontech). Examples of animal expression vectors include pEUK-Cl, pMAM and pMAMneo (Clontech). Preferably, the recombinant expression vector is a viral vector, e.g., a retroviral vector.

The recombinant expression vectors of the disclosures may be prepared using standard recombinant DNA techniques described in, for example, Sambrook et al., supra, and Ausubel et al., supra. Constructs of expression vectors, which are circular or linear, may be prepared to contain a replication system functional in a prokaryotic or eukaryotic host cell. Replication systems may be derived, e.g., from CoIEl, 2μ plasmid, λ, SV40, bovine papilloma virus, and the like.

In exemplary aspects, the recombinant expression vector comprises regulatory sequences, such as transcription and translation initiation and termination codons, which are specific to the type of host (e.g., bacterium, fungus, plant, or animal) into which the vector is to be introduced, as appropriate and taking into consideration whether the vector is DNA- or RNA-based.

The recombinant expression vector may include one or more marker genes, which allow for selection of transformed or transfected hosts. Marker genes include biocide resistance, e.g., resistance to antibiotics, heavy metals, etc., complementation in an auxotrophic host to provide prototrophy, and the like. Suitable marker genes for the inventive expression vectors include, for instance, neomycin/G418 resistance genes, hygromycin resistance genes, histidinol resistance genes, tetracycline resistance genes, and ampicillin resistance genes.

The recombinant expression vector may comprise a native or normative promoter operably linked to the nucleotide sequence encoding the binding agent or conjugate or to the nucleotide sequence which is complementary to or which hybridizes to the nucleotide sequence encoding the binding agent or conjugate. The selection of promoters, e.g., strong, weak, inducible, tissue-specific and developmental-specific, is within the ordinary skill of the artisan.

Similarly, the combining of a nucleotide sequence with a promoter is also within the skill of the artisan. The promoter may be a non-viral promoter or a viral promoter, e.g., a cytomegalovirus (CMV) promoter, an SV40 promoter, an RSV promoter, and a promoter found in the long-terminal repeat of the murine stem cell virus.

The inventive recombinant expression vectors may be designed for either transient expression, for stable expression, or for both. Also, the recombinant expression vectors may be made for constitutive expression or for inducible expression. Further, the recombinant expression vectors may be made to include a suicide gene.

As used herein, the term “suicide gene” refers to a gene that causes the cell expressing the suicide gene to die. The suicide gene may be a gene that confers sensitivity to an agent, e.g., a drug, upon the cell in which the gene is expressed, and causes the cell to die when the cell is contacted with or exposed to the agent. Suicide genes are known in the art (see, for example, Suicide Gene Therapy: Methods and Reviews. Springer, Caroline J. (Maycer Research UK Centre for Maycer Therapeutics at the Institute of Maycer Research, Sutton, Surrey, UK), Humana Press, 2004) and include, for example, the Herpes Simplex Virus (HSV) thymidine kinase (TK) gene, cytosine deaminase, purine nucleoside phosphorylase, and nitroreductase.

The disclosures further provides a host cell comprising any of the nucleic acids or vectors described herein. As used herein, the term “host cell” refers to any type of cell that may contain the nucleic acid or vector described herein. In exemplary aspects, the host cell is a eukaryotic cell, e.g., plant, animal, fungi, or algae, or may be a prokaryotic cell, e.g., bacteria or protozoa. In exemplary aspects, the host cells is a cell originating or obtained from a subject, as described herein. In exemplary aspects, the host cell originates from or is obtained from a mammal. As used herein, the term “mammal” refers to any mammal, including, but not limited to, mammals of the order Rodentia, such as mice and hamsters, and mammals of the order Lagomorpha, such as rabbits. It is preferred that the mammals are from the order Carnivora, including Felines (cats) and Canines (dogs). It is more preferred that the mammals are from the order Artiodactyla, including bovines (cows) and swines (pigs) or of the order Perssodactyla, including equines (horses). It is most preferred that the mammals are of the order Primates, Ceboids, or Simoids (monkeys) or of the order Anthropoids (humans and apes). An especially preferred mammal is the human.

In exemplary aspects, the host cell is a cultured cell or a primary cell, i.e., isolated directly from an organism, e.g., a human. The host cell in exemplary aspects is an adherent cell or a suspended cell, i.e., a cell that grows in suspension. Suitable host cells are known in the art and include, for instance, DH5α E. coli cells, Chinese hamster ovarian (CHO) cells, monkey VERO cells, T293 cells, COS cells, HEK293 cells, and the like. For purposes of amplifying or replicating the recombinant expression vector, the host cell is preferably a prokaryotic cell, e.g., a DH5α cell. For purposes of producing a binding agent or a conjugate, the host cell is in some aspects a mammalian cell. In exemplary aspects, the host cell is a human cell. While the host cell may be of any cell type, the host cell may originate from any type of tissue, and may be of any developmental stage. In exemplary aspects, the host cell is a hematopoietic stem cell or progenitor cell. See, e.g, Nakamura De Oliveira et al., Human Gene Therapy 24:824-839 (2013). The host cell in exemplary aspects is a peripheral blood lymphocyte (PBL). In exemplary aspects, the host cell is a natural killer cell. In exemplary aspects, the host cell is a T cell.

For purposes herein, the T cell may be any T cell, such as a cultured T cell, e.g., a primary T cell, or a T cell from a cultured T cell line, e.g., Jurkat, SupTl, etc., or a T cell obtained from a mammal. If obtained from a mammal, the T cell may be obtained from numerous sources, including but not limited to blood, bone marrow, lymph node, the thymus, or other tissues or fluids. T cells may also be enriched for or purified. The T cell may be obtained by maturing hematopoietic stem cells, either in vitro or in vivo, into T cells. In exemplary aspects, the T cell is a human T cell. In exemplary aspects, the T cell is a T cell isolated from a human. The T cell may be any type of T cell, including NKT cell, and may be of any developmental stage, including but not limited to, CD4+/CD8+ double positive T cells, CDA+ helper T cells, e.g., Th1 and Th2 cells, CD8+ T cells (e.g., cytotoxic T cells), peripheral blood mononuclear cells (PBMCs), peripheral blood leukocytes (PBLs), tumor infiltrating cells (TILs), memory T cells, naive T cells, and the like. Preferably, the T cell is a CD8+ T cell or a CD4+ T cell.

Also provided by the disclosures is a population of cells comprising at least one host cell described herein. The population of cells may be a heterogeneous population comprising the host cell comprising any of the recombinant expression vectors described, in addition to at least one other cell, e.g., a host cell (e.g., a T cell), which does not comprise any of the recombinant expression vectors, or a cell other than a T cell, e.g., a B cell, a macrophage, a neutrophil, an erythrocyte, a hepatocyte, an endothelial cell, an epithelial cells, a muscle cell, a brain cell, etc. Alternatively, the population of cells may be a substantially homogeneous population, in which the population comprises mainly of host cells (e.g., consisting essentially of) comprising the recombinant expression vector. The population also may be a clonal population of cells, in which all cells of the population are clones of a single host cell comprising a recombinant expression vector, such that all cells of the population comprise the recombinant expression vector. In exemplary embodiments of the disclosures, the population of cells is a clonal population comprising host cells expressing a nucleic acid or a vector described herein.

Pharmaceutical Compositions and Routes of Administration

In some embodiments of the disclosures, the binding agents, conjugates, nucleic acids, vectors, host cells, or populations of cells, are admixed with a pharmaceutically acceptable carrier. Accordingly, pharmaceutical compositions comprising any of the binding agents, conjugates, nucleic acids, vectors, host cells, or populations of cells described herein and comprising a pharmaceutically acceptable carrier, diluent, or excipient are contemplated.

The pharmaceutically acceptable carrier is any of those conventionally used and is limited only by physico-chemical considerations, such as solubility and lack of reactivity with the active binding agent(s), and by the route of administration. The pharmaceutically acceptable carriers described herein, for example, vehicles, adjuvants, excipients, and diluents, are well-known to those skilled in the art and are readily available to the public. In one aspect the pharmaceutically acceptable carrier is one that is chemically inert to the active ingredient(s) of the pharmaceutical composition, e.g., the first binding agent and the second binding agent, and one which has no detrimental side effects or toxicity under the conditions of use. The carrier in some embodiments does not produce adverse, allergic, or other untoward reactions when administered to an animal or a human. The pharmaceutical composition in some aspects is free of pyrogens, as well as other impurities that could be harmful to humans or animals. Pharmaceutically acceptable carriers include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like; the use of which are well known in the art.

Acceptable carriers, excipients or stabilizers are nontoxic to recipients and are preferably inert at the dosages and concentrations employed, and include buffers such as phosphate, citrate, or other organic acids; antioxidants such as ascorbic acid; low molecular weight polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, arginine or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; salt-forming counterions such as sodium; and/or nonionic surfactants such as Tween, Pluronics or polyethylene glycol (PEG).

Therapeutic formulations of the compositions useful for practicing the methods disclosed herein, such as polypeptides, polynucleotides, or antibodies, may be prepared for storage by mixing the selected composition having the desired degree of purity with optional physiologically pharmaceutically-acceptable carriers, excipients, or stabilizers (Remington's Pharmaceutical Sciences, 18th edition, A. R. Gennaro, ed., Mack Publishing Company (1990)) in the form of a lyophilized cake or an aqueous solution. Pharmaceutical compositions may be produced by admixing with one or more suitable carriers or adjuvants such as water, mineral oil, polyethylene glycol, starch, talcum, lactose, thickeners, stabilizers, suspending agents, and the like. Such compositions may be in the form of solutions, suspensions, tablets, capsules, creams, salves, ointments, or other conventional forms.

The composition to be used for in vivo administration should be sterile. This is readily accomplished by filtration through sterile filtration membranes, prior to or following lyophilization and reconstitution. Therapeutic compositions generally are placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle. The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In some cases the form should be sterile and should be fluid to the extent that easy syringability exists. It should be stable under the conditions of manufacture and storage and should be preserved against the contaminating action of microorganisms, such as bacteria and fungi. The composition for parenteral administration ordinarily will be stored in lyophilized form or in solution.

The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils. The proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion, and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and/or antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the inclusion in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.

The choice of carrier will be determined in part by the particular type of binding agents of the pharmaceutical composition, as well as by the particular route used to administer the pharmaceutical composition. Accordingly, there are a variety of suitable formulations of the pharmaceutical composition.

The pharmaceutical composition of the present disclosures can comprise any pharmaceutically acceptable ingredient including, for example, acidifying agents, additives, adsorbents, aerosol propellants, air displacement agents, alkalizing agents, anticaking agents, anticoagulants, antimicrobial preservatives, antioxidants, antiseptics, bases, binders, buffering agents, chelating agents, coating agents, coloring agents, desiccants, detergents, diluents, disinfectants, disintegrants, dispersing agents, dissolution-enhancing agents, dyes, emollients, emulsifying agents, emulsion stabilizers, fillers, film-forming agents, flavor enhancers, flavoring agents, flow enhancers, gelling agents, granulating agents, humectants, lubricants, mucoadhesives, ointment bases, ointments, oleaginous vehicles, organic bases, pastille bases, pigments, plasticizers, polishing agents, preservatives, sequestering agents, skin penetrants, solubilizing agents, solvents, stabilizing agents, suppository bases, surface active agents, surfactants, suspending agents, sweetening agents, therapeutic agents, thickening agents, tonicity agents, toxicity agents, viscosity-increasing agents, water-absorbing agents, water-miscible cosolvents, water softeners, or wetting agents.

The pharmaceutical compositions may be formulated to achieve a physiologically compatible pH. In some embodiments, the pH of the pharmaceutical composition may be at least 5, at least 5.5, at least 6, at least 6.5, at least 7, at least 7.5, at least 8, at least 8.5, at least 9, at least 9.5, at least 10, or at least 10.5 up to and including pH 11, depending on the formulation and route of administration. In certain embodiments, the pharmaceutical compositions may comprise buffering agents to achieve a physiologically compatible pH. The buffering agents may include any compounds capable of buffering at the desired pH such as, for example, phosphate buffers (e.g., PBS), triethanolamine, Tris, bicine, TAPS, tricine, HEPES, TES, MOPS, PIPES, cacodylate, MES, and others known in the art.

In some embodiments, the pharmaceutical composition comprising the binding agents described herein is formulated for parenteral administration, subcutaneous administration, intravenous administration, intramuscular administration, intraarterial administration, intrathecal administration, or interperitoneal administration. In other embodiments, the pharmaceutical composition is administered via nasal, spray, oral, aerosol, rectal, or vaginal administration. The compositions may be administered by infusion, bolus injection or by implantation device.

The following discussion on routes of administration is merely provided to illustrate exemplary embodiments and should not be construed as limiting the scope of the disclosed subject matter in any way.

Formulations suitable for oral administration can consist of (a) liquid solutions, such as an effective amount of the composition of the present disclosure dissolved in diluents, such as water, saline, or orange juice; (b) capsules, sachets, tablets, lozenges, and troches, each containing a predetermined amount of the active ingredient, as solids or granules; (c) powders; (d) suspensions in an appropriate liquid; and (e) suitable emulsions. Liquid formulations may include diluents, such as water and alcohols, for example, ethanol, benzyl alcohol, and the polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant. Capsule forms can be of the ordinary hard- or soft-shelled gelatin type containing, for example, surfactants, lubricants, and inert fillers, such as lactose, sucrose, calcium phosphate, and corn starch. Tablet forms can include one or more of lactose, sucrose, mannitol, corn starch, potato starch, alginic acid, microcrystalline cellulose, acacia, gelatin, guar gum, colloidal silicon dioxide, croscarmellose sodium, talc, magnesium stearate, calcium stearate, zinc stearate, stearic acid, and other excipients, colorants, diluents, buffering agents, disintegrating agents, moistening agents, preservatives, flavoring agents, and other pharmacologically compatible excipients. Lozenge forms can comprise a composition of the disclosure in a flavor, usually sucrose and acacia or tragacanth, as well as pastilles comprising a composition of the disclosure in an inert base, such as gelatin and glycerin, or sucrose and acacia, emulsions, gels, and the like, optionally also containing such excipients as are known in the art.

The compositions of the disclosure, alone or in combination with other suitable components, can be delivered via pulmonary administration and can be made into aerosol formulations to be administered via inhalation. These aerosol formulations can be placed into pressurized acceptable propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like. They also may be formulated as pharmaceuticals for non-pressured preparations, such as in a nebulizer or an atomizer. Such spray formulations also may be used to spray mucosa. In some embodiments, the composition is formulated into a powder blend or into microparticles or nanoparticles. Suitable pulmonary formulations are known in the art. See, e.g., Qian et al., Int J Pharm 366: 218-220 (2009); Adjei and Garren, Pharmaceutical Research, 7(6): 565-569 (1990); Kawashima et al., J Controlled Release 62(1-2): 279-287 (1999); Liu et al., Pharm Res 10(2): 228-232 (1993); International Patent Application Publication Nos. WO 2007/133747 and WO 2007/141411.

Topical formulations are well-known to those of skill in the art. Such formulations are particularly suitable in the context of the invention for application to the skin.

In some embodiments, the pharmaceutical composition described herein is formulated for parenteral administration. For purposes herein, parenteral administration includes, but is not limited to, intravenous, intraarterial, intramuscular, intracerebral, intracerebroventricular, intracardiac, subcutaneous, intraosseous, intradermal, intrathecal, intraperitoneal, retrobulbar, intrapulmonary, intravesical, and intracavernosal injections or infusions. Administration by surgical implantation at a particular site is contemplated as well.

Formulations suitable for parenteral administration include aqueous and non-aqueous, isotonic sterile injection solutions, which can contain anti-oxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives. The term, “parenteral” means not through the alimentary canal but by some other route such as subcutaneous, intramuscular, intraspinal, or intravenous. The composition of the present disclosure can be administered with a physiologically acceptable diluent in a pharmaceutical carrier, such as a sterile liquid or mixture of liquids, including water, saline, aqueous dextrose and related sugar solutions, an alcohol, such as ethanol or hexadecyl alcohol, a glycol, such as propylene glycol or polyethylene glycol, dimethylsulfoxide, glycerol, ketals such as 2,2-dimethyl-1,5,3-dioxolane-4-methanol, ethers, poly(ethyleneglycol) 400, oils, fatty acids, fatty acid esters or glycerides, or acetylated fatty acid glycerides with or without the addition of a pharmaceutically acceptable surfactant, such as a soap or a detergent, a suspending agent, such as pectin, carbomers, methylcellulose, hydroxypropylmethylcellulose, or carboxymethylcellulose, or emulsifying agents and other pharmaceutical adjuvants.

Oils, which can be used in parenteral formulations include petroleum, animal, vegetable, or synthetic oils. Specific examples of oils include peanut, soybean, sesame, cottonseed, corn, olive, petrolatum, and mineral. Suitable fatty acids for use in parenteral formulations include oleic acid, stearic acid, and isostearic acid. Ethyl oleate and isopropyl myristate are examples of suitable fatty acid esters.

The parenteral formulations in some embodiments contain preservatives or buffers. In order to minimize or eliminate irritation at the site of injection, such compositions optionally contain one or more nonionic surfactants having a hydrophile-lipophile balance (HLB) of from about 12 to about 17. The quantity of surfactant in such formulations will typically range from about 5% to about 15% by weight. Suitable surfactants include polyethylene glycol sorbitan fatty acid esters, such as sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed by the condensation of propylene oxide with propylene glycol. The parenteral formulations can be presented in unit-dose or multi-dose sealed containers, such as ampoules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid excipient, for example, water, for injections, immediately prior to use. Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules, and tablets of the kind previously described and known in the art.

Injectable formulations are in accordance with the invention. The requirements for effective pharmaceutical carriers for injectable compositions are well-known to those of ordinary skill in the art (see, e.g., Pharmaceutics and Pharmacy Practice, J. B. Lippincott Company, Philadelphia, PA, Banker and Chalmers, eds., pages 238-250 (1982), and ASHP Handbook on Injectable Drugs, Toissel, 4th ed., pages 622-630 (1986)).

It will be appreciated by one of skill in the art that, in addition to the above-described pharmaceutical compositions, the composition of the disclosure can be formulated as inclusion complexes, such as cyclodextrin inclusion complexes, or liposomes.

Dose

For purposes herein, the amount or dose of the pharmaceutical composition administered is sufficient to effect, e.g., a therapeutic or prophylactic response, in the subject or animal over a reasonable time frame. For example, the dose of the pharmaceutical composition is sufficient to treat or prevent a disease or medical condition in a period of from about 12 hours, about 18 hours, about 1 to 4 days or longer, e.g., 5 days, 6 days, 1 week, 10 days, 2 weeks, 16 to 20 days, or more, from the time of administration. In certain embodiments, the time period is even longer. The dose is determined by the efficacy of the particular pharmaceutical composition and the condition of the animal (e.g., human), as well as the body weight of the animal (e.g., human) to be treated.

Many assays for determining an administered dose are known in the art. In some embodiments, an assay which comprises comparing the extent to which the binding agents block IL13Rα2-mediated cell growth upon administration of a given dose to a mammal among a set of mammals each of which is given a different dose of binding agents is used to determine a starting dose to be administered to a mammal. The extent to which the binding agents block IL13Rα2 mediated cell growth upon administration of a certain dose can be assayed by methods known in the art.

The dose of the pharmaceutical composition also will be determined by the existence, nature and extent of any adverse side effects that might accompany the administration of a particular pharmaceutical composition. Typically, the attending physician will decide the dosage of the pharmaceutical composition with which to treat each individual patient, taking into consideration a variety of factors, such as age, body weight, general health, diet, sex, binding agents of the pharmaceutical composition to be administered, route of administration, and the severity of the condition being treated.

By way of example and not intending to limit the invention, the dose of the binding agent of the present disclosure can be about 0.0001 to about 1 g/kg body weight of the subject being treated/day, from about 0.0001 to about 0.001 g/kg body weight/day, or about 0.01 mg to about 1 g/kg body weight/day. The pharmaceutical composition in some aspects comprise the binding agent of the present disclosure at a concentration of at least A, wherein A is about 0.001 mg/ml, about 0.01 mg/ml, 0 about 1 mg/ml, about 0.5 mg/ml, about 1 mg/ml, about 2 mg/ml, about 3 mg/ml, about 4 mg/ml, about 5 mg/ml, about 6 mg/ml, about 7 mg/ml, about 8 mg/ml, about 9 mg/ml, about 10 mg/ml, about 11 mg/ml, about 12 mg/ml, about 13 mg/ml, about 14 mg/ml, about 15 mg/ml, about 16 mg/ml, about 17 mg/ml, about 18 mg/ml, about 19 mg/ml, about 20 mg/ml, about 21 mg/ml, about 22 mg/ml, about 23 mg/ml, about 24 mg/ml, about 25 mg/ml or higher. In some embodiments, the pharmaceutical composition comprises the binding agent at a concentration of at most B, wherein B is about 30 mg/ml, about 25 mg/ml, about 24 mg/ml, about 23, mg/ml, about 22 mg/ml, about 21 mg/ml, about 20 mg/ml, about 19 mg/ml, about 18 mg/ml, about 17 mg/ml, about 16 mg/ml, about 15 mg/ml, about 14 mg/ml, about 13 mg/ml, about 12 mg/ml, about 11 mg/ml, about 10 mg/ml, about 9 mg/ml, about 8 mg/ml, about 7 mg/ml, about 6 mg/ml, about 5 mg/ml, about 4 mg/ml, about 3 mg/ml, about 2 mg/ml, about 1 mg/ml, or about 0.1 mg/ml. In some embodiments, the compositions may contain an analog at a concentration range of A to B mg/ml, for example, about 0.001 to about 30.0 mg/ml.

Additional dosing guidance can be gauged from other antibody therapeutics, such as bevacizumab (Avastin™ Genentech); Cetuximab (Exbitux™ Imclone), Panitumumab (Vectibix™ Amgen), and Trastuzumab (Herceptin™ Genentech).

Timing of Administration

The disclosed pharmaceutical formulations may be administered according to any regimen including, for example, daily (1 time per day, 2 times per day, 3 times per day, 4 times per day, 5 times per day, 6 times per day), every two days, every three days, every four days, every five days, every six days, weekly, bi-weekly, every three weeks, monthly, or bi-monthly. Timing, like dosing can be fine-tuned based on dose-response studies, efficacy, and toxicity data, and initially gauged based on timing used for other antibody therapeutics.

Controlled Release Formulations

The pharmaceutical composition is in certain aspects modified into a depot form, such that the manner in which the active ingredients of the pharmaceutical composition (e.g., the binding agents) is released into the body to which it is administered is controlled with respect to time and location within the body (see, for example, U.S. Pat. No. 4,450,150). Depot forms in various aspects, include, for example, an implantable composition comprising a porous or non-porous material, such as a polymer, wherein the binding agents are encapsulated by or diffused throughout the material and/or degradation of the non-porous material. The depot is then implanted into the desired location within the body and the binding agents are released from the implant at a predetermined rate.

Accordingly, the pharmaceutical composition in certain aspects is modified to have any type of in vivo release profile. In some aspects, the pharmaceutical composition is an immediate release, controlled release, sustained release, extended release, delayed release, or bi-phasic release formulation. Methods of formulating peptides (e.g., peptide binding agents) for controlled release are known in the art. See, for example, Qian et al., J Pharm 374: 46-52 (2009) and International Patent Application Publication Nos. WO 2008/130158, WO2004/033036; WO2000/032218; and WO 1999/040942. Suitable examples of sustained-release preparations include semipermeable polymer matrices in the form of shaped articles, e.g., films, or microcapsules. Sustained release matrices include polyesters, hydrogels, polylactides (U.S. Pat. No. 3,773,919, EP 58,481), copolymers of L-glutamic acid and gamma ethyl-L-glutamate (Sidman, et al., Biopolymers, 22: 547-556 (1983)), poly (2-hydroxyethyl-methacrylate) (Langer, et al., J. Biomed. Mater. Res., 15:167-277 (1981) and Langer, Chem. Tech., 12:98-105 (1982)), ethylene vinyl acetate (Langer, et al, supra) or poly-D(−)-3-hydroxybutyric acid (EP 133,988). Sustained-release compositions also may include liposomes, which can be prepared by any of several methods known in the art (e.g., DE 3,218,121; Epstein, et al., Proc. Natl. Acad. Sci. USA, 82:3688-3692 (1985); Hwang, et al., Proc. Natl. Acad. Sci. USA, 77:4030-4034 (1980); EP 52,322; EP 36,676; EP 88,046; EP 143,949).

Combinations

The compositions of the disclosures may be employed alone, or in combination with other agents. In some embodiments, more than one type of binding agent are administered. For example, the administered composition, e.g., pharmaceutical composition, may comprise an antibody as well as an scFv. In some embodiments, the compositions of the disclosure are administered together with another therapeutic agent or diagnostic agent, including any of those described herein. Certain diseases, e.g., cancers, or patients may lend themselves to a treatment of combined agents to achieve an additive or even a synergistic effect compared to the use of any one therapy alone.

Uses

Based in part on the data provided herein, the binding agents, conjugates, host cells, populations of cells, and pharmaceutical compositions are useful for treating a neoplasm, tumor, or a cancer.

For purposes of the present disclosure, the term “treat” and “prevent” as well as words stemming therefrom, as used herein, do not necessarily imply 100% or complete treatment (e.g., cure) or prevention. Rather, there are varying degrees of treatment or prevention of which one of ordinary skill hi the art recognizes as having a potential benefit or therapeutic effect. In this respect, the methods of the present disclosures can provide any amount or any level of treatment or prevention of a cancer in a patient, e.g., a human. Furthermore, the treatment or prevention provided by the method disclosed herein can include treatment or prevention of one or more conditions or symptoms of the disease, e.g., cancer, being treated or prevented. Also, for purposes herein, “prevention” can encompass delaying the onset of the disease, or a symptom or condition thereof.

The materials and methods described herein are especially useful for inhibiting neoplastic cell growth or spread; particularly neoplastic cell growth for which the IL13Rα2 targeted by the binding agents plays a role.

Neoplasms treatable by the binding agents, conjugates, host cells, populations of cells, and pharmaceutical compositions of the disclosures include solid tumors, for example, carcinomas and sarcomas. Carcinomas include malignant neoplasms derived from epithelial cells which infiltrate, for example, invade, surrounding tissues and give rise to metastases. Adenocarcinomas are carcinomas derived from glandular tissue, or from tissues that form recognizable glandular structures. Another broad category of cancers includes sarcomas and fibrosarcomas, which are tumors whose cells are embedded in a fibrillar or homogeneous substance, such as embryonic connective tissue. The invention also provides methods of treatment of cancers of myeloid or lymphoid systems, including leukemias, lymphomas, and other cancers that typically are not present as a tumor mass, but are distributed in the vascular or lymphoreticular systems. Further contemplated are methods for treatment of adult and pediatric oncology, growth of solid tumors/malignancies, myxoid and round cell carcinoma, locally advanced tumors, cancer metastases, including lymphatic metastases. The cancers listed herein are not intended to be limiting. Both age (child and adult), sex (male and female), primary and secondary, pre- and post-metastatic, acute and chronic, benign and malignant, anatomical location cancer embodiments and variations are contemplated targets. Cancers are grouped by embryonic origin (e.g., carcinoma, lymphomas, and sarcomas), by organ or physiological system, and by miscellaneous grouping. Particular cancers may overlap in their classification, and their listing in one group does not exclude them from another.

Carcinomas that may be targeted include adrenocortical, acinar, acinic cell, acinous, adenocystic, adenoid cystic, adenoid squamous cell, cancer adenomatosum, adenosquamous, adnexel, cancer of adrenal cortex, adrenocortical, aldosterone-producing, aldosterone-secreting, alveolar, alveolar cell, ameloblastic, ampullary, anaplastic cancer of thyroid gland, apocrine, basal cell, basal cell, alveolar, comedo basal cell, cystic basal cell, morphea-like basal cell, multicentric basal cell, nodulo-ulcerative basal cell, pigmented basal cell, sclerosing basal cell, superficial basal cell, basaloid, basosquamous cell, bile duct, extrahepatic bile duct, intrahepatic bile duct, bronchioalveolar, bronchiolar, bronchioloalveolar, bronchoalveolar, bronchoalveolar cell, bronchogenic, cerebriform, cholangiocellular, chorionic, choroids plexus, clear cell, cloacogenic anal, colloid, comedo, corpus, cancer of corpus uteri, cortisol-producing, cribriform, cylindrical, cylindrical cell, duct, ductal, ductal cancer of the prostate, ductal cancer in situ (DCIS), eccrine, embryonal, cancer en cuirasse, endometrial, cancer of endometrium, endometroid, epidermoid, cancer ex mixed tumor, cancer ex pleomorphic adenoma, exophytic, fibrolamellar, cancer fibrosum, follicular cancer of thyroid gland, gastric, gelatinform, gelatinous, giant cell, giant cell cancer of thyroid gland, cancer gigantocellulare, glandular, granulose cell, hepatocellular, Hurthle cell, hypernephroid, infantile embryonal, islet cell carcinoma, inflammatory cancer of the breast, cancer in situ, intraductal, intraepidermal, intraepithelial, juvenile embryonal, Kulchitsky-cell, large cell, leptomeningeal, lobular, infiltrating lobular, invasive lobular, lobular cancer in situ (LCIS), lymphoepithelial, cancer medullare, medullary, medullary cancer of thyroid gland, medullary thyroid, melanotic, meningeal, Merkel cell, metatypical cell, micropapillary, cancer molle, mucinous, cancer muciparum, cancer mucocellulare, mucoepidermoid, cancer mucosum, mucous, nasopharyngeal, neuroendocrine cancer of the skin, noninfiltrating, non-small cell, non-small cell lung cancer (NSCLC), oat cell, cancer ossificans, osteoid, Paget's disease of the bone or breast, papillary, papillary cancer of thyroid gland, periampullary, preinvasive, prickle cell, primary intrasseous, renal cell, scar, schistosomal bladder, Schneiderian, scirrhous, sebaceous, signet-ring cell, cancer simplex, small cell, small cell lung cancer (SCLC), spindle cell, cancer spongiosum, squamous, squamous cell, terminal duct, anaplastic thyroid, follicular thyroid, medullary thyroid, papillary thyroid, trabecular cancer of the skin, transitional cell, tubular, undifferentiated cancer of thyroid gland, uterine corpus, verrucous, villous, cancer villosum, yolk sac, squamous cell particularly of the head and neck, esophageal squamous cell, and oral cancers and carcinomas.

Sarcomas that may be targeted include adipose, alveolar soft part, ameloblastic, avian, botryoid, sarcoma botryoides, chicken, chloromatous, chondroblastic, clear cell sarcoma of kidney, embryonal, endometrial stromal, epithelioid, Ewing's, fascial, fibroblastic, fowl, giant cell, granulocytic, hemangioendothelial, Hodgkin's, idiopathic multiple pigmented hemorrhagic, immunoblastic sarcoma of B cells, immunoblastic sarcoma of T cells, Jensen's, Kaposi's, Kupffer cell, leukocytic, lymphatic, melanotic, mixed cell, multiple, lymphangio, idiopathic hemorrhagic, multipotential primary sarcoma of bone, osteoblastic, osteogenic, parosteal, polymorphous, pseudo-Kaposi, reticulum cell, reticulum cell sarcoma of the brain, rhabdomyosarcoma, Rous, soft tissue, spindle cell, synovial, telangiectatic, sarcoma (osteosarcoma)/malignant fibrous histiocytoma of bone, and soft tissue sarcomas.

Lymphomas that may targeted include AIDS-related, non-Hodgkin's, Hodgkin's, T-cell, T-cell leukemia/lymphoma, African, B-cell, B-cell monocytoid, bovine malignant, Burkitt's, centrocytic, lymphoma cutis, diffuse, diffuse, large cell, diffuse, mixed small and large cell, diffuse, small cleaved cell, follicular, follicular center cell, follicular, mixed small cleaved and large cell, follicular, predominantly large cell, follicular, predominantly small cleaved cell, giant follicle, giant follicular, granulomatous, histiocytic, large cell, immunoblastic, large cleaved cell, large noncleaved cell, Lennert's, lymphoblastic, lymphocytic, intermediate; lymphocytic, intermediately differentiated, plasmacytoid; poorly differentiated lymphocytic, small lymphocytic, well differentiated lymphocytic, lymphoma of cattle; MALT, mantle cell, mantle zone, marginal zone, Mediterranean lymphoma mixed lymphocytic-histiocytic, nodular, plasmacytoid, pleomorphic, primary central nervous system, primary effusion, small b-cell, small cleaved cell, small noncleaved cell, T-cell lymphomas; convoluted T-cell, cutaneous t-cell, small lymphocytic T-cell, undefined lymphoma, u-cell, undifferentiated, aids-related, central nervous system, cutaneous T-cell, effusion (body cavity-based), thymic lymphoma, and cutaneous T cell lymphomas.

Leukemias and other blood cell malignancies that may be targeted include acute lymphoblastic, acute myeloid, lymphocytic, chronic myelogenous, hairy cell, lymphoblastic, myeloid, lymphocytic, myelogenous, leukemia, hairy cell, T-cell, monocytic, myeloblastic, granulocytic, gross, hand mirror-cell, basophilic, hemoblastic, histiocytic, leukopenic, lymphatic, Schilling's, stem cell, myelomonocytic, prolymphocytic, micromyeloblastic, megakaryoblastic, megakaryocytic, Rieder cell, bovine, aleukemic, mast cell, myelocytic, plasma cell, subleukemic, multiple myeloma, nonlymphocytic, and chronic myelocytic leukemias.

Brain and central nervous system (CNS) cancers and tumors that may be targeted include astrocytomas (including cerebellar and cerebral), gliomas (including malignant gliomas, glioblastomas, brain stem gliomas, visual pathway and hypothalamic gliomas), brain tumors, ependymoma, medulloblastoma, supratentorial primitive neuroectodermal tumors, primary central nervous system lymphoma, extracranial germ cell tumor, myelodysplastic syndromes, oligodendroglioma, myelodysplastic/myeloproliferative diseases, myelogenous leukemia, myeloid leukemia, multiple myeloma, myeloproliferative disorders, neuroblastoma, plasma cell neoplasm/multiple myeloma, central nervous system lymphoma, intrinsic brain tumors, astrocytic brain tumors, and metastatic tumor cell invasion in the central nervous system.

Gastrointestinal cancers that may be targeted include extrahepatic bile duct cancer, colon cancer, colon and rectum cancer, colorectal cancer, gallbladder cancer, gastric (stomach) cancer, gastrointestinal carcinoid tumor, gastrointestinal carcinoid tumors, gastrointestinal stromal tumors, bladder cancers, islet cell carcinoma (endocrine pancreas), pancreatic cancer, islet cell pancreatic cancer, prostate cancer rectal cancer, salivary gland cancer, small intestine cancer, colon cancer, and polyps associated with colorectal neoplasia. A discussion of colorectal cancer is described in Barderas et al., Cancer Research 72: 2780-2790 (2012).

Bone cancers that may be targeted include osteosarcoma and malignant fibrous histiocytomas, bone marrow cancers, bone metastases, osteosarcoma/malignant fibrous histiocytoma of bone, and osteomas and osteosarcomas. Breast cancers that may be targeted include small cell carcinoma and ductal carcinoma.

Lung and respiratory cancers that may be targeted include bronchial adenomas/carcinoids, esophagus cancer esophageal cancer, esophageal cancer, hypopharyngeal cancer, laryngeal cancer, hypopharyngeal cancer, lung carcinoid tumor, non-small cell lung cancer, small cell lung cancer, small cell carcinoma of the lungs, mesothelioma, nasal cavity and paranasal sinus cancer, nasopharyngeal cancer, nasopharyngeal cancer, oral cancer, oral cavity and lip cancer, oropharyngeal cancer; paranasal sinus and nasal cavity cancer, and pleuropulmonary blastoma.

Urinary tract and reproductive cancers that may be targeted include cervical cancer, endometrial cancer, ovarian epithelial cancer, extragonadal germ cell tumor, extracranial germ cell tumor, extragonadal germ cell tumor, ovarian germ cell tumor, gestational trophoblastic tumor, spleen, kidney cancer, ovarian cancer, ovarian epithelial cancer, ovarian germ cell tumor, ovarian low malignant potential tumor, penile cancer, renal cell cancer (including carcinomas), renal cell cancer, renal pelvis and ureter (transitional cell cancer), transitional cell cancer of the renal pelvis, and ureter, gestational trophoblastic tumor, testicular cancer, ureter and renal pelvis, transitional cell cancer, urethral cancer, endometrial uterine cancer, uterine sarcoma, vaginal cancer, vulvar cancer, ovarian carcinoma, primary peritoneal epithelial neoplasms, cervical carcinoma, uterine cancer and solid tumors in the ovarian follicle), superficial bladder tumors, invasive transitional cell carcinoma of the bladder, and muscle-invasive bladder cancer.

Skin cancers and melanomas (as well as non-melanomas) that may be targeted include cutaneous t-cell lymphoma, intraocular melanoma, tumor progression of human skin keratinocytes, basal cell carcinoma, and squamous cell cancer. Liver cancers that may be targeted include extrahepatic bile duct cancer, and hepatocellular cancers. Eye cancers that may be targeted include intraocular melanoma, retinoblastoma, and intraocular melanoma Hormonal cancers that may be targeted include: parathyroid cancer, pineal and supratentorial primitive neuroectodermal tumors, pituitary tumor, thymoma and thymic carcinoma, thymoma, thymus cancer, thyroid cancer, cancer of the adrenal cortex, and ACTH-producing tumors.

Miscellaneous other cancers that may be targeted include advanced cancers, AIDS-related, anal cancer, adrenal, cortical, aplastic anemia, aniline, betel or buyo cheek, cerebriform, chimney-sweeps, clay pipe, colloid, contact, cystic, dendritic, cancer a deux, duct, dye workers, encephaloid, cancer en cuirasse, endometrial, endothelial, epithelial, glandular, cancer in situ, kang, kangri, latent, medullary, melanotic, mule-spinners', non-small cell lung, occult cancer, paraffin, pitch workers', scar, schistosomal bladder, scirrhous, lymph node, small cell lung, soft, soot, spindle cell, swamp, tar, and tubular cancers.

Miscellaneous other cancers that may be targeted also include carcinoid (gastrointestinal and bronchial) Castleman's disease chronic myeloproliferative disorders, clear cell sarcoma of tendon sheaths, Ewing's family of tumors, head and neck cancer, lip and oral cavity cancer, Waldenstrom's macroglobulinemia, metastatic squamous neck cancer with occult primary, multiple endocrine neoplasia syndrome, multiple myeloma/plasma cell neoplasm, Wilms' tumor, mycosis fungoides, pheochromocytoma, sezary syndrome, supratentorial primitive neuroectodermal tumors, unknown primary site, peritoneal effusion, malignant pleural effusion, trophoblastic neo-plasms, and hemangiopericytoma.

In exemplary aspects, the cancer is any one of the foregoing described in which IL13Rα2 is expressed on the cells of the cancer. In exemplary aspects, the cancer is colon cancer. In exemplary aspects, the cancer is Glioblastoma Multiforme. In exemplary aspects, the method of treating cancer in a subject in need thereof comprises administering to the subject any of the binding agents, conjugates, nucleic acids, vectors, host cells, cell populations, or pharmaceutical compositions described herein, in an amount effective to treat the cancer. In exemplary aspects, the method comprises administering a conjugate described herein. In exemplary aspects, the method comprises administering host cells of the disclosures and the host cells are autologous cells in relation to the subject being treated. In exemplary aspects, the method comprises administering host cells of the disclosures and the host cells are cells obtained from the subject being treated. In exemplary aspects, the cells are T-lymphocytes. In alternative aspects, the cells are natural killer cells.

The disclosure will be more fully understood by reference to the following examples, which detail exemplary embodiments of the disclosure. The examples should not, however, be construed as limiting the scope of the disclosure.

Example 1

Materials

Lipofectamine 2000 and the pEF6/Myc-His vector were obtained from Invitrogen. Monoclonal antibodies to IL13Rα2 (clones YY-23Z and B-D13) and the IsoStrip mouse monoclonal antibody isotyping kit were purchased from Santa Cruz Biotechnology (Santa Cruz, CA). The mAb to IL13Rα2 (clone 83807) and recombinant human and mouse IL13Rα2hFc and IL13Rα1hFc chimeras were purchased from R&D Systems (Minneapolis, MN). Biotinylated horse anti-mouse antibodies and the Elite kit were obtained from Vector Laboratories (Burlingame, CA). 3,3′-Diaminobenzidine substrate was purchased from Dako (Carpinteria, CA). Goat anti-mouse antibody conjugated with peroxidase was purchased from Chemicon International (Temicula, CA), and Pngase F was purchased from New England Biolabs (Ipswich, MA). The QuikChange Lightning™ site-directed mutagenesis kit was purchased from Agilent Technologies, Inc. (Santa Clara, CA), and the RNeasy Plus™ kit was received from Qiagen (Valencia, CA). The cDNA iScript™ kit, 7.5% Tris-HCl gel, and ImmunStar™ WesternC™ developing reagent and protein marker were purchased from Bio-Rad. The human IL-13 ELISA kit was purchased from eBioscience (San Diego, CA). GBM12 and GBM43 were kindly provided by Dr. David C. James (University of California-San Francisco), and the cDNA encoding human wild-type IL13Rα2 was obtained from Dr. Waldemar Debinski (Wake Forest University). Obtaining the cDNA encoding the human wild-type IL13Rα2 or most other proteins involves the use of well-known techniques and readily available reagents.

Cell Lines

U373 (GBM), 293T (human embryonic kidney), and Raji (Burkitt's lymphoma) cell line were purchased from the American Type Culture Collection (ATCC; Manassas, VA). The generation of U373 cells expressing enhanced green fluorescent protein and firefly luciferase (U373.eGFP.ffLuc), 293T cells expressing green fluorescent protein (293T.GFP) or IL13Rα2 and GFP (293T.IL13Rα2.GFP) were previously reported. See Chow et al., Mol. Ther. 21:629-637 (2013); Krebs et al., Cytotherapy 16:1121-1131 (2014). Cell lines were grown in RPMI or DMEM (Thermo Scientific HyClone, Waltham, MA; Lonza, Basel, Switzerland) with 10% fetal calf serum (FCS; HyClone, Logan, UT) and 2 mM GlutaMAX-I™ (Invitrogen, Carlsbad, CA). The Characterized Cell Line Core Facility at MD Anderson Cancer Center, Houston, Texas, performed cell line validation.

Immunization

To obtain monoclonal antibodies with specificity to native IL13Rα2, the human recombinant IL13Rα2hFc fusion was used for immunization of animals and in all screening assays. Two 6-week-old female BALB/c mice were immunized with intraperitoneal injection of 10 μg of rhIL13Rα2hFc protein in complete Freund's adjuvant followed by intraperitoneal injection of 10 μg of rhIL13Rα2hFc protein in incomplete Freund's adjuvant at a 2-week interval for 2 months. Two weeks after the last intraperitoneal injection and 3 days before the fusion, a boost was performed by the combination of intravenous and intraperitoneal injection of 10 μg of antigen without Freund's adjuvant. The fusion of mouse spleen cells with the mouse myeloma cell line X63.Ag8.653 subclone P3O1 was performed by using a procedure described by Köhler and Milstein (27). Hybridoma supernatants were assayed for the presence of IL13Rα2 antibodies using an enzyme-linked immunosorbent assay (ELISA). Selected populations were cloned, and supernatants were assayed to identify the clones with strongest binding.

Generation of CHO Cell Line Expressing Human IL13Rα2

The cDNA encoding human wild-type IL13Rα2 was amplified with the following primer pair: forward, 5′-GCTTGGTACCGAATGGCTTTCGTTTGCTTGGC-3′ (SEQ ID NO: 17) and reverse, 5′-GTTTTTGTTCGAATGTATCACAGAAAAATTCTGG-3′ (SEQ ID NO: 18). The purified PCR product was restricted with KpnI and BstBI enzymes, agarose gel-purified, and subsequently cloned into the pEF6/Myc-His vector in a reading frame with Myc and His6 tags. CHO cells were plated at 80% confluence and transfected with a plasmid encoding the IL13Rα2 using Lipofectamine 2000. The following day, 4 μg/ml blasticidin was added for selection of cells that had stably incorporated and expressed the IL13Rα2 transcript. A stable population of cells was further subcloned in 96-well plates at a density of one cell/well. Ten days later, single clones were screened by flow cytometry for cell surface expression of IL13Rα2 using an antibody to IL13Rα2 (clone B-D13). The clone with the highest level of IL13Rα2 expression was selected and expanded for subsequent screening of hybridomas secreting IL13Rα2 antibodies.

ELISA

96-well plates were coated with 50 μl of human or mouse recombinant IL13Rα2hFc or IL13Rα1hFc or human control IgG at a concentration of 1 μg/ml overnight at 4° C. Following washes with TBS-Tween 20 buffer and blocking with 1% nonfat dry milk, 50 μl of purified antibodies, serum, or hybridoma supernatants at various dilutions were applied to the plate and incubated for 1 hour at room temperature. Bound antibodies were detected with goat anti-mouse antibodies conjugated to alkaline phosphatase following the development with alkaline phosphatase substrate. Plates were read at A405 using a UniRead 800 plate reader (BioTek).

Flow Cytometry

CHO or HEK cells expressing IL13Rα2; the glioma cell lines A172, N10, U251, U87, and U118; patient-derived GBM12 and GBM43, and primary human astrocytes were stained with IL13Rα2 (clone 47) monoclonal antibody at 1 μg/ml followed by goat anti-mouse Alexa Fluor 647 (1:500). All staining procedures were performed on ice. Samples were analyzed using the BD FACSCanto flow cytometer and FACSDiVa™ software.

For the experiments disclosed in Examples 13-16, a FACSCalibur instrument (BD Bioscience, Mountain View, CA) was used to acquire immunofluorescence data that were analyzed with CellQuest (BD) or FCS Express software (De Novo Software, Los Angeles, CA). Isotype controls were immunoglobulin Gi-fluorescein isothiocyanate (IgG1-FITC; BD Bioscience) and IgG1-phycoerythrin (IgG1-PE; BD Bioscience). SSR 47-CAR expression was detected by staining T cells with an IL13Rα2 chimera followed by Fc-FITC (Milipore) or Fc-PE (SouthernBiotech). LSR 47-CARs were detected using Fc-FITC or Fc-PE. U373 cells were analyzed for PD-L1 expression using a CD271 PE antibody (BD Bioscience). Forward- and side-scatter gating were used to discriminate live cells from dead cells. Cells were collected and washed once with phosphate-buffered saline (PBS) containing 1% FBS (Sigma; FACS buffer) prior to the addition of antibodies. Cell were incubated for 30 minutes on ice in the dark, washed once, and fixed in 0.5% paraformaldehyde/FACS buffer prior to analysis.

PCR

To determine the expression of IL13Rα2 in various glioma cells and astrocytes, total RNA was generated from the cell pellets using the RNeasy Plus kit. 200 ng of total RNA was then converted into cDNA using the cDNA iScript kit. The cDNA was further amplified by PCR for IL13Rα2 and GAPDH for 30 cycles using IL13Rα2 and GAPDH primers and visualized on a 1% agarose gel.

Surface Plasmon Resonance

The affinity and rates of interaction between IL13Rα2 (clone 47) monoclonal antibody, commercially available IL13Rα2 monoclonal antibodies (clones 83807 and B-D13), and target (rhIL13Rα2) were measured with a Biacore 3000 biosensor through surface plasmon resonance (SPR). The monoclonal antibodies were immobilized (covalently) to the dextran matrix of the sensor chip (CM5) using the amino coupling kit. The carboxyl groups on the sensor surfaces were activated with an injection of a solution containing 0.2M N-ethyl-N′-(3-diethylamino-propyl)-carbodiimide and 0.05M N-hydroxysuccinimide. The immobilization procedure was completed by the injection of 1Methanolamine hydrochloride to block the remaining ester groups. All steps of the immobilization process were carried out at a flow rate of 10 μl/minute. The control surface was prepared similarly with the exception that running buffer was injected rather than monoclonal antibodies. Binding reactions were performed at 25° C. in HBS-P buffer (20 mM HEPES, pH 7.4, 150 mM NaCl, and 0.005% (v/v) surfactant P20) using a flow rate of 20 μl/minute. Target (rhIL13Rα2) was added at various concentrations in the flow during the binding phase. The amount of protein bound to the sensor chip was monitored by the change in refractive index (represented by response units (RU)). The instrument was programmed to perform a series of binding measurements with increasing concentrations of target over the same surface. Triplicate injections of each concentration of target were performed. Sensorgrams (plots of changes in RU on the surface as a function of time) were analyzed using BIAevaluation v4.1. Affinity constants were estimated by curve fitting using a 1:1 binding model.

Data Preparation and Kinetic Analysis

The estimation of kinetic parameters was performed by repetitive injections of a range of target concentrations over the immobilized mAbs. Data were prepared by the method of “double referencing.” This method utilizes parallel injections of each target sample over a control dextran surface as well as running buffer injections over both the immobilized mAbs and control dextran surfaces. Subtraction of these sensorgrams yielded the control; this was subtracted from the experimental sensorgram. Each data set (consisting of sensorgrams of increasing target concentrations over the same level of immobilized mAbs) was analyzed using various kinetic models. The BIAevaluation v 4.1 software was then used for data analysis. Affinity constants were estimated by curve fitting using a 1:1 binding model. Sensorgram association and dissociation curves were fit locally or globally. The rate of complex formation during the sample injection is described by an equation of the following type: dR/dt=kaC (Rmax−R)−kdR (for a 1:1 interaction) where R is the SPR signal in RU, C is the concentration of analyte, Rmax is the maximum analyte binding capacity in RU, and dR/dt is the rate of change of SPR signal. The early binding phase (300 s) was used to determine the association constant (ka) between mAb and target. The dissociation phase (kd) was measured using the rate of decline in RU on introduction of free buffer at the end of target injections. Data were simultaneously fit by the software program (global fitting algorithm), and the dissociation constant (KD) of the complexes was determined as the ratio ka/kd. For quantitative analysis, three independent replicates were performed for each sample. Data are expressed as mean±S.E.

Competitive Binding Assay

For the competitive binding plate assay, a 96-well plate was coated with 50 μl of affinity-purified hrIL13Rα2hFc at 1 μg/ml in carbonate buffer, pH 9.6 and stored overnight at 4° C. After washing with PBS containing 0.05% Tween 20, mAbs to IL13Rα2 (10 μg/ml) or control mIgG were added for 30 minutes at room temperature. After washing, 50 μl of purified rhIL-13 in PBS and 0.1% BSA at 10 ng/ml were added for a 1-hour incubation at room temperature and assayed for bound rhIL-13 using detection reagents from a human IL-13 ELISA kit. Separately, HEK cells expressing wild-type IL13Rα2 or 4-amino-acid mutants (see Example 10) in the IL13Rα2 sequence were pretreated with either rhIL-13 or mAb IL13Rα2 (clone 47) at 2 μg/ml for 30 minutes on ice followed by a 1-hour incubation with IL13Rα2 (clone 47) mAb or rhIL-13 at 100 ng/ml, respectively. Binding of rhIL-13 to IL13Rα2 alone or in the presence of competitor was detected with human IL-13 mAb-FITC. Binding of IL13Rα2 (clone 47) mAb to rhIL13Rα2 alone or in the presence of competitor was detected with anti-mouse antibody conjugated to Alexa Fluor 649 and analyzed by flow cytometry.

Mutagenesis of IL13Rα

Previously, Tyr207, Asp271, Tyr315, and Asp318 of the human IL13Rα2 were identified as residues crucial for interaction with human IL-13 (28). To determine whether those residues were important for binding of IL13Rα2 (clone 47) mAb to IL13Rα2, the Tyr207, Asp271, Tyr315, and Asp318 residues were mutated to Ala separately or at the same time (4-amino-acid mutant) using the QuikChange Lightning site-directed mutagenesis kit according to the manufacturer's recommendations. Sequencing of selected clones was performed using conventional techniques, which confirmed the presence of the selected mutation. HEK cells were transfected with wild-type or mutated variants of IL13Rα2 cDNA in the pEF6 Myc-His vector using Lipofectamine Plus transfection reagent. 48 hours after transfection, the cells were collected and analyzed for binding to IL13Rα2 (clone 47) mAb via flow cytometry.

Western Blot

The rhIL13Rα2 was applied to a 7.5% Tris-HCl gel (Bio-Rad) at 200 ng/lane and resolved under reducing conditions. After the transfer of proteins to a PVDF membrane (Bio-Rad) and blocking with 2% nonfat dry milk, the membrane was stained with anti-IL13Rα2 mAb (clones YY-23Z and B-D13) at 2 μg/ml or with supernatant collected from hybridoma clones (diluted 10 times), followed by goat anti-mouse antibody conjugated to peroxidase. ImmunStar™ WesternC™ was used to develop reactions. Images were captured using a Bio-Rad ChemiDoc imaging system.

For experiments disclosed in Examples 13-16, cells were dissociated with PBS+3 mM EDTA and lysed in a buffer containing 50 mM Tris, 150 mM NaCl, 5 mM EDTA, 1% Triton X-100 (all from Sigma, St. Louis, MO), and protease inhibitors (Thermo Scientific, Waltham, MA). Protein concentrations were determined using a Bio-Rad protein assay (Bio-Rad, Hercules, CA) with bovine serum albumin (BSA) as the standard. Samples were denatured in Laemmli buffer (Bio-Rad) at 95° C. for 5 minutes. 5pg of protein were loaded per well and run on a 10% polyacrylamide gel. Proteins were transferred to nitrocellulose membranes (BioRad). Membranes were blocked with 5% milk powder (MP) in Tris-buffered saline (TBS)+0.1% Tween-20 (Sigma) and then probed with anti-CD3.ζ (sc-1239, Santa Cruz Biotechnology, Inc., Santa Cruz, CA) or GAPDH (sc-47724, Santa Cruz Biotechnology, Inc., Santa Cruz, CA) mouse monoclonal antibodies followed by a horseradish peroxidase (HRP) conjugated goat anti-mouse IgG antibody (sc-2005, Santa Cruz Biotechnology, Inc.). Blots were developed using SuperSignal West Dura Extended Duration Substrate (Thermo Scientific) and exposed to GeneMate Blue Basic Autoradiography Film (BioExpress, Kaysville, UT).

Immunohistochemistry

The GBM tissues were collected in accordance with a protocol approved by the Institutional Review Board at the University of Chicago. Flash-frozen brain-tumor tissues were cut to a thickness of 10 μm. Tissue sections were fixed with −20° C. methanol and stained for human IL13Rα2 using mouse IL13Rα2 (clone 47) mAb at a concentration of 3 pg/ml or isotype control mIgG1. The bound antibodies were detected with biotinylated horse anti-mouse antibodies (1:100). The antigen-antibody binding was detected by the Elite kit with 3,3′-diaminobenzidine substrate. Slides were analyzed using the CRI Panoramic Scan Whole Slide Scanner and Panoramic Viewer software.

Animal Study

All animals were maintained and cared for in accordance with the Institutional Animal Care and Use Committee protocol and according to National Institutes of Health guidelines. The animals used in the experiments were 6- to 7-week-old male athymic nu/nu mice. Mice were anesthetized with an intraperitoneal injection of ketamine hydrochloride/xylazine (25 mg/ml/2.5 mg/ml) mixture. To establish intracranial tumors, a midline cranial incision was made, and a right-sided burr hole was placed 2 mm lateral to the sagittal suture and about 2 mm superior to X. Animals were positioned in a stereotactic frame, and a Hamilton needle was inserted through the burr hole and advanced 3 mm. Intracranial penetration was followed by (i) injection of 2.5×104 U251 glioma cells in 2.5 μl of sterile PBS in combination with 200 ng of mIgG or IL13Rα2 (clone 47) mAb or (ii) 3 days post-intracranial injection of glioma cells with PBS or 10 μg of IL13Rα2 (clone 47 or B-D13) mAb as described previously (29, incorporated herein by reference). All mice were monitored for survival. Three animals from each group were sacrificed at day 17, and brains were harvested and frozen for sectioning, hematoxylin and eosin (H&E) staining, and microscopic analysis.

Animal experiments disclosed in Examples 13-16 followed a protocol approved by the Baylor College of Medicine Institutional Animal Care and Use Committee. Experiments were performed as described in Ahmed et al., Clin. Cancer Res. 16:474-485 (2010) (incorporated herein by reference) with a few modifications. ICR-SCID mice were purchased from Taconic (IcrTac:ICR-Prkdcscid; Fox Chase C.B-17 SCID™ ICR; Taconic, Hudson, NY). Male 7- to 9-week-old mice were anesthetized, head were shaved and the mice were immobilized in a Cunningham™ Mouse/Neonatal Rat Adaptor (Stoelting, Wood Dale, IL) stereotactic apparatus fitted into an E15600 Lab Standard Stereotaxic Instrument (Stoelting), and then scrubbed with 1% povidone-iodine. A 10 mm skin incision was made along the midline. The tip of a 30G %2 inch needle mounted on a Hamilton syringe (Hamilton, Reno, NV) served as the reference point. A 1 mm burr-hole was drilled into the skull 1 mm anterior and 2 mm to the right of the bregma. 1×105 U373.eGFP.ffLuc cells in 2.0 μL were injected 3 mm deep to the bregma, corresponding to the center of the right caudate nucleus over 5 minutes. The needle was left in place for 3 minutes to avoid tumor cell extrusion, and then withdrawn over 5 minutes. Seven days after tumor cell injection, animals were treated with 2×106 effector cells in 2 μL to the same tumor coordinates. The incision was closed with 2-3 interrupted 7.0 Ethilon sutures (Ethicon, Inc., Somerville, NJ). A subcutaneous injection of 0.03-0.1 mg/kg buprenorphine (Buprenex® RBH, Hull, England) was given for pain control.

Generation of Retroviral Vectors Encoding IL13Rα2-scFv-Specific CARs

A codon-optimized gene was synthesized by GeneArt (Invitrogen, Carlsbad, CA) containing the immunoglobulin heavy-chain leader peptide 37, and scFv47 flanked by 5′ NcoI and 3′ BamHI sites. This mini gene was subcloned into SFG retroviral vector containing IL13Rα2-specific CARs (47-CARs) with short or long spacer regions (SSRs, LSRs) and CD28.ζ, CD28.OX40.ζ, CD28.41BB.ζ, or 41BB.ζ endodomains.5,38,39 All CARs contained a CD28 transmembrane domain except for 47.SSR.CAR.41BB.ζ, which had a CD8α transmembrane domain. 47.SSR.CAR and 47.LSR.CAR without an endodomain (47.SSR.CAR.Δ and 47.LSR.CAR.Δ) were generated by PCR cloning. All cloning of the CARs were verified by sequencing (Seqwright, Houston, TX). RD114-pseudotyped retroviral particles were generated by transient transfection of 293T cells as previously described in Johnson et al., Sci. Transl. Med. 7:275ra22 (2015), incorporated herein by reference.

Generation of CAR T Cells

Human peripheral blood mononuclear cells (PBMCs) from healthy donors were obtained under a Baylor College of Medicine IRB-approved protocol, after informed consent was obtained in accordance with the Declaration of Helsinki. To generate 47-CAR T cells, PBMCs were isolated by Lymphoprep (Greiner Bio-One, Monroe, NC) gradient centrifugation and then stimulated on non-tissue culture treated 24-well plates, which were precoated with OKT3 (CRL-8001, ATCC) and CD28 (BD Bioscience, Mountain View, CA) antibodies. Recombinant human interleukin-7 (IL7) and IL15 (IL7, 10 ng/mL; IL15, 5 ng/mL; Proleukin; Chiron, Emeryville, CA) were added to cultures on day 2 (Xu et al., Blood 123:3750-3759 (2014), incorporated herein by reference). On day 3, OKT3/CD28-stimulated T cells (2.5×105 cells/well) were transduced on RetroNectin® (Clontech, Mountainview, CA)-coated plates in the presence of IL7 and IL15. On day 5 or 6, T cells were transferred into new wells and subsequently expanded with IL-7 and IL15. Non-transduced (NT) T cells were activated with OKT3/CD28 and expanded in parallel with IL-7 and IL15. 47-CAR expression was determined 3 to 4 days post-transduction.

Co-Culture Assay

Recombinant protein co-culture assay. Non-tissue culture 24-well plates were precoated with recombinant human IL13Rα1, IL13Rα2, or IL4R protein, (R&D Systems, Minneapolis, MN) at a final concentration of 500ng/well. Plates were washed once using RPMI, and CAR or NT T cells were plated. After 24 hours, supernatants were harvested and interferon γ (IFN γ) and Interleukin 2 (1L2) release were measured by ELISA according to the manufacturer's instructions (R&D Systems, Minneapolis, MN).

Cell culture co-culture assay. CAR T cells were co-cultured with target cells at a 1:2 effector to target (E:T) ratio in a 24-well plate. NT T cells served as controls. After 24 hours, culture supernatants were harvested, and the presence of IFNγ and IL2 was determined by ELISA according to the manufacturer's instructions (R&D Systems, Minneapolis, MN).

Cytotoxicity Assay

Standard chromium (51Cr) release assays were performed as described in Gottschalk et al., Blood 101:1905-1912 (2003), incorporated herein by reference. Briefly, 1×106 target cells were labeled with 0.1 mCi (3.7MBq) 51Cr and mixed with decreasing numbers of effector cells to give effector to target ratios of 40:1, 20:1, 10:1, and 5:1. Target cells incubated in complete medium alone or in 1% Triton X-100 were used to determine spontaneous and maximum 51Cr release, respectively. After 4 hours, supernatants were collected and radioactivity was measured in a gamma counter (Cobra Quantum; PerkinElmer; Wellesley; MA). The mean percentage of specific lysis of triplicate wells was calculated according to the following formula: [test release−spontaneous release]/[maximal release−spontaneous release]×100.

Bioluminescence Imaging

Isofluorane anesthetized animals were imaged using the IVIS® system (IVIS, Xenogen Corp., Alameda, CA) 10-15 minutes after 150 mg/kg D-luciferin (Xenogen) per mouse was injected intraperitoneally. The photons emitted from the luciferase-expressing tumor cells were quantified using Living Image software (Caliper Life Sciences, Hopkinton, MA). A pseudo-color image representing light intensity (blue least intense and red most intense) was generated and superimposed over the grayscale reference image. Mice were euthanized when the tumor radiance was greater than 1×109 on two occasions or when they met euthanasia criteria (neurological deficits, weight loss, signs of distress) in accordance with the Center for Comparative Medicine at Baylor College of Medicine.

Statistics

The differences between groups were evaluated by Student's t test or one-way analysis of variance with post hoc comparison Tukey's test or Dunnett's test. For the in vivo survival data, a Kaplan-Meier survival analysis was used, and statistical analysis was performed using a log rank test. P<0.05 was considered statistically significant.

For the experiments disclosed in Examples 13-16, the in vitro experiments were performed at least in triplicate, and GraphPad Prism 5 software (GraphPad software, Inc., La Jolla, CA) was used for statistical analysis. Measurement data were presented as mean±standard deviation (SD). The differences between means were tested by appropriate tests. The significance level used was P<0.05. For the mouse experiments, changes in tumor radiance from baseline at each time point were calculated and compared between groups using t-test. Survival, determined from the time of tumor cell injection, was analyzed by the Kaplan-Meier method and by the log-rank test.

Example 2

Characterization of Antigen and Screening of Hybridoma Clones Secreting Anti-IL13Rα2 Antibodies

The primary goal of this study was to generate a high affinity monoclonal antibody suitable for targeting of the IL13Rα2 expressed on the surface of tumor cells. We therefore immunized mice and screened the resulting hybridoma clones for reactivity against the antigen, rhIL13Rα2, in its native conformation. A plate-bound ELISA utilizing a hybridoma clone against rhIL13Rα2, YY-23Z, was established for the detection of rhIL13Rα2. The concentration of rhIL13Rα2 absorbed to the plastic at 1 μg/ml was found to be suitable for the detection of antibody binding (FIG. 1A). Next, the rhIL13Rα2hFc was characterized for its “nativity” by utilizing a pair of commercially available antibodies recognizing only the native (found on the cell surface) and denatured (using Western blotting under reducing conditions) forms of IL13Rα2 and for its binding properties to rhIL13Rα2 in ELISA with antibody clones B-D13 and YY-23Z, respectively. Both clones B-D13 and YY-23Z were able to recognize the rhIL13Rα2hFc in a plate-bound ELISA (FIG. 1B). Denaturation of antigen at 95° C. for 5 minutes in the presence of β-mercaptoethanol completely abolished the ability of the antibody clone B-D13 to recognize antigen by ELISA, whereas the YY-23Z clone retained the ability to bind the denatured antigen. Thus, the rhIL13Rα2hFc absorbed to the plastic of ELISA plates containing both native and denatured forms of the protein. Analysis of serum from animals immunized with a fusion of rhIL13Rα2 and hFc revealed the presence of antibodies against both rhIL13Rα2 and human Fc fragment. To select antibodies specific for the IL13Rα2 portion of the fusion, human IgG was included as an additional negative control for the screening of hybridoma populations. Of the 39 screened primary populations, only 15 populations were specific to IL13Rα2, and four were reactive with human IgG. Finally, five clones strongly reacting with native IL13Rα2 were further expanded and recloned. The two clones recognizing only denatured antigen were selected from the separate immunization set with rhIL13Rα2hFc chimera. Supernatants from selected clones were compared for their ability to bind hrIL13Rα2 in a plate-bound ELISA (FIG. 1C) and by Western blotting (FIG. 1D). FIG. 1C shows that clone 47 strongly binds to the antigen in plate-bound ELISA but not by Western blotting, indicating the ability of clone 47 to recognize a native conformation of the antigen. Therefore, clone 47 was selected for further characterization and for further experiments. Clone 47 was found to be of the IgG1 isotype, possessing a κ chain.

Example 3

Specificity of Binding for the IL13Rα2 (Clone 47) mAb to Recombinant Human IL13Rα2 and IL13Rα2 Expressed at the Cell Surface

We investigated the binding properties of the IL13Rα2 (clone 47) mAb to rhIL13Rα2 versus the commercially available clones 83807 and B-D13 in a plate-bound ELISA. FIG. 2A shows strong and specific binding of clone 47 to rhIL13Rα2 when compared with clones 83807 and B-D13. Clone 47 reached the plateau of binding at the low concentration of 0.05 μg/ml. None of the antibodies showed binding to human IgG utilized as an additional negative control in these experiments. To further verify the specificity of interaction for clone 47 with human IL13Rα2, a clonal line of CHO cells expressing the full size wild-type human IL13Rα2 (clone 6) was generated. Binding of the antibody to control CHO cells transfected with an empty vector was compared with that of CHO cells expressing IL13Rα2. Again, the IL13Rα2 (clone 47) mAb demonstrated strong and specific binding to IL13Rα2 expressed on the cell surface but not to control CHO cells, indicating that this antibody specifically recognizes a native conformation of the IL13Rα2 (FIG. 2B). Clone 47 demonstrated the strongest affinity for IL13Rα2 at the lowest tested concentration of 0.25 μg/ml. Notably, other selected hybridoma clones demonstrated similar specificity of interaction with IL13Rα2 expressed on the cell surface of CHO cells but not with control CHO cells. Data obtained in a plate-bound ELISA also revealed that clone 47 does not interact with the low affinity receptor for IL-13, the IL13Rα1 (FIG. 2C), or mouse recombinant IL13Rα2, further validating the specificity of interaction between clone 47 and IL13Rα2 (FIG. 2D). Clones 83807 and B-D13 did not show binding to mouse rIL13Rα2 in agreement with current understanding of the cross-reactivity of these antibodies with mouse IL13Rα2.

We next characterized the binding capacity of clone 47 with various glioma cell lines, the patient-derived glioma lines GBM12 and GBM43, and normal human astrocytes. Increased expression of the IL13Rα2 gene relative to normal brain tissue is reported in 44-47% of human GBM resected specimens (3) and in up to 82% (14 of 17) primary cell cultures derived from GBM and normal brain explants (2). FIGS. 3, A and B, show the flow charts of the comparative staining of glioma cells, human astrocytes, and HEK cells expressing recombinant human IL13Rα2 on the cell surface with the IL13Rα2 (clones 47, 83807, and B-D13) mAb. FIGS. 3, A and B, reveal (i) various levels of IL13Rα2 expression on the cell surface and (ii) superior binding of the clone 47 versus clones B-D13 (1.2-4.6-fold difference between the cell lines) and 83807 to the surface of analyzed cell lines. Interestingly, we observed a near complete absence of the binding of clone 83807 to glioma cell lines in contrast to HEK cells expressing IL13Rα2. No binding of clone 47 was detected with normal human astrocytes, confirming the specificity of interaction of clone 47 with human glioma cells expressing IL13Rα2. The expression of IL13Rα2 mRNA in these cells generally correlates with the level of IL13Rα2 expression on the cell surface. Moreover, cells expressing low to no mRNA expression for IL13Rα2, including U118 and primary human astrocytes, demonstrated low to no expression for IL13Rα2 on the cell surface (FIG. 3B). In additional experiments, N10 glioma cells were incubated with either the IL13Rα2 (clone 47) mAb at 1 μg/ml or the IL13Rα2 (clone 47) mAb preincubated with a 10-fold excess of rhIL13Rα2 (FIG. 10) and analyzed by flow cytometry. A significant ablation of interaction between the IL13Rα2 (clone 47) mAb in the presence of a 10-fold excess of rhIL13Rα2 was found when compared with clone 47 alone. Similarly, preincubation of N10 cells with either a 10-fold excess of rhIL-13 or IL13Rα2 (clone 47) mAb almost completely blocked the interaction between the antibody or rhIL-13 and N10 cells (supplemental FIG. 1B), indicating a specificity of recognition between IL13Rα2 expressed on the surface of glioma cells and clone 47 (FIG. 10).

To verify that the IL13Rα2 (clone 47) mAb possessed the ability to bind IL13Rα2 on the surface of glioma cells in situ, intracranial glioma xenografts of U251 cells expressing green fluorescent protein (GFP) were established in nude mice. Three weeks later, animals were sacrificed, and cells were obtained and placed into in vitro culture conditions. After 48 hours, the cells were collected and stained with control mIgG or IL13Rα2 (clone 47) mAb. Cultured GFP-expressing U251 cells served as a positive control. GFP-positive U251 cells represented about 56% of the total cells (FIG. 3C, panel a), and 96% of the cells were reactive with the IL13Rα2 (clone 47) mAb (FIG. 3C, panel c), whereas GFP-negative cells did not interact with the antibody (FIG. 3C, panel b). These data further confirm that the IL13Rα2 (clone 47) mAb specifically recognizes glioma cells expressing IL13Rα2 in mouse xenografts and is not reactive with other cells from the mouse brain.

Example 4

Affinity Studies

Surface plasmon resonance was used to determine the affinity and rate of interaction between the IL13Rα2 (clone 47) mAb and rhIL13Rα2. All measurements were done in comparison with two commercial antibodies against IL13Rα2, clones 83807 and B-D13. FIG. 4 shows the sensorgrams for each antibody. The measurements are summarized in Table 1.

TABLE 1 Kinetics of monoclonal antibodies binding to the human recombinant IL13Rα2 mAbs to ka kd KD Rmax IL13Rα2 1/MS 1/S M RU Clone 47 9.06e4 ± 322 1.26e−4 = 1.07e−6 1.39 × 10−9 390 Clone 83807 2.23e4 ± 620 2.31e−3 = 1.03e−5  104 × 10−9 250 Clone B-D13    1.08e5 ± 5.71e3 4.99e−3 = 1.45e−4 46.1 × 10−9 8-16

The estimation of kinetic parameters was performed as described in Example 1. The dissociation constant (KD) of the complexes was determined as the ratio ka/kd. For quantitative analysis, three independent replicates were performed for each sample. Data are expressed as mean f S.E. These data demonstrate that the affinity of IL13Rα2 (clone 47) mAb to recombinant IL13Rα2 exceeds the affinity of commercially available mAb clones 83807 and B-D13 by 75-fold and 33-fold, respectively.

FIG. 4A shows that clone 47 demonstrates a prolonged and stable association with rhIL13Rα2 measured over a 30-minute time frame, whereas clones 83807 (FIG. 4B) and B-D13 (FIG. 4C) dissociate relatively quickly. The affinity of binding for the IL13Rα2 (clone 47) mAb to rhIL13Rα2 was calculated at 1.39×10−9 M. This value exceeded the affinity of the commercially available antibody clones 83807 and B-D13 to rhIL13Rα2 by 75-fold and 33-fold, respectively. Clone 47 demonstrated the highest binding affinity (Rmax) to rhIL13Rα2 at 390 RU when compared with 250 and 8-16 RU for clones 83807 and B-D13, respectively. These data indicate that the IL13Rα2 (clone 47) mAb possesses properties superior to clones 83807 and B-D13 as well as demonstrates a higher affinity toward rhIL13Rα2.

Example 5

A Monoclonal Antibody Competes with rhIL-13for Binding to IL13Rα2

To determine whether the IL13Rα2 (clone 47) mAb possesses inhibitory properties, competitive binding assays utilizing a rhIL13Rα2hFc chimera and HEK cells transiently expressing the human IL13Rα2 were performed. The competitive binding assay was set up in a plate-bound ELISA format. The rhIL13Rα2hFc absorbed to the plate served as the target antigen. To determine whether the IL13Rα2 mAb specifically inhibits the binding of IL-13 to rhIL13Rα2, plates were preincubated with a 100-fold excess of mIgG, the IL13Rα2 (clone 47) mAb, or other IL13Rα2 mAb clones, including 83807, YY-23Z, and B-D13, followed by incubation with rhIL13. FIG. 5A shows that the IL13Rα2 (clone 47) mAb significantly abolished the binding of rhIL-13 to rhIL13Rα2, whereas the IL13Rα2 mAb clones B-D13 and 83807 exhibited significantly less competition for binding of human IL-13.

To further verify the inhibitory properties of the IL13Rα2 (clone 47) mAb, HEK 293T cells were transfected with an agent encoding wild-type or a 4-amino-acid mutant form of IL13Rα2 cDNA in which Tyr207, Asp271, Tyr315, and Asp318 residues were substituted with Ala. Previously, these residues of the human IL13Rα2 were identified as amino acids required for the interaction with the cognate ligand, IL-13. The presence of all four mutations in one molecule has been shown to result in near complete loss of the binding of IL-13 to the mutated form of IL13Rα2 (28). After 48 hours, the cells were pretreated with a 20-fold excess of rhIL-13 or the IL13Rα2 (clone 47) mAb, followed by incubation of the IL13Rα2 (clone 47) mAb or rhIL-13, respectively. FIG. 5B shows about 50% binding inhibition of IL13Rα2 (clone 47) mAb by a 20-fold excess of rhIL-13 to wild-type (WT) IL13Rα2 but not to the 4-amino-acid mutant form of IL13Rα2. A 20-fold excess of antibody abolished the binding of rhIL-13 to IL13Rα2 when expressed on the cell surface by 80%, which is similar to the result observed in plate ELISA. The residual binding of IL-13 to the 4-amino-acid mutant form of IL13Rα2 was further decreased by an excess of the IL13Rα2 (clone 47) mAb (FIG. 5C). Collectively, these data indicate that the IL13Rα2 (clone 47) mAb specifically competes with rhIL-13 for the binding site on IL13Rα2. Also, these data indicate that the IL13Rα2 (clone 47) mAb and IL-13 have a significant overlap in their recognition site of the IL13Rα2 molecule.

Example 6

Role of the Tyr207, Asp271, Tyr315, and Asp318 Residues for IL13Rα2 (Clone 47) mAb Binding

Taking into consideration that IL-13 and the IL13Rα2 (clone 47) monoclonal antibody can significantly compete with one other for binding of IL13Rα2, we determined whether the residues Tyr207, Asp271, Tyr315, and Asp318 contributing to the interaction of IL-13 with IL13Rα2 (28) were also important for binding of the IL13Rα2 (clone 47) mAb to IL13Rα2. The plasmids encoding cDNA for IL13Rα2 carrying individual mutations of Tyr207, Asp271, Tyr315, or Asp318 residues to Ala or a combination of all four mutations in one molecule were generated and transiently expressed in HEK cells. Binding of the IL13Rα2 (clone 47) mAb to wild-type and mutant forms of IL13Rα2 was analyzed by flow cytometry. The IL13Rα2 mAbs 83807 and B-D13 were used as reference antibodies to exclude a possible influence of variations in the level of expression of wild-type or mutated variants of IL13Rα2 on the surface of HEK cells (FIG. 6A). Data were calculated as a ratio of IL13Rα2 (clone 47) binding to IL13Rα2 when compared with both antibody clones 83807 and B-D13. FIG. 6A demonstrates that the binding of IL13Rα2 (clone 47) mAb was not significantly affected by either the individual mutations or the 4-amino-acid mutant form of IL13Rα2 when compared with wild-type receptor. In contrast, binding of IL-13 to the 4-amino-acid mutant form of IL13Rα2 was nearly abolished (FIG. 6B). These data indicate that the Tyr207, Asp271, Tyr315, and Asp318 residues are not crucial for the interaction of IL13Rα2 (clone 47) mAb with IL13Rα2 but are necessary for binding to IL-13.

Example 7

N-Linked Glycosylation Affects the Affinity of the IL13Rα2 mAb for IL13Rα2

N-Linked glycosylation has previously been demonstrated to be important for efficient binding of IL-13 to the cognate receptor, IL13Ra2 (30). Taking into consideration the significant overlap in epitope recognition between the IL13Rα2 (clone 47) mAb and IL-13, we expected N-linked glycosylation of IL13Ra2 to contribute to binding of the IL13Rα2 (clone 47) mAb. To confirm this expectation, rhIL13Rα2hFc was treated with Pngase F to remove N-linked glycosylation from the protein. The binding of the IL13Rα2 (clone 47) mAb to control and deglycosylated target protein was investigated. Treatment of rhIL13Rα2 with Pngase F was performed under native conditions (in the absence of SDS) to avoid denaturation of the rhIL13Rα2 affecting the binding of antibodies. Additional mAbs to IL13Rα2 (clones 83807, B-D13, and YY23Z) and rhIL-13 were included in the assay to demonstrate the specificity of binding. In a plate-bound ELISA, binding of the IL13Rα2 (clone 47) mAb to Pngase F-treated IL13Rα2 was decreased by 35% when compared with untreated protein (n=4; p<0.001). The binding of the IL13Rα2 (clone 83807) was reduced by 80% when compared with untreated protein and completely absent for the IL13Rα2 mAbs B-D13 and YY-23Z, respectively (n=4; p<0.001) (FIG. 7A). Binding of rhIL-13 with Pngase F-treated rhIL13Rα2 was also significantly diminished. To verify that Pngase F treatment resulted in deglycosylation of the protein, control and Pngase F-treated rhIL13Rα2hFc protein was resolved by Western blot. FIG. 7B shows that Pngase F-treated protein has a lower molecular weight, confirming the removal of N-linked glycans from the IL13Rα2 molecule. Binding of the IL13Rα2 (clone 47) mAb to Pngase F-treated U251 glioma and HEK 293 cells expressing wild-type IL13Rα2 was also decreased by about 30% (n=3; p<0.05) when compared with control untreated cells (FIG. 7C).

Example 8

Immunohistochemistry

The ability of the IL13Rα2 (clone 47) mAb to detect IL13Rα2 was evaluated in fresh frozen tissues. Flash-frozen human GBM samples or the U251 glioma flank xenograft was stained with either isotype control mIgG1 or the IL13Rα2 (clone 47) mAb. FIG. 8 shows positive (brown) staining in the two human GBM samples, albeit with different frequency of positive cells in the sample as well as a U251 glioma cell-based glioma xenograft. Positive staining was detected in two of the three GBM samples analyzed, which is consistent with the expectation that fewer than 50% of primary GBM express IL13Rα2 (3). These data are also consistent with the ability of this antibody to recognize the native form of IL13Rα2 expressed on the cell surface and in ELISA applications, as well as the compromised ability of this mAb to detect denatured antigen by Western blotting.

Example 9

The IL13Rα2 Monoclonal Antibody Prolongs the Survival of Animals with an Intracranial Glioma Xenograft

The potential therapeutic properties of the IL13Rα2 (clone 47) mAb were also determined in an orthotopic mouse model of human glioma. U251 glioma cells were intracranially injected into the brain of nude mice alone, in the presence of control mIgG, or with the IL13Rα2 (clone 47) mAb. FIG. 9A shows that animals in the control PBS (n=15) and mIgG (n=16) groups demonstrated a similar median survival of 27 and 25 days, respectively. In contrast, the survival of animals co-injected with the IL13Rα2 (clone 47) mAb (n=13) was significantly increased to a median of 34 days (p=0.0001; mIgG versus the IL13Rα2 mAb group). Analysis of H&E staining of the glioma xenografts from brains collected on day 17 revealed a similar pattern of glioma cell distribution in the brain of control groups. In contrast, the tumor mass in the group of animals co-injected with IL13Rα2 mAb was significantly decreased in size (FIG. 9B). Independently, U251 cells were inoculated in the brains of mice and 3 days later injected through the same burr hole with either PBS or the IL13Rα2 (clone 47 or B-D13) mAb as described previously (29). Interestingly, the mice injected with clone 47 demonstrated improvement in median survival when compared with PBS and clone B-D13 groups (35 days versus 27 and 23 days, respectively; n=7; p>0.05) (FIG. 11), similar to what was found in the co-injection experiment (FIG. 9A). Nevertheless, all animals ultimately succumbed to the disease. These data indicate that the IL13Rα2 (clone 47) mAb shows promise in promoting tumor rejection of IL13Rα2-expressing U251 glioma cells in the mouse brain. This finding leads to the expectation that antibody agent incorporating the IL13Rα2-binding domain of the IL13Rα2 (clone 47) mAb will be efficacious in treating a variety of human and non-human cancers characterized by the presentation of IL13Rα2, such as IL13Rα2-expressing glioma cells and other malignant cell types.

REFERENCES—FOR EXAMPLES 1-9 AND FOR CITATIONS THROUGHOUT THE APPLICATION UNLESS SPECIFICALLY IDENTIFIED OTHERWISE

  • 1. Debinski, W., Obiri, N. I., Powers, S. K., Pastan, I., and Puri, R. K. (1995) Clin. Cancer Res. 1, 1253-1258.
  • 2. Joshi, B. H., Plautz, G. E., and Puri, R. K. (2000) Cancer Res. 60, 1168-1172.
  • 3. Jarboe, J. S., Johnson, K. R., Choi, Y., Lonser, R. R., and Park, J. K. (2007) Cancer Res. 67, 7983-7986.
  • 4. Kawakami, K., Terabe, M., Kawakami, M., Berzofsky, J. A., and Puri, R. K. (2006) Cancer Res. 66, 4434-4442.
  • 5. Fujisawa, T., Joshi, B., Nakajima, A., and Puri, R. K. (2009) Cancer Res. 69, 8678-8685.
  • 6. Debinski, W., Slagle, B., Gibo, D. M., Powers, S. K., and Gillespie, G. Y. (2000) J. Neurooncol. 48, 103-111.
  • 7. Aman, M. J., Tayebi, N., Obiri, N. I., Puri, R. K., Modi, W. S., and Leonard, W. J. (1996) J. Biol. Chem. 271, 29265-29270.
  • 8. Gauchat, J. F., Schlagenhauf, E., Feng, N. P., Moser, R., Yamage, M., Jeannin, P., Alouani, S., Elson, G., Notarangelo, L. D., Wells, T., Eugster, H. P., and Bonnefoy, J. Y. (1997) Eur. J. Immunol. 27, 971-978.
  • 9. Akaiwa, M., Yu, B., Umeshita-Suyama, R., Terada, N., Suto, H., Koga, T., Arima, K., Matsushita, S., Saito, H., Ogawa, H., Furue, M., Hamasaki, N., Ohshima, K., and Izuhara, K. (2001) Cytokine 13, 75-84.
  • 10. Rahaman, S. O., Sharma, P., Harbor, P. C., Aman, M. J., Vogelbaum, M. A., and Haque, S. J. (2002) Cancer Res. 62, 1103-1109.
  • 11. Fichtner-Feigl, S., Strober, W., Kawakami, K., Puri, R. K., and Kitani, A. (2006) Nat. Med. 12, 99-106.
  • 12. Shimamura, T., Fujisawa, T., Husain, S. R., Joshi, B., and Puri, R. K. (2010) Clin. Cancer Res. 16, 577-586.
  • 13. Fujisawa, T., Joshi, B. H., and Puri, R. K. (2012) Int. J. Cancer 131, 344-356.
  • 14. Murphy, E. V., Zhang, Y., Zhu, W., and Biggs, J. (1995) Gene 159, 131-135.
  • 15. Rich, T., Chen, P., Furman, F., Huynh, N., and Israel, M. A. (1996) Gene 180, 125-130.
  • 16. Stupp, R., Mason, W. P., van den Bent, M. J., Weller, M., Fisher, B., Taphoorn, M. J., Belanger, K., Brandes, A. A., Marosi, C., Bogdahn, U., Curschmann, J., Janzer, R. C., Ludwin, S. K., Gorlia, T., Allgeier, A., Lacombe, D., Cairncross, J. G., Eisenhauer, E., and Mirimanoff, R. O. (2005) N. Engl. J. Med. 352, 987-996.
  • 17. Kunwar, S., Prados, M. D., Chang, S. M., Berger, M. S., Lang, F. F., Piepmeier, J. M., Sampson, J. H., Ram, Z., Gutin, P. H., Gibbons, R. D., Aldape, K. D., Croteau, D. J., Sherman, J. W., and Puri, R. K. (2007) J. Clin. Oncol. 25, 837-844.
  • 18. Wykosky, J., Gibo, D. M., Stanton, C., and Debinski, W. (2008) Clin. Cancer Res. 14, 199-208.
  • 19. Debinski, W., Gibo, D. M., Obiri, N. I., Kealiher, A., and Puri, R. K. (1998) Nat. Biotechnol. 16, 449-453.
  • 20. Kawakami, M., Kawakami, K., and Puri, R. K. (2002) Mol. Cancer Ther. 1, 999-1007.
  • 21. Husain, S. R., and Puri, R. K. (2000) Blood 95, 3506-3513.
  • 22. Bartolazzi, A., Nocks, A., Aruffo, A., Spring, F., and Stamenkovic, I. (1996) J. Cell Biol. 132, 1199-1208.
  • 23. Kioi, M., Seetharam, S., and Puri, R. K. (2008) Mol. Cancer Ther. 7, 1579-1587.
  • 24. Pini, A., and Bracci, L. (2000) Curr. Protein Pept. Sci. 1, 155-169.
  • 25. Ross, J. S., Gray, K., Gray, G. S., Worland, P. J., and Rolfe, M. (2003) Am. J. Clin. Pathol. 119, 472-485.
  • 26. Souriau, C., and Hudson, P. J. (2003) Expert Opin. Biol. Ther. 3, 305-318.
  • 27. Köhler, G., and Milstein, C. (1975) Nature 256, 495-497.
  • 28. Arima, K., Sato, K., Tanaka, G., Kanaji, S., Terada, T., Honjo, E., Kuroki, R., Matsuo, Y., and Izuhara, K. (2005) J. Biol. Chem. 280, 24915-24922.
  • 29. Sampson, J. H., Crotty, L. E., Lee, S., Archer, G. E., Ashley, D. M., Wikstrand, C. J., Hale, L. P., Small, C., Dranoff, G., Friedman, A. H., Friedman, H. S., and Bigner, D. D. (2000) Proc. Natl. Acad. Sci. U.S.A. 97, 7503-7508.
  • 30. Kioi, M., Seetharam, S., and Puri, R. K. (2006) FASEB J. 20, 2378-2380.
  • 31. McKenzie, A. N., Culpepper, J. A., de Waal Malefyt, R., Briere, F., Punnonen, J., Aversa, G., Sato, A., Dang, W., Cocks, B. G., and Menon, S. (1993) Proc. Natl. Acad. Sci. U.S.A. 90, 3735-3739.
  • 32. Donaldson, D. D., Whitters, M. J., Fitz, L. J., Neben, T. Y., Finnerty, H., Henderson, S. L., O'Hara, R. M., Jr., Beier, D. R., Turner, K. J., Wood, C. R., and Collins, M. (1998) J. Immunol. 161, 2317-2324.
  • 33. Hansen, H. J., Goldenberg, D. M., Newman, E. S., Grebenau, R., and Sharkey, R. M. (1993) Cancer 71, 3478-3485.
  • 34. Kuan, C. T., Wikstrand, C. J., Archer, G., Beers, R., Pastan, I., Zalutsky, M. R., and Bigner, D. D. (2000) Int. J. Cancer 88, 962-969.
  • 35. Imperiali, B., and Rickert, K. W. (1995) Proc. Natl. Acad. Sci. U.S.A. 92, 97-101.
  • 36. Sondermann, P., Huber, R., Oosthuizen, V., and Jacob, U. (2000) Nature 406, 267-273.
  • 37. Krapp, S., Mimura, Y., Jefferis, R., Huber, R., and Sondermann, P. (2003) J. Mol. Biol. 325, 979-989.
  • 38. Fernandes, H., Cohen, S., and Bishayee, S. (2001) J. Biol. Chem. 276, 5375-5383.
  • 39. Hsu, Y. F., Ajona, D., Corrales, L., Lopez-Picazo, J. M., Gurpide, A., Montuenga, L. M., and Pio, R. (2010) Mol. Cancer 9, 139.
  • 40. Cartron, G., Watier, H., Golay, J., and Solal-Celigny, P. (2004) Blood 104, 2635-2642.
  • 41. de Haij, S., Jansen, J. H., Boross, P., Beurskens, F. J., Bakema, J. E., Bos, D. L., Martens, A., Verbeek, J. S., Parren, P. W., van de Winkel, J. G., and Leusen, J. H. (2010) Cancer Res. 70, 3209-3217.
  • 42. Minn, A. J., Kang, Y., Serganova, I., Gupta, G. P., Giri, D. D., Doubrovin, M., Ponomarev, V., Gerald, W. L., Blasberg, R., and Massague, J. (2005) J. Clin. Investig. 115, 44-55.
  • 43. Kawakami, M., Kawakami, K., Kasperbauer, J. L., Hinkley, L. L., Tsukuda, M., Strome, S. E., and Puri, R. K. (2003) Clin. Cancer Res. 9, 6381-6388.
  • 44. Husain, S. R., Obiri, N. I., Gill, P., Zheng, T., Pastan, I., Debinski, W., and Puri, R. K. (1997) Clin. Cancer Res. 3, 151-156.
  • 45. Puri, R. K., Leland, P., Obiri, N. I., Husain, S. R., Kreitman, R. J., Haas, G. P., Pastan, I., and Debinski, W. (1996) Blood 87, 4333-4339.
  • 46. Joshi, B. H., Leland, P., and Puri, R. K. (2003) Croat. Med. J. 44, 455-462.
  • 47. Barderas R, Bartolomé R A, Fernandez-Aceñero M J, Torres S, Casal J I. Cancer Res. 2012 Jun. 1; 72(11):2780-90.

Example 10

A Single-Chain Antibody for Selective Targeting of IL13Rα2-Expressing Brain Tumors

IL13Rα2 is overexpressed in a majority of high-grade astrocytomas and other malignancies, and has been validated as a target for therapeutic applications in various preclinical models. However, current IL13-based therapeutic agents lack specificity due to interaction with the IL13Rα1 receptor, which is widely expressed by normal or healthy cells. The generation of a targeting agent that strictly binds to IL13Rα2 would significantly expand the therapeutic potential for the treatment of IL13Rα2-expressing cancers. Recently, a monoclonal antibody 47 (mAb47) has been developed and extensively characterized. The mAb47 exclusively binds to a native form of human IL13Rα2. Using mAb47, a single-chain antibody (scFv) fragment was engineered from mAb47 expressed by the parental hybridoma cell line. The single-chain antibody (scFv) fragment was tested for its targeting properties as a soluble agent, and an adenovirus (Ad) with a modified fiber incorporating scFv47 as a targeting motif was agented.

The phage-display approach was utilized for selection of a functional combination of variable heavy (VH) and light (VL) chains from established hybridoma cells producing mAb47. Purified phages displaying scFv47 were tested for their interaction with IL13Rα2hFc recombinant protein, i.e., a fusion of IL13Rα2 and the Fc region of an antibody. A competitive ELISA was utilized to verify that the parental mAb47 and the scFv47 fragment bind to the same epitope. The soluble form of scFv47 expressed in E. coli and CHO cells was analyzed by SDS-PAGE, and tested for stability and targeting properties. To generate IL13Rα2-specific Ad, the fiber of a replication-deficient Ad5 encoding green fluorescent protein was replaced with a chimeric fiber gene composed of a T4 fibritin trimerization domain linked at its C-terminal to scFV47 (AdFFscFv47-CMV-GFP). To generate viral particles, an agent encoding the adenoviral genome was rescued in HEK293F28 cells, propagated, and purified. IL13Rα2+ and IL13Rα2 U251 cell lines were established via stable transfection with either control or IL13Rα2-specific shRNAs (U251-IL13Ra2.KO), respectively. The AdFFscFv47-CMV-GFP virus was tested for targeting properties in these U251 cell lines and in IL13Rα2-expressing U87 cells.

The biopanning-selected pool of phages, as well several individual clones, demonstrated specific binding to IL13Rα2hFc protein, but not to hIgG in plate ELISA. Binding of scFv47-displayed phages to IL13Rα2 was completely abolished by mAb47, but not by control IgG or other tested IL13Rα2 mAbs, thus confirming the same IL13Rα2 epitope was recognized by scFv47 as was recognized by the parental mAb47. Similarly to phage-displayed scFv47, the soluble scFv47 showed specific binding to IL123Ra2, but not to IL13Rα1. Interaction of Ad5FFscFv47-CMV-GFP was also specific to IL13Rα2-expressing U251 cells, as judged by flow cytometry for GFP expression in U251-IL13Rα2+ versus U251-IL13Rα2.KO cells. Furthermore, GFP expression in cells infected with Ad5FFscFv47-CMV-GFP strongly correlated with the level of surface expression of IL13Rα2. The specificity of viral infection was further validated in a U251 glioma model.

The data validate scFv47 as a highly selective IL13Rα2 targeting agent that provides a soluble, single-chain biologic useful in diagnosing and treating IL13Rα2-expressing cancers, such as gliomas, colon cancers (see Example 12) and others.

Example 11

Generation of an IL13Rα2-CAR

To generate an IL13Rα2-specific T cell, an IL13Rα2-specific chimeric antigen receptor (CAR) was initially constructed. A codon-optimized minigene was synthesized that contained the immunoglobulin heavy-chain leader peptide and the heavy and light chains of the IL13Rα2-specific single-chain variable fragment (scFv) separated by a linker (the scFv was derived from hybridoma 47, Balyasnikova et al. J Biol. Chem. 2012; 287(36):30215-30277). The minigene was subcloned into an SFG retroviral vector containing the human IgG1-CH2CH3 domain, a CD28 transmembrane domain, and costimulatory domains derived from CD28 and the CD3ζ-chain. CD3/CD28-activated human T cells were transduced with RD114-pseudotyped retroviral particles and subsequently expanded using IL2. Functional analysis revealed that T cells expressing IL13Rα2-specific CARs (IL13Rα2-CAR T cells) recognized recombinant IL13Rα2 protein as judged by cytokine production (IFNγ and IL2; FIGS. 19 and 20), and killed IL13Rα2-positive cells in a cytotoxicity assay (FIG. 18). Non-transduced (NT) T cells did not produce cytokines and had no cytolytic activity.

Example 12

Redirecting T Cells to IL13Rα2-Positive Pediatric Glioma

IL13Rα2 is aberrantly expressed in Glioblastoma Multiforme and is, therefore, a promising target for CAR T-cell immunotherapy. The antigen recognition domain of CARs normally consists of a single-chain variable fragment (scFv), but current IL13Rα2-specific CARs use IL13 muteins as an antigen recognition domain. IL13 mutein-based CARs, however, have been shown to also recognize IL13Rα1, raising significant safety concerns. To overcome this obstacle, a high affinity IL13Rα2-specific scFv has been agented. This scFv is used in developing a scFv-based IL13Rα2-specific CAR (IL13Rα2-CAR), which, when expressed in T cells, will provide IL13Rα2-CAR T cells having cytotoxic effector function.

Antigen-specific T cells were incorporated into an effective immunotherapy for diffuse intrinsic pontine glioma (DIPG) and glioblastoma (GBM), which are the most aggressive, uniformly fatal, primary human brain tumors in children. IL13Rα2 is expressed at a high frequency in both DIPG and GBM, but not in normal brain, making it a promising target for T-cell immunotherapy, including scFv-based therapy, scFv-CAR T-cell-based therapy, and scFv fusions to other frameworks providing effector function, such as BiTEs and scFv-CAR-NKs. IL13-binding CARs have been generated using mutated forms of IL13 as CAR binding domains, but these CARs also recognize IL13Rα1, raising significant toxicity concerns.

To overcome this limitation, a high-affinity IL13Rα2-specific scFv that does not recognize IL13Rα1 was generated. A panel of IL13Rα2-CARs were agented that contain the IL13Rα2-specific scFv as an ectodomain, a short hinge (SH) or a long hinge (LH), a CD28 transmembrane domain, and endodomains that contain signaling domains derived from CD3ζ and co-stimulatory molecules (e.g., CD28.ζ, CD137.ζ, CD28.CD137.ζ, CD28.CD134.ζ). IL13Rα2-CAR T cells were generated by retroviral transduction, and effector function was determined in vitro, using co-culture and cytotoxicity assays, and in vivo, using the U373 brain xenograft model (FIG. 21).

Expression of all CARs in T cells was similar, as judged by Western blot analyses. CAR cell-surface expression varied, however, depending on the hinge and endodomain of the agent. In cytotoxicity assays, the various IL13Rα2-CAR T cells only killed target cells that expressed IL13Rα2 and not IL13Rα1, confirming specificity (FIG. 18). While all IL13Rα2-CAR T cells secreted significant levels of IFNγ in co-culture assays with the IL13Rα2+ glioma cell line U373 (FIG. 19), only short-hinge CAR T cells secreted significant amounts of IL2 (FIG. 20). T cells expressing IL13Rα2-CARs with a deleted endodomain (IL13Rα2Δ-CAR) secreted no cytokines, confirming that cytokine production depends on the presence of a functional IL13Rα2-CAR. In vivo, injection of IL13Rα2.SH.CD28.ζ-CAR T cells into U373-bearing mice resulted in regression of glioma xenografts, as judged by bioluminescence imaging (FIG. 21). IL13Rα2.LH.CD28.ζ- or IL13Rα2.Δ-CAR T cells had no antitumor effects. The data establish that a CAR that only recognizes IL13Rα2 and not IL13Rα1 was generated, and that CAR preferentially targets tumor cells expressing IL13Rα2. Comparison of several IL13Rα2-CARs revealed that a CAR with a SH and a CD28.ζ endodomain resulted in significant T cell activation, as judged by IL2 production and in vivo anti-glioma activity. The results show that adoptive immunotherapy of primary human brain tumors, e.g., high-grade gliomas, in children is both feasible and promising.

Example 13

Generation of 47-CAR T Cells

Two retroviral vectors encoding CARs based on scFv47 (47-CARs; FIG. 31A)24,25 were initially generated. Both CARs contained an N-terminal leader sequence, a codon-optimized synthetic gene encoding scFv47, a spacer region, a CD28 transmembrane domain, and signaling domains derived from CD28 and CD3.ζ (FIG. 31A). The spacer region was either the IgG1 hinge (16 amino acids; short spacer region (SSR); 47-CAR.SSR.CD28.ζ) or the IgG1-CH2CH3 domain (293 amino acids; long spacer region (LSR); 47-CAR.LSR.CD28.ζ). As controls, LSR and SSR 47-CARs without signaling domains were constructed (47-CAR.SSR.Δ, 47-CAR.LSR.Δ; FIG. 31A). CD3/CD28-activated T cells from healthy donors were transduced with RD114-pseudotyped retroviral particles, and 4 to 5 days post-transduction, T-cell phenotype and CAR expression was determined by FACS analysis. CARs were expressed on the cell surface, and the transduction efficiency ranged from 69.2%-98.5% with no significant differences between constructs (FIG. 31B, C). Expression of full-length 47-CAR.SSR.CD28.ζ and 47-CAR.LSR.CD28.ζ was confirmed by Western blot using a CD3.ζ antibody for detection (FIG. 31D). Phenotypic analysis revealed a mixture of CD4- and CD8-positive T cells. While the ratio of CD8- to CD4-positive T cells was about 3:1 for 47-CAR.SSR.CD28.ζ, 47-CAR.SSR.Δ, and 47-CAR.LSR.Δ T-cell lines, it was about 1.5:1 for 47-CAR.LSR.CD28.ζ (FIG. 2).

Example 14

47-CAR T Cells Only Recognize IL13Rα2

To initially determine the specificity of 47-CARs, T cells expressing 47-CAR.SSR.CD28.ζ, 47-CAR.LSR.CD28.ζ, M47-CAR.SSR.Δ, or M47-CAR.LSR.Δ were cultured on tissue culture plates that were uncoated or coated with recombinant proteins encoding IL13Rα1, IL13Rα2, or IL4R. Non-transduced (NT) T cells and T cells expressing an IL13mutein-CAR.LSR.CD28.ζ10 that recognizes IL13Rα1 and IL13Rα2, served as controls. T cells expressing 47-CAR.SSR.CD28.ζ or 47-CAR.LSR.CD28.ζ produced significant levels of IFNγ (p<0.001) when stimulated with recombinant IL13Rα2 proteins in comparison to IL13Rα1- or IL4R-stimulated T cells (FIG. 33A). In contrast, T cells expressing 47-CAR.SSR.Δ or 47-CAR.LSR.Δ produced no IFNγ in response to all three proteins, indicating that IFNγ production depends on an intact 47-CAR signaling domain. 47-CAR.LSR.CD28.ζ T cells also produced low levels of IFNγ without activation, indicating baseline T-cell activation, which was confirmed by intracellular staining for phosphorylated CD3.ζ (FIG. 34). IL13mutein-CAR.LSR.CD28.ζ T cells produced significant levels of IFNγ in the presence of IL13Rα (p<0.001) and IL13Rα2 (p<0.05) in comparison to NT T cells.

The specificity of 47-CAR T cells was then confirmed using cell lines that were negative for IL13Rα1 and IL13Rα2 (Raji), positive for IL13Rα1 (293T-GFP cells), or positive for IL13Rα1 and IL13Rα2 (U373, 293T-GFP/IL13Rα2; FIG. 35). T cells expressing 47-CAR.SSR.CD28.ζ, 47-CAR.LSR.CD28.ζ, 47-CAR.SSR.Δ, or 47-CAR.LSR.Δ were co-cultured with Raji, 293T-GFP, or 293T-GFP/IL13Rα2 cells. NT T cells served as controls. After 24 hours, media was collected and the concentrations of IFNγ and IL2 were determined by ELISA. 47-CAR.SSR.CD28.ζ and 47-CAR.LSR.CD28.ζ T cells produced significant amounts of IFNγ only in the presence of U373 or 293T-GFP/IL13Rα2 cells (FIG. 33B) with SSR.CAR T cells producing significantly more IFNγ than LSR.CAR T cells (p<0.001). 47-CAR.SSR.CD28.ζ T cells produced also significant amounts of IL2 in the presence of 293T-GFP/IL13Rα2 and U373 cells, while 47-CAR.LSR.CD28.ζ T cells did not (FIG. 33C). NT-T cells and T cells expressing 47-CAR.SSR.Δ or 47-CAR.LSR.Δ produced no IFNγ or IL2 in response to any target cells. Finally, we confirmed the specificity of 47-CAR T cells in standard cytotoxicity assays using Raji, 293T-GFP, 293T-GFP/IL13Rα2, and U373 as targets (FIG. 33D).

Example 15

Generation of Short Spacer Region (SSR) 47-CARs with CD28.OX40/41BB

While the results described above demonstrated that 47-CAR T cells only recognize IL13Rα2, as judged by cytokine production and cytolytic activity, the results also highlighted differences between LSR and SSR 47-CARs. Because only 47-CAR.SSRs produced IL2 in the presence of IL13Rα2-positive target cells, the focus in the next set of experiments was shifted to 47-CARs with SSRs, and additional CARs were generated with CD28.OX40.ζ, CD28.41BB.ζ or 41BB.ζ endodomains (FIG. 36A). CAR T cells were generated by retroviral transduction and CAR expression was determined by FACS analysis (FIG. 36B, C) and Western blot (FIG. 36D). While all CARs were expressed, as judged by Western blot analysis, 47-CAR.SSR.CD28.41BB.ζ was not expressed on the cell surface, and was excluded from further analysis.

Example 16

Comparison of Short Spacer Region 47-CARs

To compare the ability of 47-CAR.SSR T cells to produce IFNγ and IL2 in response to antigen exposure, co-culture assays were performed with U373 cells. T cells expressing 47-CAR.SSR.Δ served as controls. All 47-CAR.SSRs with functional endodomains induced IFNγ and IL2 production in the presence of U373 cells; however, 47-CAR.SSR.41BB.ζ T cells produced significantly less (p<0.05) IFNγ in comparison to 47-CAR.SSR.CD28.ζ and 47-CAR.SSR.CD28.OX40.ζ T cells (FIG. 37A). 47-CAR.SSR.CD28.ζ T cells produced the highest amount of IL2, followed by 47-CAR.SSR.41BB.ζ and 47-CAR.SSR.CD28.OX40.ζ T cells. In cytotoxicity assays, no significant difference was observed between all three constructs using Raji, 293T-GFP, 293T-GFP/IL13Rα2, and U373 cells as targets (FIG. 37B).

Because all three 47-CAR.SSRs T cells with functional endodomains produced 1L2, all three constructs were tested in an orthotopic U373 glioma xenograft mouse model in which T cells are directly injected into tumors.6 The model allows for serial bioluminescence imaging because U373 cells are genetically modified to express an eGFP.ffLuc fusion protein (U373.eGFP.ffLuc). On day 0, U373.eGFP.ffLuc cells were injected stereotactically into brains of SCID mice and, on day 7, T cells expressing 47-CAR.SSR.CD28.ζ, 47-CAR.SSR.41BB.ζ, 47-CAR.SSR.CD28.OX40.ζ or 47-CAR.SSR.Δ were injected intratumorally. While mice treated with 47-CAR.SSR.Δ T cells showed continuous tumor growth within 4 days of T-cell injection, mice treated with 47-CAR.SSR T cells that had functional endodomains did not (FIG. 38A, B). Comparison of bioluminescence imaging results revealed no significant difference between 47-CAR.SSR.Δ T cells and the 47-CAR.SSR T cells groups on the day of T-cell injection. Mice treated with 47-CAR.SSR.CD28.ζ or 47-CAR.SSR.CD28.OX40.ζ T cells, however, had significantly lower tumor signals as early as one day post-treatment in comparison to mice treated with 47-CAR.SSR.Δ T cells (p=0.012; Table 2). This resulted in a significant survival advantage for 47-CAR.SSR.CD28.ζ or 47-CAR.SSR.CD28.OX40.ζ T-cell-treated mice (p=0.0002 and p=0.0092; FIG. 40C). While 47-CAR.SSR.41BB.ζ T-cell-treated mice responded slower, resulting in a significant difference between 47-CAR.SSR.Δ T-cell treated on day 14 (p=0.005; Table 2), treatment with this CAR T cell also resulted in a significant survival advantage (p=0.0039; FIG. 5C FIG. 40C). 47-CAR.SSR.CD28.ζ T-cell-treated mice had the longest median survival (84 days). There was no statistical difference, however, in comparison to the median survival of 47-CAR.SSR.41BB.ζ (63 days) or 47-CAR.SSR.CD28.OX40.ζ (56 days) T-cell-treated mice.

TABLE 2 Tumor signal comparison P Day 7 SSR.Δ vs. SSR.41BB.ζ 0.917 SSR.Δ vs. SSR.CD28.ζ 0.111 SSR.Δ vs. SSR.CD28.OX40.ζ 0.917 Day 8 SSR.Δ vs. SSR.41BB.ζ 0.835 SSR.Δ vs. SSR.CD28.ζ 0.012 SSR.Δ vs. SSR.CD28.OX40.ζ 0.023 Day 14 SSR.Δ vs. SSR.41BB.ζ 0.005 SSR.Δ vs. SSR.CD28.ζ 0.015 SSR.Δ vs. SSR.CD28.OX40.ζ 0.015 Day 21 SSR.Δ vs. SSR.41BB.ζ 0.010 SSR.Δ vs. SSR.CD28.ζ 0.010 SSR.Δ vs. SSR.CD28.OX40.ζ 0.012 Day 28 SSR.Δ vs. SSR.41BB.ζ 0.051 SSR.Δ vs. SSR.CD28.ζ 0.008 SSR.Δ vs. SSR.CD28.OX40.ζ 0.034 Wilcoxon rank-sum test

While 47-CAR T cells had potent anti-glioma activity, mice developed recurrent gliomas. To investigate the etiology of tumor recurrence, U373 cells were isolated from two tumor-bearing mice that had been treated either with 47-CAR.SSR.CD28.ζ or 47-CAR.SSR.CD28.OX40.ζ T cells. FACS analysis after short-term culture revealed cell surface expression of IL13Rα2, and these cells were readily killed by 47-CAR T cells in cytotoxicity assays (FIG. 39). Next, the persistence of T-cells was determined by genetically modifying T cells with 47-CAR.SSR.CD28.ζ and eGFP.ffLuc (Luc/47-CAR T cells), and injecting them into U373 tumor-bearing mice. T cells persisted for less than 7 days. Without wishing to be bound by theory, limited persistence appears to be the most likely explanation for tumor recurrence (FIG. 40).

REFERENCES FOR EXAMPLES 13-16

  • 1. Okada, H, Kohanbash, G, Zhu, X, Kastenhuber, E R, Hoji, A, Ueda, R et al. (2009). Immunotherapeutic approaches for glioma. Crit Rev Immunol 29: 1-42.
  • 2. Suryadevara, C M, Verla, T, Sanchez-Perez, L, Reap, E A, Choi, B D, Fecci, P E et al. (2015). Immunotherapy for malignant glioma. Surg Neurol Int 6: S68-S77.
  • 3. Van Gool, S W (2015). Brain Tumor Immunotherapy: What have We Learned so Far?Front Oncol 5: 98.
  • 4. Reardon, D A, Freeman, G, Wu, C, Chiocca, E A, Wucherpfennig, K W, Wen, P Y et al. (2014). Immunotherapy advances for glioblastoma. Neuro Oncol 16: 1441-1458.
  • 5. Ahmed, N, Salsman, V S, Kew, Y, Shaffer, D, Powell, S, Zhang, Y J et al. (2010). HER2-specific T cells target primary glioblastoma stem cells and induce regression of autologous experimental tumors. Clin Cancer Res 16: 474-485.
  • 6. Chow, K K, Naik, S, Kakarla, S, Brawley, V S, Shaffer, D R, Yi, Z et al. (2013). T Cells Redirected to EphA2 for the Immunotherapy of Glioblastoma. Mol Ther 21: 629-637.
  • 7. Johnson, L A, Scholler, J, Ohkuri, T, Kosaka, A, Patel, P R, McGettigan, S E et al. (2015). Rational development and characterization of humanized anti-EGFR variant III chimeric antigen receptor T cells for glioblastoma. Sci Transl Med 7: 275ra22.
  • 8. Brown, C E, Badie, B, Barish, M E, Weng, L, Ostberg, J R, Chang, W C et al. (2015). Bioactivity and Safety of IL13Ralpha2-Redirected Chimeric Antigen Receptor CD8+ T cells in Patients with Recurrent Glioblastoma. Clin Cancer Res
  • 9. Brown, C E, Starr, R, Aguilar, B, Shami, A F, Martinez, C, D'Apuzzo, M et al. (2012). Stem-like tumor-initiating cells isolated from IL13Ralpha2 expressing gliomas are targeted and killed by IL13-zetakine-redirected T Cells. Clin Cancer Res 18: 2199-2209.
  • 10. Krebs, S, Chow, K K, Yi, Z, Rodriguez-Cruz, T, Hegde, M, Gerken, C et al. (2014). T cells redirected to interleukin-13Ralpha2 with interleukin-13 mutein-chimeric antigen receptors have anti-glioma activity but also recognize interleukin-13Ralpha1. Cytotherapy 16: 1121-1131.
  • 11. Kong, S, Sengupta, S, Tyler, B, Bais, A J, Ma, Q, Doucette, S et al. (2012). Suppression of human glioma xenografts with second-generation IL13R-specific chimeric antigen receptor-modified T cells. Clin Cancer Res 18: 5949-5960.
  • 12. Choi, B D, Suryadevara, C M, Gedeon, P C, Herndon Ii, J E, Sanchez-Perez, L, Bigner, D D et al. (2013). Intracerebral delivery of a third generation EGFRvIII-specific chimeric antigen receptor is efficacious against human glioma. J Clin Neurosci.
  • 13. Kawakami, M, Kawakami, K, Takahashi, S, Abe, M, Puri, R K (2004). Analysis of interleukin-13 receptor alpha2 expression in human pediatric brain tumors. Cancer 101: 1036-1042.
  • 14. Okada, H, Low, K L, Kohanbash, G, McDonald, H A, Hamilton, R L, Pollack, I F (2008). Expression of glioma-associated antigens in pediatric brain stem and non-brain stem gliomas. J Neurooncol 88: 245-250.
  • 15. Joshi, B H, Puri, R A, Leland, P, Varricchio, F, Gupta, G, Kocak, M et al. (2008). Identification of interleukin-13 receptor alpha2 chain overexpression in situ in high-grade diffusely infiltrative pediatric brainstem glioma. Neuro Oncol 10: 265-274.
  • 16. Brown, C E, Warden, C D, Starr, R, Deng, X, Badie, B, Yuan, Y C et al. (2013). Glioma IL13Ralpha2 is associated with mesenchymal signature gene expression and poor patient prognosis. PLoS One 8: e77769.
  • 17. Hsi, L C, Kundu, S, Palomo, J, Xu, B, Ficco, R, Vogelbaum, M A et al. (2011). Silencing IL-13Ralpha2 promotes glioblastoma cell death via endogenous signaling. Mol Cancer Ther 10: 1149-1160.
  • 18. Fichtner-Feigl, S, Strober, W, Kawakami, K, Puri, R K, Kitani, A (2006). IL-13 signaling through the IL-13alpha2 receptor is involved in induction of TGF-beta1 production and fibrosis. Nat Med 12: 99-106.
  • 19. Dotti, G, Gottschalk, S, Savoldo, B, Brenner, M K (2014). Design and development of therapies using chimeric antigen receptor-expressing T cells. Immunol Rev 257: 107-126.
  • 20. Sadelain, M, Brentjens, R, Riviere, I (2013). The basic principles of chimeric antigen receptor design. Cancer Discov 3: 388-398.
  • 21. Jensen, M C, Riddell, S R (2015). Designing chimeric antigen receptors to effectively and safely target tumors. Curr Opin Immunol 33: 9-15.
  • 22. Brown, C E, Starr, R, Aguilar, B, Shami, A, Martinez, C, D'Apuzzo, M et al. (2012). Stem-like tumor initiating cells isolated from IL13Ralpha2-expressing gliomas are targeted and killed by IL13-zetakine redirected T cells. Clin Cancer Res
  • 23. Kahlon, K S, Brown, C, Cooper, L J, Raubitschek, A, Forman, S J, Jensen, M C (2004). Specific recognition and killing of glioblastoma multiforme by interleukin 13-zetakine redirected cytolytic T cells. Cancer Res 64: 9160-9166.
  • 24. Balyasnikova, I V, Wainwright, D A, Solomaha, E, Lee, G, Han, Y, Thaci, B et al. (2012). Characterization and immunotherapeutic implications for a novel antibody targeting interleukin (IL)-13 receptor alpha2. J Biol Chem 287: 30215-30227.
  • 25. Kim, J, Young, J, Solomaha, E, Kanojia, D, Lesniak, M S, Balyasnikova, I V (2015). A novel single-chain antibody redirects adenovirus to IL13Rα2-expressing brain tumors. (submitted for publication)
  • 26. Brown, C E, Starr, R, Naranjo, A, Wright, C, Bading, J, Ressler, J et al. (2011). Adoptive Transfer of Autologous IL13-zetakine+ Engineered T Cell Clones for the Treatment of Recurrent Glioblastoma: Lessons from the Clinic. Molecular Therapy 19: S136-S137.
  • 27. Beard, R E, Abate-Daga, D, Rosati, S F, Zheng, Z, Wunderlich, J R, Rosenberg, S A et al. (2013). Gene expression profiling using nanostring digital RNA counting to identify potential target antigens for melanoma immunotherapy. Clin Cancer Res 19: 4941-4950.
  • 28. Kunwar, S, Prados, M D, Chang, S M, Berger, M S, Lang, F F, Piepmeier, J M et al. (2007). Direct intracerebral delivery of cintredekin besudotox (IL13-PE38QQR) in recurrent malignant glioma: a report by the Cintredekin Besudotox Intraparenchymal Study Group. J Clin Oncol 25: 837-844.
  • 29. Okada, H, Kalinski, P, Ueda, R, Hoji, A, Kohanbash, G, Donegan, T E et al. (2011). Induction of CD8+ T-cell responses against novel glioma-associated antigen peptides and clinical activity by vaccinations with (alpha)-type 1 polarized dendritic cells and polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose in patients with recurrent malignant glioma. J Clin Oncol 29: 330-336.
  • 30. Iwami, K, Shimato, S, Ohno, M, Okada, H, Nakahara, N, Sato, Y et al. (2012). Peptide-pulsed dendritic cell vaccination targeting interleukin-13 receptor alpha2 chain in recurrent malignant glioma patients with HLA-A*24/A*02 allele. Cytotherapy
  • 31. Kioi, M, Seetharam, S, Puri, R K (2008). Targeting IL-13Ralpha2-positive cancer with a novel recombinant immunotoxin composed of a single-chain antibody and mutated Pseudomonas exotoxin. Mol Cancer Ther 7: 1579-1587.
  • 32. Haso, W, Lee, D W, Shah, N N, Stetler-Stevenson, M, Yuan, C M, Pastan, I H et al. (2013). Anti-CD22-chimeric antigen receptors targeting B-cell precursor acute lymphoblastic leukemia. Blood 121: 1165-1174.
  • 33. Hudecek, M, Lupo-Stanghellini, M T, Kosasih, P L, Sommermeyer, D, Jensen, M C, Rader, C et al. (2013). Receptor affinity and extracellular domain modifications affect tumor recognition by ROR1-specific chimeric antigen receptor T cells. Clin Cancer Res 19: 3153-3164.
  • 34. Guest, R D, Hawkins, R E, Kirillova, N, Cheadle, E J, Arnold, J, O'Neill, A et al. (2005). The role of extracellular spacer regions in the optimal design of chimeric immune receptors: evaluation of four different scFvs and antigens. J Immunother 28: 203-211.
  • 35. Kunkele, A, Johnson, A J, Rolczynski, L S, Chang, C A, Hoglund, V, Kelly-Spratt, K S et al. (2015). Functional Tuning of CARs Reveals Signaling Threshold above Which CD8+ CTL Antitumor Potency Is Attenuated due to Cell Fas-FasL-Dependent AICD. Cancer Immunol Res 3: 368-379.
  • 36. Hoyos, V, Savoldo, B, Quintarelli, C, Mahendravada, A, Zhang, M, Vera, J et al. (2010). Engineering CD19-specific T lymphocytes with interleukin-15 and a suicide gene to enhance their anti-lymphoma/leukemia effects and safety. Leukemia 24: 1160-1170.
  • 37. Wels, W, Harwerth, I M, Zwickl, M, Hardman, N, Groner, B, Hynes, N E (1992). Construction, bacterial expression and characterization of a bifunctional single-chain antibody-phosphatase fusion protein targeted to the human erbB-2 receptor. Biotechnology (N Y) 10: 1128-1132.
  • 38. Pule, M A, Straathof, K C, Dotti, G, Heslop, H E, Rooney, C M, Brenner, M K (2005). A chimeric T cell antigen receptor that augments cytokine release and supports clonal expansion of primary human T cells. Mol Ther 12: 933-941.
  • 39. Vera, J, Savoldo, B, Vigouroux, S, Biagi, E, Pule, M, Rossig, C et al. (2006). T lymphocytes redirected against the kappa light chain of human immunoglobulin efficiently kill mature B lymphocyte-derived malignant cells. Blood 108: 3890-3897.
  • 40. Xu, Y, Zhang, M, Ramos, C A, Durett, A, Liu, E, Dakhova, O et al. (2014). Closely related T-memory stem cells correlate with in vivo expansion of CAR.CD19-T cells and are preserved by IL-7 and IL15. Blood 123: 3750-3759.
  • 41. Gottschalk, S, Edwards, O L, Sili, U, Huls, M H, Goltsova, T, Davis, A R et al. (2003). Generating CTL against the subdominant Epstein-Barr virus LMP1 antigen for the adoptive Immunotherapy of EBV-associated malignancies. Blood 101: 1905-1912.

Example 17

Transgenic Expression of IL15 Increases 47-CAR T-Cell Persistence Resulting in Enhanced Anti-Tumor Activity

IL13Rα2-CAR.CD28.ζ T cells expressing IL15 (IL13Rα2-CAR.IL15 T cells) were generated by double transducing T cells with retroviruses containing expression cassettes encoding i) IL13Rα2-CAR.CD28.ζ or ii) IL15, A Nerve Growth Factor Receptor (ΔNGFR), and inducible Caspase 9 (iC9) separated by 2A sequences. Suitable 2A sequences include any 2A sequence known in the art, as exemplified by the 2A amino acid sequence from porcine teschovirus-1 (SEQ ID NO:109) encoded by the polynucleotide sequence set forth as SEQ ID NO:110, the 2A amino acid sequence from Thoseaasigna virus (SEQ ID NO:111) encoded by the polynucleotide sequence set forth as SEQ ID NO:112, the 2A amino acid sequence from Equine rhinitis A virus (ERAV) (SEQ ID NO:113) encoded by the polynucleotide sequence set forth as SEQ ID NO:114, or the 2A amino acid sequence from Foot and Mouth Disease Virus (FMDV) (SEQ ID NO:115) encoded by the polynucleotide sequence set forth as SEQ ID NO:116. Kim et al., PLoS One 6(4):1-8 (2011). The effector function of IL13Rα2-CAR.IL15 T cells was determined in vitro using standard assays, and in the U373 GBM xenograft model.

Double transduction of CD3/CD28-activated T cells resulted in T-cell lines that expressed both transgenes in 45-50% of T cells. At base line, IL13Rα2-CAR.IL15 T cells produced on average 69.5 pg/ml of IL15. Production was significantly increased after CD3 or antigen-specific T-cell stimulation (176.7 μg/ml; n=6; p<0.001). IL13Rα2-CAR.IL15 T cells were as efficient as IL13Rα2-CAR T cells in killing IL13Rα2-positive GBM cells in vitro. After intratumoral injection into U373 glioma-bearing mice, IL13Rα2-CAR.IL15 T cells persisted significantly longer than IL13Rα2-CAR T cells (p<0.05). This resulted in a significant increase in progression-free (98 versus 49 days; p=0.004) and overall survival (p=0.006) of treated mice.

The data disclosed in this Example demonstrate that transgenic expression of IL15 enhances the in vivo persistence of IL13Rα2-CAR T cells, resulting in improved anti-glioma activity.

Each of the references cited herein is hereby incorporated by reference in its entirety or in relevant part, as would be apparent from the context of the citation.

FROM THE DISCLOSURE HEREIN IT WILL BE APPRECIATED THAT, ALTHOUGH SPECIFIC EMBODIMENTS OF THE DISCLOSURE HAVE BEEN DESCRIBED HEREIN FOR PURPOSES OF ILLUSTRATION, VARIOUS MODIFICATIONS MAY BE MADE WITHOUT DEVIATING FROM THE SPIRIT AND SCOPE OF THE DISCLOSURE.

Claims

1-31. (canceled)

32. An IL13Rα2-specific chimeric antigen receptor (CAR) comprising: (i) (SEQ ID NO: 1) NYLMN; (ii) (SEQ ID NO: 2) RIDPYDGDIDYNQNFKD; (III) (SEQ ID NO: 3) GYGTAYGVDY; (iv) (SEQ ID NO: 4) RASESVDNYGISFMN; (v) (SEQ ID NO: 5) AASRQGSG; and (vi) (SEQ ID NO: 6) QQSKEVPWT;

(A) an ectodomain comprising each of the amino acid sequences of:
(B) a spacer region;
(C) a transmembrane domain; and
(D) an endodomain selected from the group consisting of CD3.ζ, CD28.ζ, CD28.OX40.ζ, CD28.41BB.ζ and 41BB.ζ.

33-35. (canceled)

36. The IL13Rα2-specific CAR of claim 32 wherein the spacer region comprises SEQ ID NO:103, SEO ID NO:35, or SEO ID NO:37.

37. The IL13Rα2-specific CAR of claim 32 wherein the transmembrane domain is the transmembrane domain of CD28 or CD8α.

38. The IL13Rα2-specific CAR of claim 37, wherein the transmembrane domain of CD28 comprises the amino acid sequence of SEQ ID NO: 39.

39. The IL13Rα2-specific CAR of claim 32, wherein the endodomain comprises a signaling domain of one or more of: CD137, CD134, CD27, CD40, ICOS, or Myd88.

40-41. (canceled)

42. The IL13Rα2-specific CAR of claim 32, comprising the amino acid sequence of SEQ ID NO: 47.

43-47. (canceled)

48. The IL13Rα2-specific CAR of claim 32, comprising the amino acid sequence of SEQ ID NO: 53 or SEQ ID NO: 55.

49. A nucleic acid encoding the IL13Rα2-specific CAR of claim 32.

50. (canceled)

51. A vector comprising the nucleic acid of claim 49.

52. (canceled)

53. A host cell comprising the vector of claim 51.

54. The host cell of claim 53, which is a human host cell.

55. The host cell of claim 53, which is a T-lymphocyte or a natural killer cell.

56-59. (canceled)

60. A pharmaceutical composition comprising the IL13Rα2-specific CAR of claim 32.

61. A method of treating a cancer in a subject, comprising administering to the subject a population of cells comprising the IL13Rα2-specific CAR of claim 32, in an amount effective to treat the cancer in the subject.

62-63. (canceled)

64. The method of claim 61, wherein the population of cells are T-lymphocytes or natural killer cells.

65. (canceled)

66. An IL13Rα2 binding agent comprising each of the amino acid sequences of: (a) complementarity determining region 1 (NYLMN (SEQ ID NO: 1)); (b) complementarity determining region 2 (RIDPYDGDIDYNQNFKD (SEQ ID NO: 2)); (c) complementarity determining region 3 (GYGTAYGVDY (SEQ ID NO: 3)); (d) complementarity determining region 4 (RASESVDNYGISFMN (SEQ ID NO: 4)); (e) complementarity determining region 5 (AASRQGSG (SEQ ID NO: 5)); and (f) complementarity determining region 6 (QQSKEVPWT (SEQ ID NO: 6)).

67. The IL13Rα2 binding agent of claim 66, comprising one or both of the amino acid sequences of SEQ ID NO: 7 and/or SEQ ID NO: 8.

68. The IL13Rα2 binding agent of claim 66, wherein the agent comprises a polypeptide of SEQ ID NO: 13.

69. A nucleic acid encoding the IL13Rα2 binding agent of claim 66.

70. A method of treating glioblastoma multiforme or colon cancer in a subject, comprising administering to the subject a binding agent of claim 66, in an amount effective to treat the cancer in the subject.

Patent History
Publication number: 20240025961
Type: Application
Filed: May 1, 2023
Publication Date: Jan 25, 2024
Applicants: The University of Chicago (Chicago, IL), Baylor College of Medicine (Houston, TX)
Inventors: Irina V. BALYASNIKOVA (Chicago, IL), Maciej S LESNIAK (Chicago, IL), Stephen M.G. GOTTSCHALK (Houston, TX)
Application Number: 18/310,069
Classifications
International Classification: C07K 14/725 (20060101); C07K 14/715 (20060101); C07K 16/28 (20060101); C07K 14/705 (20060101); A61P 35/00 (20060101); C12N 15/79 (20060101);