Chimeric antigen receptors targeting tumor antigens

文档序号:1173741 发布日期:2020-09-18 浏览:18次 中文

阅读说明:本技术 靶向肿瘤抗原的嵌合抗原受体 (Chimeric antigen receptors targeting tumor antigens ) 是由 何苗壮 李楠 李丹 于 2018-11-07 设计创作,主要内容包括:描述了编码嵌合抗原受体(CAR)和截短的人表皮生长因子受体(huEGFRt)的核酸构建体。所编码的CAR包括与CD8α铰链区、CD8α跨膜区、4-1BB共刺激结构域和CD3ζ信号传导结构域融合的肿瘤抗原特异性单克隆抗体,例如glypican-3(GPC3)-特异性、GPC2-特异性或间皮素-特异性单克隆抗体。还描述了分离的宿主细胞,例如共表达所公开的CAR和huEGFRt的分离的T细胞。用所公开的CAR构建体转导的T细胞可用于癌症免疫疗法。(Nucleic acid constructs encoding Chimeric Antigen Receptors (CARs) and truncated human epidermal growth factor receptors (huEGFRt) are described. The encoded CAR comprises a tumor antigen-specific monoclonal antibody, such as a glypican-3(GPC3) -specific, GPC 2-specific or mesothelin-specific monoclonal antibody, fused to a CD8a hinge region, a CD8a transmembrane region, a 4-1BB co-stimulatory domain and a CD3zeta signaling domain. Also described are isolated host cells, e.g., isolated T cells, that co-express the disclosed CARs and huEGFRt. T cells transduced with the disclosed CAR constructs can be used for cancer immunotherapy.)

1. A nucleic acid molecule encoding a Chimeric Antigen Receptor (CAR) comprising, in a5 'to 3' direction:

a nucleic acid encoding a first granulocyte-macrophage colony stimulating factor receptor signal sequence (GMCSFRss);

a nucleic acid encoding an antigen-specific antibody or antigen-binding fragment thereof;

a nucleic acid encoding an extracellular hinge region;

a nucleic acid encoding a transmembrane domain;

a nucleic acid encoding an intracellular co-stimulatory domain;

a nucleic acid encoding an intracellular signaling domain;

a nucleic acid encoding a self-cleaving 2A peptide;

a nucleic acid encoding a second GMCSFRss; and

a nucleic acid encoding a truncated human epidermal growth factor receptor (huEGFRT).

2. The nucleic acid molecule of claim 1, wherein the extracellular hinge region comprises a CD8a hinge region or a CD28 hinge region.

3. The nucleic acid molecule of claim 1 or claim 2, wherein the transmembrane domain comprises a CD8a transmembrane domain or a CD28 transmembrane domain.

4. The nucleic acid molecule of any one of claims 1-3, wherein the intracellular co-stimulatory domain comprises a 4-1BB, CD28, ICOS, OX40, CD27, or DAP10 co-stimulatory domain.

5. The nucleic acid molecule of any one of claims 1-4, wherein the intracellular signaling domain comprises a CD3 ζ or FcRIy signaling domain.

6. The nucleic acid molecule of any one of claims 1-5, wherein the extracellular hinge region comprises a CD8a hinge region, the transmembrane domain comprises a CD8a transmembrane domain, the intracellular co-stimulatory domain comprises a 4-1BB co-stimulatory domain, and the intracellular signaling domain comprises a CD3zeta signaling domain.

7. The nucleic acid molecule of claim 6, wherein the nucleic acid encoding the CD8a hinge comprises the sequence of SEQ ID NO. 3.

8. The nucleic acid molecule of claim 6 or claim 7, wherein the nucleic acid encoding the CD8a transmembrane domain comprises the sequence of SEQ ID NO 5.

9. The nucleic acid molecule of any one of claims 6-8, wherein the nucleic acid encoding the 4-1BB co-stimulatory domain comprises the sequence of SEQ ID No. 7.

10. The nucleic acid molecule of any one of claims 6-9, wherein the nucleic acid encoding the CD3zeta signaling domain comprises the sequence of SEQ ID No. 9.

11. The nucleic acid molecule of any one of claims 1-10, wherein the nucleic acid encoding the first GMCSFRss and the nucleic acid encoding the second GMCSFRss each comprise the sequence of SEQ ID No. 1.

12. The nucleic acid molecule of any one of claims 1-11, wherein the self-cleaving 2A peptide is a T2A peptide and the nucleic acid encoding the self-cleaving 2A peptide includes the sequence of SEQ ID No. 11.

13. The nucleic acid molecule of any one of claims 1-12, wherein the nucleic acid encoding the huEGFRT comprises the sequence of SEQ ID NO 13.

14. The nucleic acid molecule of any one of claims 1-13, further comprising a human elongation factor 1 α (EF1 α) promoter sequence 5' to the nucleic acid encoding said first GMCSFRss.

15. The nucleic acid molecule of any one of claims 1-14, wherein the antigen-binding fragment is a single chain variable fragment (scFv) or a single domain antibody.

16. The nucleic acid molecule of any one of claims 1-15, wherein the antibody or antigen-binding fragment specifically binds to a tumor antigen.

17. The nucleic acid molecule of claim 16, wherein said tumor antigen is glypican-3(GPC 3).

18. The nucleic acid molecule of claim 17, wherein the nucleic acid encoding the antibody binding fragment comprises the heavy chain Variable (VH) domain complementarity determining region 1(CDR1), CDR2 and CDR3 nucleic acid sequences of SEQ ID No. 25 and the light chain Variable (VL) domain CDR1, CDR2 and CDR3 nucleic acid sequences of SEQ ID No. 27.

19. The nucleic acid molecule of claim 17 wherein the nucleic acid encoding the antibody binding fragment comprises the sequence of nucleotides 73-807 of SEQ ID NO 15.

20. The nucleic acid molecule of claim 17, wherein the nucleic acid encoding the antibody binding fragment comprises the CDR1, CDR2, and CDR3 nucleic acid sequences of SEQ ID No. 29.

21. The nucleic acid molecule of claim 17, wherein the nucleic acid encoding the antibody binding fragment comprises the sequence of nucleotides 73-420 of SEQ ID No. 17.

22. The nucleic acid molecule of claim 16, wherein said tumor antigen is glypican-2(GPC 2).

23. The nucleic acid molecule of claim 22, wherein the nucleic acid encoding the antibody binding fragment comprises the CDR1, CDR2, and CDR3 nucleic acid sequences of SEQ ID No. 31.

24. The nucleic acid molecule of claim 22, wherein the nucleic acid encoding the antibody binding fragment comprises the sequence of nucleotides 73-432 of SEQ ID No. 19.

25. The nucleic acid molecule of claim 16, wherein the tumor antigen is mesothelin.

26. A vector comprising the nucleic acid molecule of any one of claims 1-25.

27. The vector of claim 26, wherein the vector is a viral vector.

28. The vector of claim 27, wherein the viral vector is a lentiviral vector.

29. An isolated host cell comprising the nucleic acid molecule of any one of claims 1-25 or the vector of any one of claims 26-28.

30. An isolated host cell that co-expresses a Chimeric Antigen Receptor (CAR) and a truncated human epidermal growth factor receptor (huEGFRt), wherein:

the CAR comprises an antigen-specific antibody or antigen-binding fragment thereof, an extracellular hinge region, a transmembrane domain, an intracellular co-stimulatory domain, and an intracellular signaling domain; and

the huEGFRT includes domain III, domain IV and a transmembrane domain from human EGFR, but lacks an Epidermal Growth Factor (EGF) -binding domain and a cytoplasmic domain.

31. The isolated host cell of claim 30, wherein the extracellular hinge region comprises a CD8a hinge region or a CD28 hinge region.

32. The isolated host cell of claim 30 or claim 31, wherein the transmembrane domain comprises a CD8a transmembrane domain or a CD28 transmembrane domain.

33. The isolated host cell of any one of claims 30-32, wherein the intracellular co-stimulatory domain comprises a 4-1BB, CD28, ICOS, OX40, CD27, or DAP10 co-stimulatory domain.

34. The isolated host cell of any one of claims 30-33, wherein the intracellular signaling domain comprises a CD3 ζ or FcRI γ signaling domain.

35. The isolated host cell of any one of claims 30-34, wherein the extracellular hinge region comprises a CD8a hinge region, the transmembrane domain comprises a CD8a transmembrane domain, the intracellular co-stimulatory domain comprises a 4-1BB co-stimulatory domain, and the intracellular signaling domain comprises a CD3zeta signaling domain.

36. The isolated host cell of claim 35, wherein the CD8a hinge region comprises the amino acid sequence of SEQ ID No. 4.

37. The isolated host cell of claim 35 or claim 36, wherein the CD8a transmembrane domain comprises the amino acid sequence of seq id No. 6.

38. The isolated host cell of any one of claims 35-37, wherein the 4-1BB co-stimulatory domain comprises the amino acid sequence of seq id No. 8.

39. The isolated host cell of any one of claims 35-38, wherein the CD3zeta signaling domain comprises the amino acid sequence of SEQ ID No. 10.

40. The isolated host cell of any one of claims 30-39, wherein the huEGFRT comprises the amino acid sequence of SEQ ID NO. 14.

41. The isolated host cell of any one of claims 30-40, wherein the antigen-binding fragment is a single chain variable fragment (scFv) or a single domain antibody.

42. The isolated host cell of claim 41, wherein the antibody or antigen-binding fragment specifically binds to a tumor antigen.

43. The isolated host cell of claim 42 wherein said tumor antigen is glypican-3(GPC 3).

44. The isolated host cell of claim 43, wherein the amino acid sequence of the antigen-binding fragment comprises the heavy chain Variable (VH) domain complementarity determining region 1(CDR1), CDR2 and CDR3 sequences of SEQ ID NO. 26 and the light chain Variable (VL) domain CDR1, CDR2 and CDR3 sequences of SEQ ID NO. 28.

45. The isolated host cell of claim 43 wherein the amino acid sequence of the antibody binding fragment comprises residues 25-269 of SEQ ID NO 16.

46. The isolated host cell of claim 43, wherein the amino acid sequence of the antigen-binding fragment comprises the CDR1, CDR2, and CDR3 sequences of SEQ ID NO. 30.

47. The isolated host cell of claim 43 wherein the amino acid sequence of the antibody binding fragment comprises residues 25-140 of SEQ ID NO. 18.

48. The isolated host cell of claim 42 wherein said tumor antigen is glypican-2(GPC 2).

49. The isolated host cell of claim 48, wherein the amino acid sequence of the antigen-binding fragment comprises the CDR1, CDR2, and CDR3 sequences of SEQ ID NO. 32.

50. The isolated host cell of claim 48, wherein the amino acid sequence of the antibody binding fragment comprises residues 25-144 of SEQ ID NO. 20.

51. The isolated host cell of claim 42 wherein the tumor antigen is mesothelin.

52. The isolated host cell of any one of claims 29-51, wherein the cell is a T lymphocyte.

53. The isolated host cell of claim 52, wherein said T lymphocyte is an autologous T lymphocyte or an allogeneic T lymphocyte.

54. A composition comprising the isolated host cell of any one of claims 29-53 and a pharmaceutically acceptable carrier.

55. A method of treating a GPC 3-positive cancer in a subject, comprising administering to the subject a therapeutically effective amount of an isolated host cell comprising the nucleic acid molecule of any one of claims 17-21, or administering a therapeutically effective amount of the isolated host cell of any one of claims 43-47.

56. The method of claim 55, wherein the GPC 3-positive cancer is hepatocellular carcinoma (HCC), melanoma, clear ovarian cell carcinoma, Yolk Sac Tumor (YST), neuroblastoma, hepatoblastoma, or Wilms tumor.

57. A method of treating a GPC 2-positive cancer in a subject, comprising administering to the subject a therapeutically effective amount of an isolated host cell comprising the nucleic acid molecule of any one of claims 22-24, or administering a therapeutically effective amount of the isolated host cell of any one of claims 48-50.

58. The method of claim 57, wherein the GPC 2-positive cancer is neuroblastoma, acute lymphocytic leukemia, embryonic rhabdomyosarcoma, alveolar rhabdomyosarcoma, Ewing's sarcoma, desmoplastic small round cell tumor, or osteosarcoma.

59. A method of treating a mesothelin-positive cancer in a subject, comprising administering to the subject a therapeutically effective amount of an isolated host cell comprising the nucleic acid molecule of claim 25, or administering a therapeutically effective amount of the isolated host cell of claim 51.

60. The method of claim 59, wherein the mesothelin-positive cancer is mesothelioma, prostate cancer, lung cancer, gastric cancer, squamous cell carcinoma, pancreatic cancer, cholangiocarcinoma, triple negative breast cancer, or ovarian cancer.

61. The method of any one of claims 55-60, wherein the isolated host cell is a T lymphocyte.

62. The method of claim 61, wherein the T lymphocytes are autologous T lymphocytes or allogeneic T lymphocytes.

63. A nucleic acid molecule encoding a Chimeric Antigen Receptor (CAR) comprising, in a5 'to 3' direction:

a nucleic acid encoding a first granulocyte-macrophage colony stimulating factor receptor signal sequence (GMCSFRss);

a nucleic acid encoding an antigen-specific antibody or antigen-binding fragment thereof;

a nucleic acid encoding a CD8a hinge region;

a nucleic acid encoding a transmembrane domain of CD8 a;

a nucleic acid encoding a 4-1BB co-stimulatory domain;

a nucleic acid encoding a CD3zeta signaling domain;

a nucleic acid encoding a self-cleaving 2A peptide;

a nucleic acid encoding a second GMCSFRss; and

a nucleic acid encoding a truncated human epidermal growth factor receptor (huEGFRT).

64. The nucleic acid molecule of claim 63, wherein the nucleic acid encoding the antibody binding fragment comprises the sequence of nucleotides 73-807 of SEQ ID NO. 15, nucleotides 73-420 of SEQ ID NO. 17, or nucleotides 73-432 of SEQ ID NO. 19.

65. The nucleic acid molecule of claim 63 or claim 64, which comprises the nucleotide sequence of SEQ ID NO 15, SEQ ID NO 17 or SEQ ID NO 19.

66. An isolated host cell that co-expresses a Chimeric Antigen Receptor (CAR) and a truncated human epidermal growth factor receptor (huEGFRt), wherein:

the CAR comprises an antigen-specific antibody or antigen-binding fragment thereof, a CD8a hinge region, a CD8a transmembrane domain, a 4-1BB co-stimulatory domain, and a CD3zeta signaling domain; and

the huEGFRT includes domain III, domain IV and a transmembrane domain from human EGFR, but lacks an EGF-binding domain and a cytoplasmic domain.

67. The isolated host cell of claim 66, wherein the amino acid sequence of the antigen-binding fragment comprises residues 25-269 of SEQ ID NO. 16, residues 25-140 of SEQ ID NO. 18, or residues 25-144 of SEQ ID NO. 20.

68. The isolated host cell of claim 66 or claim 67, wherein the amino acid sequence of the CAR comprises residues 25-491 of SEQ ID NO 16, residues 25-362 of SEQ ID NO 18, or residues 25-366 of SEQ ID NO 20, and the amino acid sequence of the huEGFRT comprises SEQ ID NO 14.

Technical Field

The present disclosure relates to chimeric antigen receptors specific for tumor antigens, and their use for cancer immunotherapy.

Government supported credit

The invention was made with government support under item number Z01 BC010891 awarded by the National Institutes of health, National Cancer Institute. The united states government has certain rights in this invention.

Background

Chimeric Antigen Receptors (CARs) consist of an antibody fragment specific for a tumor antigen fused to a transmembrane domain and a T cell signaling moiety. When the receptor is expressed on the surface of a T cell, it mediates binding to the target and activates the T cell, eventually inducing lysis of the target cell. CAR is becoming one of the most promising approaches to the treatment of hematological malignancies (Kochenderfer et al, Blood 119: 2709-. Two CD 19-targeted CARs have been approved in the United states, axicabtagene ciloleucel (Yescatta)TM) And tisagenlecucel (Kymriah)TM) They are useful in the treatment of B-cell non-Hodgkin's lymphoma and B-cell acute lymphocytic leukemia, respectively. Clinical trials are currently being conducted to test various CAR T cell therapies for the treatment of solid tumors (Yuet al, J Hematol Oncol10 (1):78,2017).

Summary of The Invention

Disclosed herein are nucleic acid constructs encoding a Chimeric Antigen Receptor (CAR) and a truncated human epidermal growth factor receptor (huEGFRt). The encoded CAR comprises a tumor antigen-specific monoclonal antibody fragment fused to an extracellular hinge region, a transmembrane region, an intracellular costimulatory domain, and an intracellular signaling domain. huEGFRt includes two EGFR extracellular domains (domain III and domain IV) and one EGFR transmembrane domain, but lacks two membrane-distal extracellular domains and all intracellular domains. Also disclosed are isolated cells, such as T lymphocytes, that co-express the disclosed CARs and huEGFRt. T cells transduced with the CAR constructs can be used for cancer immunotherapy.

Provided herein are nucleic acid molecules encoding a CAR and a huEGFRt. In some embodiments, the nucleic acid molecule comprises in the 5 'to 3' direction a nucleic acid encoding a first signal sequence; a nucleic acid encoding an antigen-specific antibody or antigen-binding fragment thereof; a nucleic acid encoding an extracellular hinge region; a nucleic acid encoding a transmembrane domain; a nucleic acid encoding an intracellular co-stimulatory domain; a nucleic acid encoding an intracellular signaling domain; a nucleic acid encoding a self-cleaving 2A peptide; a nucleic acid encoding a second signal sequence; and a nucleic acid encoding huEGFRT. In some examples, the first and/or second signal sequence is a granulocyte-macrophage colony stimulating factor receptor signal sequence (GMCSFRss), the extracellular hinge region is a CD8a hinge region, the transmembrane domain is a CD8a transmembrane domain, the intracellular costimulatory domain is a 4-1BB costimulatory domain, and the intracellular signaling domain is a CD3 ζ signaling domain. In some examples, the antibody or antigen binding fragment specifically binds to a tumor antigen, such as glypican-3(GPC3), GPC2, or mesothelin. Also provided are vectors, e.g., viral vectors, comprising the nucleic acid molecules disclosed herein. In a specific non-limiting example, the viral vector is a lentiviral vector. Further provided are isolated host cells comprising the nucleic acid molecules disclosed herein.

Also provided are isolated host cells co-expressing a CAR and a huEGFRt. In some embodiments, the CAR comprises an antigen-specific antibody or antigen-binding fragment thereof, an extracellular hinge region, a transmembrane domain, an intracellular costimulatory domain, and an intracellular signaling domain; and/or huEGFRT comprises domain III, domain IV and a transmembrane domain from human EGFR, but lacks an Epidermal Growth Factor (EGF) -binding domain and a cytoplasmic domain. In some examples, the extracellular hinge region comprises a CD8a hinge region, the transmembrane domain comprises a CD8a transmembrane domain, the intracellular co-stimulatory domain comprises a 4-1BB co-stimulatory domain, and the intracellular signaling domain comprises a CD3zeta signaling domain. In some examples, the antibody or antigen binding fragment specifically binds a tumor antigen, such as GPC3, GPC2, or mesothelin.

Further provided are compositions comprising the isolated host cells disclosed herein and a pharmaceutically acceptable carrier. In some embodiments, the isolated host cell is a T lymphocyte.

Further provided are methods of treating a GPC 3-positive cancer, a GPC 2-positive cancer, or a mesothelin-positive cancer in a subject by administering to the subject an isolated host cell disclosed herein. In some embodiments, the isolated host cell is a T lymphocyte, e.g., an autologous T lymphocyte.

The foregoing and other objects, features, and advantages of the invention will become more apparent from the following detailed description, which proceeds with reference to the accompanying drawings.

Drawings

Figure 1 is a schematic of a lentiviral construct for generating a tumor-targeting Chimeric Antigen Receptor (CAR). The lentiviral construct comprises a CAR coding region and a region encoding a truncated human epidermal growth factor receptor (huEGFRt), each preceded by a granulocyte-macrophage colony stimulating factor receptor signal sequence (GMCSFRss). The two regions are separated by a self-cleaving T2A sequence, such that upon expression of the construct, the CAR is cleaved from the huEGFRt. Expression of this construct is driven by the human elongation factor 1 α (EF1 α) promoter. The CAR includes an antigen binding region, a CD8a hinge region, a CD8a Transmembrane (TM) domain, a 4-1BB costimulatory region, and a CD3zeta signaling domain. The huEGFRt comprises two extracellular domains (domain III and domain IV) and one TM domain.

FIGS. 2A-2C are vector maps of the following constructs: pMH288 encoding car.hn3 (fig. 2A), pMH289 encoding car.yphp 7 (fig. 2B) and pMH290 encoding car.lh7 (fig. 2C).

Figures 3A-3C are flow cytograms showing the transduction efficiency of GPC 3-targeted CAR T cells. Transduction efficiency was determined using anti-huEGFRt antibody cetuximab (cetuximab). Lentiviral vectors encoding car. hn3 (fig. 3A) and car. hpyp7 (fig. 3B) transduced 65% and 45.4% of T cells, respectively. (FIG. 3C) control human serum IgG.

Figures 4A-4G are graphs showing the cytotoxicity of GPC 3-targeted CAR T cells against human cell lines. Effectors of 1:2, 1.5:1, 5:1 and 16:1 were used: target ratio at GPC3+G1 cells (FIG. 4A), GPC3+Hep3B cells (FIG. 4B), GPC3+HepG2 cell (FIG. 4C), GPC3+Huh7 cells (FIG. 4D), GPC3-A431 cells (FIG. 4E), GPC3-T3M4 cells (FIG. 4F) and GPC3-Car. hpyp7 was tested on IMR32 cells (fig. 4G). Hhyp7 is cytotoxic to GPC 3-positive cell line but not to GPC 3-negative cell line.

Figure 5 is a graph showing that car. hpyp7T cells induce Interferon (IFN) - γ secretion by target GPC-positive Hep3B, Huh7 and G1 tumor cells.

Figure 6 shows bioluminescence images of Hep3B tumor suppression in mice treated with GPC 3-targeted T cells. Mice were injected intravenously with 400 ten thousand Hep3B cells on day 0. On day 10, mice were subjected to a blank injection or injection of PBS, 1000 million car.hn3T cells (HN 3-10M), 1000 million car.hpyp7T cells (hYP 7-10M), 2000 million car.hpyp7T cells (hYP 7-20M), or 4000 million car.hpyp7T cells (hYP 7-40M). Tumor size was measured by bioluminescence imaging.

Fig. 7A-7C are graphs showing that car. hpyp7T cells have durable anti-tumor activity against Hep3B xenograft tumors in mice. (figure 7A) tumor volume of Hep3B tumor-bearing mice treated with PBS treatment, blank treatment, with 1000 million car.hn3T cells, 1000 million car.yhyp7T cells, 2000 million car.yhyp7T cells, or 4000 million car.yhyp7T cells up to 3 weeks after treatment. (figure 7B) tumor volume of Hep3B tumor-bearing mice treated with PBS, 1000 ten thousand car.hn3T cells, 1000 ten thousand car.yhsp 7T cells or 4000 ten thousand car.yhsp 7T cells up to 7 weeks after treatment. (FIG. 7C) survival curves of Hep3B tumor-bearing mice. Mice were injected with PBS, 1000 ten thousand car.yhpp 7T cells or 4000 ten thousand car.yhpp 7T cells 10 days after Hep3B inoculation and the survival period was evaluated for 70 days. Treatment with 4000 million car.

Figures 8A-8D are graphs showing tumor volume of HepG2 xenografted NSG mice treated with mock-treated (figure 8A) or 1000 ten thousand car. In fig. 10A-10C, each row represents a single animal. Figure 8D shows the mean tumor volume for all three treatment groups.

FIG. 9 shows GPC3 mRNA levels in human normal tissue as measured by quantitative real-time PCR. The relative GPC3 levels in different normal tissues were compared to the expression of GPC3 in the placenta.

Figures 10A-10E show the production and expression of GPC3 CAR T cells. (FIG. 10A) schematic structure of the HN3 and hYP7 antibodies binding to the N-leaflet (N-lobe) and C-leaflet (C-lobe), respectively, of mature GPC 3. (figure 10B) schematic representation of a bicistronic lentiviral construct expressing a GPC 3-targeted CAR with huEGFRt using T2A ribosome-hopping sequence. (FIG. 10C) CAR expression on healthy donor-derived T cells transduced with lentiviral particles was analyzed by detecting EGFR expression using flow cytometry. (FIG. 10D) blank T cells and CAR (hYP7) T cells from healthy donors and CD3 of CAR (hYP7) T cells derived from HCC patients+、CD4+And CD8+T cell population analysis. (FIG. 10E) proliferation of CAR (hYP7) T cells in 8 different healthy donors and 4 different HCC patients assessed by trypan blue (trypan blue) exclusion assay.

Figures 11A-11G are graphs showing that GPC 3-targeted CAR T cells killed GPC 3-positive HCC cells in vitro. (FIG. 11A) GPC 3-positive target cells lysed (G1), but GPC 3-negative target cells did not lyse (A431 and T3M4), as measured by luciferase activity. Blank or GPC 3-targeted CAR T cells with luciferase-expressing target cells at the indicated effector (E): target (T) ratios were co-cultured for 24 hours and specific lysis was measured using a fluorescence-based cytolysis assay. (FIGS. 11B-11C) cytolytic activity of CAR (HN3) T cells and CAR (hYP7) T cells from healthy donors (FIG. 11B) or HCC patients (FIG. 11C) 24 hours after co-culture with Hep3B cells. (FIG. 11D) CAR (hYP7) T cells proliferated strongly for 35 days after stimulation with anti-CD 3/CD28 beads. (FIG. 11E) cytolytic activity of CAR (hYP7) T cells at day 14 and day 28 post-activation after 24 hours of coculture with Hep3B cells. (FIG. 11F) cytolytic activity of healthy donor-derived GPC3 specific CAR T cells after 24 h co-culture with HepG2 and Huh-7 cells. (FIG. 11G) GPC 3-targeted CAR T cell-mediated killing of HepG2 cells using IncuCyte zoom assay. HepG2 cells were mixed with CAR T cells at 2: 1E: t ratio incubation was up to 140 hours.

Figure 12 shows the cytokine/chemokine profile and versatility of T cells redirected with GPC 3-CAR. Hep3B and HepG2 cells were compared to GPC 3-targeted CAR T cells at various E: t-ratio co-cultures were performed for 24 hours and the indicated cytokine/chemokine levels in the supernatants were measured using Luminex. The bars are from left to right: blank, hYP7 and HN 3. The mean sum SD is shown. P < 0.05; p < 0.01; p < 0.001.

FIGS. 13A-13F show that targeting GPC3 induces apoptosis of HCC cells by inhibiting Wnt/β -catenin signaling (FIG. 13A) 6 hours after treatment, CAR (hYP7) T cells inhibited β -catenin expression and increased expression of apoptosis markers (cleaved PARP and cleaved caspase-9) in Hep3B cells (FIG. 13B) CAR (hYP7) T cells inhibited β -catenin expression in Hep3B cells in a time-dependent manner (FIG. 13C) after CRISPR/Cas 9-mediated GPC3 knockout, 3 protein expression in Hep3B cells (FIG. 13D) antitumor activity of sgRNA5-2 targeting exon 5 of GPC 3. Thymus-free nu/nu nude mice were inoculated subcutaneously with 5 × 106Hep3B cells. When the average tumor volume reaches 150mm3At that time, mice were treated every other day by intratumoral injection of sgRNA5-2 plasmid or empty vector, 6 injections (fig. 13E) knockout of GPC3 reduced expression of β -catenin in mouse tumors (fig. 13F) serum AFP levels before and after sgRNA5-2 plasmid or empty vector control treatment<0.05;**p<0.01。

FIGS. 14A-14F show schematic diagrams of CAR (hYP7) T cell eradication in a Hep3B peritoneal dissemination (periapical differentiation) xenograft mouse model (FIG. 14A). In 12 days post-tumor cell inoculation, NSG mice bearing Hep3B tumors were injected peritoneally with naive T cells, 5 × 106CAR (HN3) T cell, 5 × 106CAR (hYP7) T FineCell, 10 × 106CAR (hYP7) T cells or 20 × 106CAR (hYP7) T cells for treatment. Tumor burden was monitored by bioluminescence imaging. (FIG. 14B) CAR (hYP7) T cells regressed the established Hep3B xenograft at high dose (2,000 ten thousand cells) and inhibited tumor growth at low dose (500 or 1,000 ten thousand cells). (FIG. 14C) tumor bioluminescence in the mice treated in FIG. 14B, counted as mean photons. (FIG. 14D) Kaplan-Meier survival curves of tumor-bearing mice after treatment with 500 or 2000 million CAR (hYP7) T cells. (FIG. 14E) alpha-fetoprotein levels in sera collected from the panel shown in FIG. 14B two or six weeks after CAR T treatment. Sera from three different mice per group were collected for ELISA analysis. (figure 14F) CAR T cell persistence in xenograft tumor tissues 3 weeks after treatment as measured by digital pcr with microdroplet (ddpcr). Values represent mean ± SD. P<0.05;**p<0.01;***p<0.001。

FIGS. 15A-15D show that CAR (hYP7) T cells eliminate tumor cells in a HepG2 peritoneal disseminated xenograft mouse model (FIG. 15A) experimental schematic representation on day 21, NSG mice bearing Hep3B tumors were injected peritoneally with either blank T cells or 20 × 106CAR (hYP7) T cells for treatment. (FIG. 15B) CAR (hYP7) T cells demonstrated potent antitumor activity and mediated eradication of HepG2 xenograft tumors. (FIG. 15C) tumor bioluminescence in the mice treated in FIG. 15B, counted as mean photons. (figure 15D) CAR T cell persistence in xenograft tumor tissues and mouse spleen 5 weeks after treatment as measured by ddPCR.

FIGS. 16A-16D show schematic experimental representations of HCC eradication by CAR (hYP7) T cells in a Hep3B orthotopic xenograft mouse model (FIG. 16A). In day 21, 20 × 10 was injected intraperitoneally or intravenously into Hep3B orthotopic tumor-bearing NSG mice6CAR (hYP7) T cells. Mice were imaged weekly. (FIG. 16B) mice treated with CAR (hYP7) T cells via the tail vein demonstrated tumor eradication, while intraperitoneal treatment resulted in tumor growth inhibition. (FIG. 16C) tumor bioluminescence in the mice treated in FIG. 16B, counted as mean photons. (figure 16D) CAR T cell persistence in tumor tissues and mouse spleen 5 weeks after treatment as measured by ddPCR. Value represents flatMean ± SD. P<0.01。

Figure 17 is a series of flow cytograms and Scatchard plots showing binding of GPC 3-targeted (HN3 and hYP7) Jurkat CAR T cells to GPC 3-human fc (hfc) fusion proteins.

Figure 18 is a series of graphs showing differential cytokine and chemokine secretion measured by Luminex after GPC 3-targeted CAR T cells were incubated with Hep3B and HepG2 tumor cells for 24 hours. The bars are from left to right: blank, hYP7 and HN 3. P <0.05, P <0.01, P < 0.001.

Figures 19A-19B show body weights of Hep3B and HepG2 tumor xenograft mice after treatment with GPC 3-targeted CAR T cells. (figure 19A) body weight of Hep3B tumor model mice after intraperitoneal injection of PBS, blank T-cells, CAR (hYP7) T cells or CAR (HN3) T cells. (FIG. 19B) body weight of HepG2 tumor model mice after intraperitoneal injection of 2000 ten thousand blank T cells or CAR (hYP7) T cells.

Sequence listing

The nucleic acid and amino acid sequences listed in the attached sequence listing are shown using the standard nucleotide base letter abbreviations and amino acid three letter codes as defined in 37 c.f.r.1.822. Only one strand is shown for each nucleic acid sequence, but it is understood that the complementary strand is included by any reference to the strand shown. The sequence listing was filed as an ASCII text file, generated on 29 months 10.8 years 2018, 59.3KB, which is incorporated herein by reference. In the attached sequence listing:

SEQ ID NO 1 is the nucleotide sequence encoding GMCSFRss.

SEQ ID NO 2 is the amino acid sequence of GMCSFRss.

SEQ ID NO 3 is the nucleotide sequence encoding the CD8 α hinge.

SEQ ID NO 4 is the amino acid sequence of the CD8 α hinge.

SEQ ID NO 5 is a nucleotide sequence encoding the transmembrane domain of CD8 a.

SEQ ID NO 6 is the amino acid sequence of the transmembrane domain of CD8 α.

SEQ ID NO 7 is a nucleotide sequence encoding 4-1 BB.

SEQ ID NO 8 is the amino acid sequence of 4-1 BB.

SEQ ID NO 9 is a nucleotide sequence encoding CD3 ζ.

SEQ ID NO 10 is the amino acid sequence of CD3 ζ.

SEQ ID NO 11 is a nucleotide sequence encoding the self-cleaving T2A peptide.

SEQ ID NO 12 is the amino acid sequence of the self-cleaving T2A peptide.

SEQ ID NO 13 is a nucleotide sequence encoding huEGFRT.

SEQ ID NO 14 is the amino acid sequence of huEGFRT.

SEQ ID NO 15 is a nucleotide sequence encoding car.yphp 7 having the following characteristics:

nucleotide 1-66 ═ GMCSFRss coding sequence

Nucleotide 67-72 ═ NdeI restriction sites

Nucleotides 73-807 the humanized YP7 scFv coding sequence

Nucleotide 808-

Nucleotide 814-948 ═ CD8 α hinge region coding sequence

Nucleotide 949-1011-CD 8alpha transmembrane structural domain coding sequence

Nucleotide 1012-1137-4-1 BB costimulatory domain coding sequence

Nucleotide 1138-

Nucleotide 1474-

Nucleotide 1528-

Nucleotide 1594 ═ huEGFRT coding sequence 2598.

16 is the amino acid sequence of car.yphp 7, having the following characteristics:

residue 1-22 ═ GMCSFRss

Residues 23-24 ═ HM (encoded by NdeI restriction sites)

Residue 25-269 ═ humanized YP7 scFv

Residue 270-

Residue 272 ═ 316 ═ CD8 α hinge region

Residue 317-337 ═ CD8a transmembrane domain

Residue 338-containing 379 ═ 4-1BB costimulatory domain

Residue 380-491 ═ CD3zeta signaling domain

Residue 492 ═ 509 self-cleaving T2A peptide

Residue 510-

Residue 532-866 ═ huEGFRt coding sequence.

SEQ ID NO 17 is a nucleotide sequence encoding car. hn3, having the following characteristics:

nucleotide 1-66 ═ GMCSFRss coding sequence

Nucleotide 67-72 ═ NdeI restriction sites

Nucleotide 73-420 ═ HN3 coding sequence

Nucleotide 421-

Nucleotide 427-561 ═ CD8a hinge region coding sequence

Nucleotide 562-624-CD 8alpha transmembrane domain coding sequence

Nucleotide 625-750 ═ 4-1BB costimulatory domain coding sequence

Nucleotide 751-1086 ═ CD3zeta signaling domain coding sequence

Nucleotide 1087-1140 ═ T2A coding sequence

Nucleotide 1141-

Nucleotide 1207-.

18 is the amino acid sequence of car. hn3, with the following features:

residue 1-22 ═ GMCSFRss

Residues 23-24 ═ HM (encoded by NdeI restriction sites)

Residue 25-140 h HN3 single domain antibody

Residue 141-

Residue 143 ═ CD8 α hinge region

Residue 188-

Residue 209-250 ═ 4-1BB costimulatory domain

Residue 251-

Residue 363 ═ self-cleaving T2A peptide

Residue 381 ═ 402 ═ GMCSFRss

Residue 403-.

SEQ ID NO 19 is a nucleotide sequence encoding car.lh7 having the following characteristics:

nucleotide 1-66 ═ GMCSFRss coding sequence

Nucleotide 67-72 ═ NdeI restriction sites

Nucleotide 73-432 as LH7 coding sequence

Nucleotide 433-

Nucleotide 439-573-CD 8-alpha hinge region coding sequence

Nucleotide 574-636 ═ CD8alpha transmembrane domain coding sequence

Nucleotide 637-762 ═ 4-1BB costimulatory domain coding sequence

Nucleotide 763-

Nucleotide 1099-1152 ═ T2A coding sequence

Nucleotide 1153-1218 ═ GMCSFRss coding sequence

Nucleotide 1219-2223 ═ huEGFRT coding sequence.

SEQ ID No. 20 is the amino acid sequence of car.lh7, with the following features:

residue 1-22 ═ GMCSFRss

Residues 23-24 ═ HM (encoded by NdeI restriction sites)

Single domain antibody with residues 25-144 of LH7

Residue 145-

Residue 147 ═ 191-CD 8 α hinge region

Residue 192 ═ 212 ═ CD8 α transmembrane domain

Residue 213-

Residue 255 ═ CD3zeta signaling domain

Residue 367-

Residue 385 ═ GMCSFRss 406-

Residue 407 ═ huEGFRt coding sequence.

SEQ ID NO 21 is the nucleotide sequence of the YP7 VH domain.

SEQ ID NO. 22 is the amino acid sequence of the YP7 VH domain.

SEQ ID NO. 23 is the nucleotide sequence of YP7VL domain.

SEQ ID NO. 24 is the amino acid sequence of the YP7VL domain.

SEQ ID NO 25 is the nucleotide sequence of the hYP7 VH domain.

SEQ ID NO 26 is the amino acid sequence of the hYP7 VH domain.

SEQ ID NO 27 is the nucleotide sequence of the hYP7VL domain.

SEQ ID NO 28 is the amino acid sequence of the hYP7VL domain.

SEQ ID NO. 29 is the nucleotide sequence of the HN3 single domain antibody.

SEQ ID NO 30 is the amino acid sequence of the HN3 single domain antibody.

SEQ ID NO. 31 is the nucleotide sequence of a LH7 single domain antibody.

SEQ ID NO. 32 is the amino acid sequence of a LH7 single domain antibody.

SEQ ID NO 33 is the nucleotide sequence of a LH4 single domain antibody.

SEQ ID NO 34 is the amino acid sequence of a LH4 single domain antibody.

SEQ ID NO 35 is the nucleotide sequence of a LH6 single domain antibody.

SEQ ID NO 36 is the amino acid sequence of a LH6 single domain antibody.

SEQ ID NO 37 is the nucleotide sequence of the YP218 VH domain.

SEQ ID NO 38 is the amino acid sequence of the YP218 VH domain.

SEQ ID NO 39 is the nucleotide sequence of the YP218 VL domain.

SEQ ID NO 40 is the amino acid sequence of the YP218 VL domain.

SEQ ID NO 41 is the nucleotide sequence of the SD1 single domain antibody.

SEQ ID NO 42 is the amino acid sequence of the SD1 single domain antibody.

SEQ ID NOS 43-51 are sgRNA sequences.

Detailed Description

I. Abbreviations

ADCC antibody-dependent cell-mediated cytotoxicity (anti-dependent cell-mediated cytotoxicity)

CAR chimeric antigen receptor (chimeric antigen receptor)

CDR complementary determining region (complementary determining region)

CTL cytotoxic T lymphocyte (cytotoxic T lymphocyte)

ddPCR micro-droplet digital PCR (droplet digital PCR)

DMEM Dulbecco's modified Eagle Medium

EF 1alpha elongation factor 1alpha (elongation factor 1alpha)

EGF epidermal growth factor (epidermal growth factor)

EGFR epidermal growth factor receptor (epidermal growth factor receptor)

ELISA enzyme-linked immunosorbent assay (enzyme-linked immunosorbent assay)

FACS fluorescence activated cell sorting (fluorescent activated cells sorting)

FBS fetal bovine serum (total bone serum)

GPC2 glypican-2

GPC3 glypican-3

GMCSFRss granulocyte-macrophage colony stimulating factor receptor signal sequence (Granulocyte-macrophage colony stimulating factor receptor signaling)

HCC hepatocellular carcinoma (hepatocellular carcinoma)

HLA human leukocyte antigen (human leukocyte antigen)

human truncated epidermal growth factor receptor (human truncated epidermal growth factor receptor)

IFN interferon (interferon)

Ig immunoglobulin (immunoglobulin)

IL Interleukin (Interleukin)

i.p. intraperitoneal (intraperitoneal)

ITAM immune receptor tyrosine activation motif (immunoreceptor tyrosine-based activated motif)

PBMC peripheral blood mononuclear cell (periphytol blood mononar cell)

PBS phosphate buffer (phosphate-buffered saline)

scFv single-chain variable fragment (single-chain variable fragment)

TM transmembrane (transmembrane)

VH or VHHeavy chain variable (variable heavy)

VL or VLLight chain variable (variable light)

YST yolk sac tumor (yolk sac tumor)

Terms and methods

Unless otherwise indicated, technical terms are used according to conventional usage. Definitions of the commonly used terms in molecular biology can be found in the following documents: benjamin Lewis, Genes V, published by Oxford University Press,1994(ISBN 0-19-854287-9); kendrew et al (eds.), The Encyclopedia of molecular biology, published by Blackwell Science Ltd.,1994(ISBN 0-632-02182-9); and RobertA. Meyers (ed.), Molecular Biology and Biotechnology a Comprehensive desk reference, published by VCH Publishers, Inc.,1995(ISBN 1-56081-.

To facilitate review of the various embodiments of the disclosure, the following explanation of specific terms is provided:

4-1 BB: costimulatory molecules expressed by T Cell Receptor (TCR) -activated lymphocytes and other cells, including natural killer cells. Ligation of 4-1BB induces a signaling cascade (signaling cascade) that leads to cytokine production, expression of anti-apoptotic molecules and enhanced immune responses.

Acute lymphocytic leukemia (Acute lymphoblastic leukemia, ALL): acute forms of leukemia are characterized by an overproduction of lymphoblasts. ALL is most common in children, peaking in children 2-5 years of age.

Antibody: a polypeptide ligand comprising at least one variable region which recognizes and binds (e.g. specifically recognizes and specifically binds) an epitope of an antigen. Mammalian immunoglobulin molecules are composed of heavy (H) and light (L) chains, each of which has a variable regionRespectively referred to as heavy chain variable (V)H) Variable domains and light chains (V)L) And (4) a zone. VHRegion and VLThe regions are collectively responsible for binding the antigen recognized by the antibody. There are five major heavy chain classes (or isotypes) of mammalian immunoglobulins that determine the functional activity of the antibody molecules IgM, IgD, IgG, IgA and IgE. Antibody isotypes not found in mammals include IgX, IgY, IgW and IgNAR. IgY is the primary antibody produced by birds and reptiles and is somewhat similar in function to mammalian IgG and IgE. IgW and IgNAR antibodies are produced by cartilaginous fish (cartilaginous fish), while IgX antibodies are found in amphibians.

Antibody variable regions comprise "framework" regions and hypervariable regions, referred to as "complementarity determining regions" or "CDRs". The CDRs are primarily responsible for binding to epitopes of the antigen. The framework regions of the antibody are used to position and align the CDRs in three-dimensional space. Amino acid sequence boundaries of a given CDR can be readily determined using any of a variety of known numbering schemes, including those described in Kabat et Al (Sequences of proteins of Immunological Interest, U.S. department of Health and Humanservices, 1991; the "Kabat" number scheme), Chothia et Al (see Chothia and Lesk, J MolBiol 196: 901-.

A "single-domain antibody" refers to an antibody having a single domain (variable domain) that is capable of specifically binding to an antigen or an epitope of an antigen in the absence of other antibody domains. Single domain antibodies include, for example, VHDomain antibody, VNARAntibody, camelid (camelid) VHH antibody and VLA domain antibody. VNARAntibodies are produced by cartilaginous fish, such as nurse shark (null shark), wishbogong shark, white spot shark (spinydogfish) and bamboo shark (bambooo shark). Camelidae animal VHH antibodies are produced by several species, including camel, camelLlama (llama), alpaca (alpaca), dromedary (dromedary), and guanaco (guanaco), which produce heavy chain antibodies that naturally lack a light chain.

A "monoclonal antibody" is an antibody produced by a single clone of lymphocytes or by cells into which the coding sequence for a single antibody has been transfected. Antibodies are produced by methods known to those skilled in the art. Monoclonal antibodies include humanized monoclonal antibodies.

A "chimeric antibody" has framework residues from one species, e.g., human, and CDRs (which typically confer antigen binding) from another species.

A "humanized" antibody is one that includes a human framework region and one or more CDRs from a non-human (e.g., mouse, rabbit, rat, shark or synthetic) immunoglobulin. The non-human immunoglobulin providing the CDRs is referred to as the "donor" and the human immunoglobulin providing the framework is referred to as the "acceptor". In one embodiment, all CDRs in the humanized immunoglobulin are from a donor immunoglobulin. The constant regions need not be present, but if present, they must be substantially identical to human immunoglobulin constant regions, i.e., have at least about 85-90%, e.g., about 95% or greater identity. Thus, all parts of a humanized immunoglobulin, except perhaps the CDRs, are substantially identical to the corresponding parts of the natural human immunoglobulin sequence. The humanized antibody binds to the same antigen as the donor antibody that provided the CDRs. Humanized or other monoclonal antibodies may have other conservative amino acid substitutions that have substantially no effect on antigen binding or other immunoglobulin function.

Binding affinity: affinity of an antibody for an antigen. In one embodiment, affinity is calculated by the modified Scatchard method described in Frankel et al, mol. Immunol.,16: 101-. In another embodiment, binding affinity is measured by antigen/antibody off-rate. In another embodiment, high binding affinity is measured by a competitive radioimmunoassay. In another embodiment, the binding affinity is measured by ELISA. In another embodiment, antibody affinity is measured by flow cytometry. An antibody that "specifically binds" an antigen (e.g., GPC3) is one that binds that antigen with high affinity and does not significantly bind to other unrelated antigens.

Breast cancer: a cancer forms in breast tissue, which is usually ducts (ducts that deliver milk to the nipple) and lobules (glands that make milk). Triple negative breast cancer refers to a type of breast cancer in which the cancer cells do not express estrogen receptor, progesterone receptor, or significant levels of HER2/neu protein. Triple negative breast cancer is also known as ER negative PR negative HER 2/neu-negative breast cancer.

Chemotherapeutic agents: any chemical agent that has a therapeutic effect in treating a disease characterized by abnormal cell growth. These diseases include tumors, neoplasms (neoplasms) and cancers, as well as diseases characterized by hyperplastic growth, such as psoriasis. In one embodiment, the chemotherapeutic agent is a radioactive compound. The chemotherapeutic agent used can be readily determined by one skilled in the art (see, e.g., Slapak and Kufe, Principles of Cancer Therapy, Chapter 86in Harrison's Principles of Internal Medicine,14th edition; Perry et al, Chemotherpy, Ch.17in Abeloff, Clinical Oncology 2nded.,

Figure BDA0002486745430000091

2000 churchlilllingstone, Inc; baltzer, L., Berkery, R. (eds.): Oncology Pocket Guide to Chemotherapy,2nd ed.St.Louis, Mosby-Year Book, 1995; fischer, D.S., Knobf, M.F., Durivage, H.J, (eds): The Cancer chemother Handbook,4th ed.St.Louis, Mosby-Yeast Book, 1993). Combination chemotherapy is the administration of more than one agent to treat cancer. One example is the administration of CAR T cells used in combination with radioactive or chemical compounds.

Chimeric Antigen Receptor (CAR): a chimeric molecule comprising an antigen binding moiety (e.g., a single domain antibody or scFv) and a signaling domain (e.g., a signaling domain from a T cell receptor (e.g., CD3 ζ)). Typically, a CAR consists of an antigen binding portion, a transmembrane domain, and an intracellular domain. The intracellular domain typically includes a signaling chain with an Immunoreceptor Tyrosine Activation Motif (ITAM), such as CD3 ζ or FcRI γ. In some cases, the intracellular domain further comprises an intracellular portion of at least one other costimulatory domain, e.g., CD28, 4-1BB (CD137), ICOS, OX40(CD134), CD27, and/or DAP 10.

Cholangiocarcinoma (cholangiocancymoma): the type of cancer that occurs in the cells of the inner wall of the hepatobiliary tract.

Complementarity Determining Region (CDR): regions of hypervariable amino acid sequence which determine the binding affinity and specificity of the antibody. The light and heavy chains of mammalian immunoglobulins each have three CDRs, designated L-CDR1, L-CDR2, L-CDR3 and H-CDR1, H-CDR2, H-CDR 3.

Conservative variant (Conservative variant): "conservative" amino acid substitutions are those that do not substantially affect or reduce the affinity of a protein, such as an antibody, for GPC 3. For example, a monoclonal antibody that specifically binds GPC3 can include up to about 1, up to about 2, up to about 5, up to about 10, or up to about 15 conservative substitutions and specifically binds a GPC3 polypeptide. The term "conservative variant" also includes the use of a substituted amino acid in place of an unsubstituted parent amino acid, so long as the variant retains activity. Non-conservative substitutions are those that decrease the activity of the protein.

Conservative amino acid substitutions that provide functionally similar amino acids are well known to those of ordinary skill in the art. The following six groups are examples of amino acids that are considered conservative substitutions for one another:

1) alanine (a), serine (S), threonine (T);

2) aspartic acid (D), glutamic acid (E);

3) asparagine (N), glutamine (Q);

4) arginine (R), lysine (K);

5) isoleucine (I), leucine (L), methionine (M), valine (V); and

6) phenylalanine (F), tyrosine (Y), tryptophan (W).

In some embodiments herein, amino acid sequences are provided that include NO more than 10, NO more than 9, NO more than 8, NO more than 7, NO more than 6, NO more than 5, NO more than 4, NO more than 3, NO more than 2, or NO more than 1 amino acid substitution relative to SEQ ID No. 2, SEQ ID No. 4, SEQ ID No. 6, SEQ ID No. 8, SEQ ID No. 10, SEQ ID No. 12, SEQ ID No. 14, SEQ ID No. 16, SEQ ID No. 18, SEQ ID No. 20, SEQ ID No. 22, SEQ ID No. 24, SEQ ID No. 26, SEQ ID No. 28, SEQ ID No. 30, or SEQ ID No. 32.

Cytotoxic agents: any drug or compound that kills cells.

Cytotoxicity: toxicity of the molecule to the cell that is intended to be targeted (relative to the cells of the rest of the organism).

Degenerate variants: a polynucleotide encoding a polypeptide comprising a sequence that is degenerate as a result of the genetic code. There are 20 natural amino acids, most of which are designated by more than one codon. Thus, all degenerate nucleotide sequences are included as long as the amino acid sequence of the polypeptide is not altered.

Desmoplastic small round cell tumor (DRCT): soft tissue sarcomas occur mainly in childhood, especially in boys. DRCT is an invasive and rare cancer that occurs primarily as an abdominal mass, but can also be found in lymph nodes, the endocardium of the abdomen, the diaphragm, the spleen, the liver, the chest wall, the skull, the spinal cord, the intestine, the bladder, the brain, the lung, the testis, the ovary, and the pelvis.

Epitope: an antigenic determinant. These are specific chemical groups or peptide sequences that are molecularly antigenic, i.e., eliciting specific immune responses. Antibodies specifically bind to a particular epitope on a polypeptide.

Ewing's sarcoma: a rare type of malignancy found in bone or soft tissue. Ewing's sarcoma is a small, blue, round cell tumor.

Framework region (Framework region): amino acid sequence between the CDRs. Framework regions include light chain variable and heavy chain variable framework regions. The framework regions serve to maintain the CDRs in the proper orientation for antigen binding.

Fusion protein: a protein comprising at least a portion of two different (heterologous) proteins.

Glypican-2(GPC 2): members of the six-membered, glypican family of Heparan Sulfate (HS) proteoglycans that are attached to the cell surface by GPI anchors (Filmus et al, Genome Biol 9:224,2008). GPC2 is uniquely expressed in the nervous system (Stipp et al, J Cell Biol124:149-160,1994), is involved in Cell adhesion and is thought to regulate axon growth and guidance. Furthermore, GPC2 mRNA is highly expressed in neuroblastoma and other pediatric cancers (Orentas et al, Front Oncol 2:194,2012). GPC2 is also known as cerebellosan (cerebroglycan) proteoglycan and glypican 2. GPC2 genomic, mRNA, and protein sequences are publicly available (see, e.g., NCBI Gene ID 221914).

GPC 2-positive cancers: a cancer that overexpresses GPC 2. Examples of GPC 2-positive cancers include, but are not limited to, neuroblastoma, acute lymphocytic leukemia, embryonal rhabdomyosarcoma, alveolar rhabdomyosarcoma, ewing's sarcoma, desmoplastic small round cell tumor, or osteosarcoma.

Glypican-3(GPC 3): members of the glypican family of Heparan Sulfate (HS) proteoglycans that are attached to the cell surface by glycosylphosphatidylinositol anchors (Filmus and Selleck, J Clin Invest108:497-501, 2001). The GPC3 gene encodes an approximately 70kD core protein that can be cleaved by furin to produce a 40kD N-terminal fragment and a 30kD C-terminal fragment. Two HS chains are attached at the C-terminal portion of GPC 3. GPC3 and other glypican family proteins play a role in cell division and regulation of cell growth. GPC3 is highly expressed in HCC and some other human cancers, including melanoma, lung squamous cell carcinoma, and ovarian clear cell carcinoma (Ho and Kim, Eur J Cancer 47(3):333-338,2011), but is not expressed in normal tissues. GPC3 is also known as SGB, DGSX, MXR7, SDYS, SGBS, OCI-5, SGBS1, and GTR 2-2.

There are four known isoforms of human GPC3 (isoforms 1-4). The nucleic acid and amino acid sequences of the four isoforms of GPC3 are known, including GenBank accession numbers: NM _001164617 and NP _001158089 (isoform 1); NM _004484 and NP _004475 (isoform 2); NM _001164618 and NP _001158090 (isoform 3); and NM _001164619 and NP _001158091 (isoform 4).

GPC 3-positive cancers: a cancer that overexpresses GPC 3. Examples of GPC 3-positive cancers include, but are not limited to, HCC, melanoma, clear cell carcinoma of the ovary, Yolk Sac Tumor (YST), neuroblastoma, hepatoblastoma, Wilms tumor, squamous cell carcinoma of the lung, testicular non-seminoma germ cell tumor, liposarcoma, cervical intraepithelial neoplasia, adrenal adenoma, schwannoma, and embryonic tumors (Ho and Kim, Eur J Cancer 47(3):333 and 338, 2011; Baumhoe et al., Am JClin Pathol 129(6):899 and 906, 2008; Saikali and Sinnett, Int J Cancer 89(5):418 and 422, 2000).

HAMA (human anti-mouse antibody) response: the immune response of a human subject to the variable and constant regions of a murine antibody that has been administered to the patient. Repeated administration of the antibody may result in an increased rate of clearance of the antibody from the patient's serum and may also cause allergic reactions in the patient.

Hepatocellular carcinoma (Hepatocellular carcinoma, HCC): primary malignancies of the liver typically occur in patients with inflammatory liver caused by viral hepatitis, hepatotoxins or cirrhosis (usually caused by alcoholism). HCC is also known as malignant liver cancer.

Heterologous (heterolous): derived from a single genetic resource or species.

Immune reaction: the response of cells of the immune system (e.g., B cells, T cells, or monocytes) to the stimulus. In one embodiment, the reaction is specific for a particular antigen ("antigen-specific reaction"). In one embodiment, the immune response is a T cell response, e.g., CD4+Reaction or CD8+And (4) reacting. In another embodiment, the response is a B cell response and results in the production of specific antibodies.

Separating: an "isolated" biological component (e.g., a nucleic acid, protein (including antibody), or organelle) has been substantially separated from or purified away from other biological components (i.e., other chromosomal and extrachromosomal DNA and RNA, proteins, and organelles) in the environment (e.g., a cell) in which the component naturally occurs. Nucleic acids and proteins that have been "isolated" include nucleic acids and proteins purified by standard purification methods. The term also encompasses nucleic acids and proteins prepared by recombinant expression in a host cell, as well as chemically synthesized nucleic acids.

Marker (Label): a detectable compound or composition conjugated directly or indirectly to another molecule (e.g., an antibody or protein) to facilitate detection of the molecule. Non-limiting specific examples of labels include fluorescent tags, enzyme linkages, and radioisotopes. In one example, "labeled antibody" refers to the introduction of another molecule into an antibody. For example, the label is a detectable label, e.g., incorporating a radiolabeled amino acid or linked to a biotin moiety polypeptide that is detectable by labeled avidin (e.g., streptavidin containing a fluorescent label or enzymatic activity that is detectable by optical or colorimetric methods). Various methods of labeling polypeptides and glycoproteins are known in the art and can be used. Examples of labels for polypeptides include, but are not limited to, the following: radioisotopes or radionucleotides (e.g. of the type35S、11C、13N、15O、18F、19F、99mTc、131I、3H、14C、15N、90Y、99Tc、111In and125I) fluorescent labels (e.g., Fluorescein Isothiocyanate (FITC), rhodamine, lanthanide fluorescers), enzyme labels (e.g., horseradish peroxidase, β -galactosidase, luciferase, alkaline phosphatase), chemiluminescent labels, biotin groups, predetermined polypeptide epitopes recognized by secondary reporter molecules (e.g., leucine zipper pair sequences, binding sites for secondary antibodies, metal binding domains, epitope tags), or magnetic reagents such as gadolinium chelates.

Linker (Linker): in some cases, the linker is a peptide within an antibody binding fragment (e.g., an Fv fragment) that is used to indirectly bind the variable heavy chain to the variable light chain. "linker" may also refer to a peptide used to link a targeting moiety, such as an antibody, to an effector molecule, such as a cytotoxin, or a detectable label.

The terms "conjugation", "binding" or "linking" refer to bringing two polypeptides into one continuous polypeptide molecule, or covalently attaching a radionuclide or other molecule to a polypeptide, such as an scFv. In a particular context, the term includes reference to the attachment of a ligand, such as an antibody moiety, to an effector molecule. The linkage may be by chemical or recombinant means. "chemical means" refers to a reaction between an antibody moiety and an effector molecule such that a covalent bond is formed between the two molecules to form one molecule.

Lung cancer: in lung tissue, cancer often forms in the airway lining cells. Two major types are small cell lung cancer and non-small cell lung cancer (NSCLC). These types are diagnosed based on the appearance of the cells under the microscope.

Mammals: the term includes human and non-human mammals. Similarly, the term "subject" includes human and veterinary subjects.

Melanoma (Melanoma): a cancer originated from melanocyte (cell producing pigment such as melanin). Melanocytes are found primarily in the skin, but are also present in the intestine and eye. Melanomas in the skin include superficial spreading melanoma, nodular melanoma, acral freckle-like melanoma, and lentigo maligna (melanoma). Any of the above types may produce melanin or may be melanin-free. Also, any subtype may show desmoplasia (with a neurotropic dense fibrous response) which is a hallmark of aggressive behavior and a tendency to local recurrence. Other melanomas include clear cell sarcoma, mucosal melanoma and uveal melanoma.

Mesothelin (Mesothelin): 40kDa cell surface Glycosylphosphatidylinositol (GPI) -linked glycoprotein. Human mesothelin protein is synthesized as a 70kD precursor and then proteolytically processed. The 30kD amino terminus of mesothelin is secreted and is called megakaryocyte enhancing factor (Yamaguchi et al, J.biol.chem.269: 805808,1994). As mature mesothelin, the 40kD carboxy terminus remains membrane bound (Chang et al, Natl.Acad.Sci.USA 93: 136140,1996). Exemplary nucleic acid and amino acid sequences for mesothelin are described in PCT publication nos. WO 97/25,068; U.S. patent No. 6,083,502; chang and Pastan, int.J. cancer 57:90,1994; chang and Pastan, Proc.Natl.Acad.Sci USA 93:136,1996; brinkmann et al, int.j. cancer 71:638,1997; and Chowdhury et al, mol.Immunol.34:9, 1997. Mesothelin also refers to mesothelin proteins or polypeptides that remain within the cell and extracellular mesothelin proteins that are secreted and/or isolated.

Mesothelin-positive cancer: cancers that overexpress mesothelin. Examples of mesothelin-positive cancers include, but are not limited to, mesothelioma, prostate cancer, lung cancer, gastric cancer, squamous cell carcinoma, pancreatic cancer, cholangiocarcinoma, triple negative breast cancer, and ovarian cancer.

Mesothelioma (Mesothelioma): one type of neoplasm, derived from pleural and peritoneal lining cells, grows as a thick sheet covering the internal organs, consisting of spindle cells or fibrous tissue, which may surround a glandular space lined with cuboidal cells. Mesothelioma often originates in the lining tissue of the lung, heart or abdomen. In some cases, mesothelioma is caused by exposure to asbestos.

Neoplasms, malignancies, cancers or tumors (neoplasms, malignancy, cancer or tumor): neoplasms are abnormal growth of tissue or cells due to excessive cell division. Neoplastic growth can produce tumors. The amount of tumor in an individual is the "tumor burden", which can be measured as the number, volume or weight of tumors. Non-metastatic tumors are referred to as "benign". Tumors that invade surrounding tissue and/or can metastasize are referred to as "malignant".

Neuroblastoma (Neuroblastoma): solid tumors arising from embryonic neural crest cells. Neuroblastoma usually occurs in the adrenal gland and its surroundings, but may also occur anywhere where sympathetic nervous tissue is found, for example in nervous tissue near the abdomen, chest, neck or spine. Neuroblastoma usually occurs in children under the age of 5.

Operably linked (operable linked): the first nucleic acid sequence is operably linked to the second nucleic acid sequence when the first nucleic acid sequence is in a functional relationship with the second nucleic acid sequence. For example, a promoter is operably linked to a coding sequence if it affects the transcription or expression of the coding sequence. Generally, operably linked DNA sequences are contiguous and, where necessary to join two protein coding regions, in the same reading frame.

Osteosarcoma (Osteosarcoma): cancerous tumor types found in bone. Osteosarcoma is an invasive cancer that originates from primary transformed cells of mesenchymal origin. This type of cancer is most prevalent in children and young adults.

Ovarian cancer (Ovarian cancer): cancer formed in ovarian (one of a pair of female gonads in which an egg or egg is formed) tissue. Most ovarian cancers are either ovarian epithelial cancers (cancers that start from ovarian surface cells) or malignant germ cell tumors (cancers that start from egg cells).

Clear cell carcinoma of ovary (Ovarian clear cell carcinoma): the unique histopathological subtype of epithelial ovarian cancer occurs at less than 5% of all ovarian malignancies. The interior of this type of tumor cell appears transparent when viewed under a microscope.

Pancreatic cancer (Pancreatic cancer): diseases with malignant (cancerous) cells are found in pancreatic tissue. Also known as exocrine carcinoma.

Pediatric cancer (Pediatric cancer): cancer that occurs in children between 0 and 14 years of age. The major types of pediatric cancers include, for example, neuroblastoma, Acute Lymphocytic Leukemia (ALL), embryonal rhabdomyosarcoma (erm), Alveolar Rhabdomyosarcoma (ARMS), ewing's sarcoma, desmoplastic small round cell tumor (DRCT), osteosarcoma, brain and other CNS tumors, Wilm tumor, non-hodgkin lymphoma, and retinoblastoma.

Medicament: a chemical compound or composition capable of inducing a desired therapeutic or prophylactic effect when properly administered to a subject or cell.

A pharmaceutically acceptable carrier: the pharmaceutically acceptable carriers used are conventional. Remington's pharmaceutical Sciences, by e.w. martin, Mack Publishing co., Easton, PA,15th edition,1975, describe compositions and formulations suitable for pharmaceutical delivery of the compositions disclosed herein.

In general, the nature of the carrier will depend on the particular mode of administration employed. For example, parenteral formulations typically include injectable fluids, which include pharmaceutically and physiologically acceptable fluids, such as water, physiological saline, balanced salt solutions, aqueous dextrose, glycerol, and the like, as vehicles. For solid compositions (e.g., in powder, pill, tablet, or capsule form), conventional non-toxic solid carriers can include, for example, pharmaceutical grades of mannitol, lactose, starch, or magnesium stearate. In addition to the biologically neutral carrier, the pharmaceutical composition to be administered may contain minor amounts of non-toxic auxiliary substances such as wetting or emulsifying agents, preservatives, and pH buffering agents and the like, for example sodium acetate or sorbitan monolaurate.

Prevention, treatment or amelioration of disease (ameliorating): "preventing" a disease refers to inhibiting the overall progression of the disease. "treatment" refers to a therapeutic intervention that ameliorates a sign or symptom of a disease or pathological condition after it has begun to develop, such as reducing tumor burden or reducing the number size of metastases. "improvement" refers to a reduction in the number or severity of signs or symptoms of a disease, such as cancer.

Prostate cancer (Prostate cancer): cancer that develops in the tissues of the prostate gland (the gland of the male reproductive system located below the bladder and in front of the rectum). Prostate cancer commonly occurs in older men.

Purification of: the term "purified" does not require absolute purity, but rather it is intended as a relative term. Thus, for example, a purified peptide preparation is one in which the peptide or protein is enriched to a greater extent than the peptide or protein in the natural environment of the cell. In one embodiment, the preparation is purified such that the protein or peptide comprises at least 50% of the total peptide or protein content of the preparation. Substantially purified refers to purification from other proteins or cellular components. A substantially purified protein is at least 60%, 70%, 80%, 90%, 95%, or 98% pure. Thus, in one specific non-limiting example, a substantially purified protein is 90% free of other proteins or cellular components.

Recombinant: a recombinant nucleic acid is a nucleic acid having a sequence that does not occur naturally or has a sequence made by the artificial combination of two separate fragments of the sequence. Such artificial combination is usually achieved by chemical synthesis or by artificial manipulation of the isolated nucleic acid fragments, for example by genetic engineering techniques.

Rhabdomyosarcoma (Rhabdomyosarcoma, RMS): malignant tumor of soft tissue of skeletal muscle origin. The most common primary sites of rhabdomyosarcoma are the head and neck (e.g., parameningeal, orbital, pharyngeal, etc.), the urogenital tract, and the extremities. Other less common primary sites include the torso, chest wall, abdomen (including retroperitoneal cavity and biliary tract), and perineal/anal regions. There are at least two types of RMS. The most common forms are acinar rms (arms) and embryonic histology rms (erm). Approximately 20% of children with rhabdomyosarcoma have the ARMS subtype. This subtype is found to be of increased frequency in adolescents and in patients whose primary site involves the extremities, trunk and perineal/perianal regions. ARMS is associated with chromosomal translocations encoding fusion genes involved in FKHR and PAX family members on chromosome 13. The embryonic subtype is the most common subtype in children and accounts for about 60-70% of rhabdomyosarcoma in children. Tumors with embryonic histology are usually found in the head and neck or urogenital tract, although they may occur at any primary site. Erm is characterized by a small diagnostic age, loss of heterozygosity and altered genomic imprinting.

Sample (or biological sample): a biological sample comprising genomic DNA, RNA (including mRNA), protein, or a combination thereof obtained from a subject. Examples include, but are not limited to, peripheral blood, tissue, cells, urine, saliva, tissue biopsies, fine needle aspirates, surgical specimens, and autopsy material. In one example, the sample comprises a tumor biopsy, such as a tumor tissue biopsy.

Sequence identity: the similarity between amino acid or nucleic acid sequences is expressed in terms of the similarity between sequences (otherwise known as sequence identity). Sequence identity is often measured in terms of percentage identity (or similarity or homology); the higher the percentage, the more similar the two sequences. Homologues or variants of a polypeptide or nucleic acid molecule will have a considerably higher degree of sequence identity when aligned using standard methods.

Methods of sequence alignment for comparison are well known in the art. Various programs and alignment algorithms are described in: smith and Waterman, adv.Appl.Math.2:482,1981; needleman and Wunsch, J.mol.biol.48:443,1970; pearson and Lipman, proc.natl.acad.sci.u.s.a.85:2444,1988; higgins and Sharp, Gene 73:237,1988; higgins and Sharp, CABIOS 5:151,1989; corpet et al, Nucleic Acids Research 16:10881,1988; and Pearson and Lipman, Proc.Natl.Acad.Sci.U.S.A.85:2444,1988. Altschul et al, Nature Genet.6:119,1994 set forth detailed considerations for sequence alignment methods and homology calculations.

The NCBI Basic Local Alignment Search Tool (NCBI Basic Local Alignment Search Tool, BLAST) (Altschul et al, J.mol.biol.215:403,1990) is available from several sources including the National Center for Biotechnology Information, NCBI, Bethesda, Md.) and the Internet, used in conjunction with the sequence analysis programs blastp, blastn, blastx, tblastn, and tblastx. A description of how to determine sequence identity using this program is available on the NCBI website of the Internet.

V of antibody specifically binding GPC3 polypeptideLOr VHAre generally characterized by having at least about 75%, e.g., at least about 80%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity as calculated from a full-length alignment of the antibody amino acid sequences using NCBI blast2.0, gapped blast set as a default parameter. To compare amino acid sequences greater than about 30 amino acids, the Blast2 sequence function was used using a default BLOSUM62 matrix (gap existence cost of 11 and gap per residue cost of 1) set as default parameters. When aligning short peptides (less than about 30 amino acids), the alignment should be performed using the Blast2 sequence function using the PAM30 matrix set as default parameters (open gap 9, extension gap 1 penalty). Having even higher similarity to a reference sequence when assessed by this methodProteins will exhibit an increased percentage of identity, e.g., at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99% sequence identity. When comparing sequence identity to less than the complete sequence, homologues and variants will typically have at least 80% sequence identity within a short window of 10-20 amino acids, and may have at least 85% or at least 90% or 95% sequence identity, depending on their similarity to the reference sequence. Methods for determining sequence identity over these short windows are available on the NCBI website of the Internet. Those skilled in the art will recognize that these ranges of sequence identity are provided as guidance only; it is entirely possible to obtain strong significant homologues falling outside the ranges provided.

Squamous cell carcinoma (Squamous cell carcinoma): a cancer that originates in squamous cells, thin, flattened cells that form the surface of the skin, eye, various internal organs, and lining of hollow organs and ducts of certain glands. Squamous cell carcinoma is also known as epidermoid carcinoma. One squamous cell carcinoma is squamous cell carcinoma of the lung. Squamous cell carcinoma is the most common type of skin cancer.

Gastric cancer (Stomach cancer): cancer that develops in the stomach wall tissue. Also known as gastric cancer (gastrococcus).

Subject: living multicellular vertebrate organisms, including the classes of human and veterinary subjects, including human and non-human mammals.

The synthesis comprises the following steps: produced by artificial means in the laboratory, for example, synthetic nucleic acids or proteins (e.g., antibodies) which can be chemically synthesized in the laboratory.

A therapeutically effective amount of: an amount of the particular substance sufficient to achieve the desired effect in the subject being treated. For example, this may be the amount required to inhibit or prevent tumor growth. In one embodiment, a therapeutically effective amount is the amount necessary to eliminate, reduce the size of a tumor, or prevent metastasis of a tumor. When administered to a subject, a dose will generally be used that achieves a target tissue concentration (e.g., in a tumor) that has been shown to achieve a desired in vitro effect.

Carrier: a nucleic acid molecule introduced into a host cell, thereby producing a transformed host cell. A vector may include a nucleic acid sequence, such as an origin of replication, that allows it to replicate in a host cell. The vector may also include one or more selectable marker genes and other genetic elements known in the art.

Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. The singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. "comprising A or B" is meant to include A or B, or both A and B. It is also understood that all base sizes or amino acid sizes, as well as all molecular weight or molecular mass values given for a nucleic acid or polypeptide are approximate and provided for descriptive purposes. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present disclosure, particularly suitable methods and materials are described below. In case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.

Overview of several embodiments

Disclosed herein are nucleic acid molecules encoding Chimeric Antigen Receptors (CARs) and truncated human epidermal growth factor receptors (huEGFRt). The encoded CAR comprises a tumor antigen-specific monoclonal antibody fused to an extracellular hinge region, a transmembrane region, an intracellular costimulatory domain, and an intracellular signaling domain. The huEGFRt comprises two EGFR extracellular domains (domain III and domain IV) and one transmembrane domain, but lacks two membrane distal extracellular domains (domain I and domain II) and all intracellular domains (membrane proximal domain, tyrosine kinase domain and C-terminal tail). Also provided are isolated cells, such as T lymphocytes, that co-express the disclosed CARs and huEGFRt. T cells transduced with the CAR construct can be used for cancer immunotherapy.

Provided herein are nucleic acid molecules encoding a CAR and a huEGFRt. In some embodiments, the nucleic acid molecule comprises in the 5 'to 3' direction a nucleic acid encoding a first signal sequence; a nucleic acid encoding an antigen-specific antibody or antigen-binding fragment thereof; a nucleic acid encoding an extracellular hinge region; a nucleic acid encoding a transmembrane domain; a nucleic acid encoding an intracellular co-stimulatory domain; a nucleic acid encoding an intracellular signaling domain; a nucleic acid encoding a self-cleaving 2A peptide; a nucleic acid encoding a second signal sequence; and a nucleic acid encoding huEGFRT.

The first and second signal sequences may be any suitable signal sequence known in the art. The first and second signal sequences may be the same signal sequence or they may be different signal sequences. In some embodiments, the first and/or second signal sequence is a granulocyte-macrophage colony stimulating factor receptor signal sequence (GMCSFRss).

In some embodiments, the extracellular hinge region comprises a CD8a hinge region, a CD28 hinge region, or a sequence from another immunoglobulin molecule such as IgG1, IgG4, or IgD (e.g., from a CH2 and/or CH3 domain of an immunoglobulin molecule). This hinge region is also sometimes referred to in the art as a "spacer region".

In some embodiments, the transmembrane domain comprises a CD8a, CD28, CD3, CD45, CD4, CD5, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, or CD154 transmembrane domain. The transmembrane domain may also be a transmembrane region of the α, β or zeta chain of a T cell receptor.

In some embodiments, the intracellular co-stimulatory domain comprises a 4-1BB (CD137, TNFRSF9), CD28, ICOS, OX40(CD134), CD27, CD30, CD40, PD-1, lymphocyte function-associated antigen 1(LFA-1), CD2, CD7, LIGHT, NKG2C, B7-H3, or DAP10 co-stimulatory domain. In some examples, the intracellular co-stimulatory domain includes 4-1BB and CD 28.

In some embodiments, the intracellular signaling domain is a domain having an Immunoreceptor Tyrosine Activation Motif (ITAM), such as a CD3 ζ or FcRI γ signaling domain.

In a specific embodiment, the first and second signal sequences comprise GMCSFRss, the extracellular hinge region comprises a CD8a hinge region, the transmembrane domain comprises a CD8a transmembrane domain, the intracellular co-stimulatory domain comprises a 4-1BB co-stimulatory domain, and the intracellular signaling domain comprises a CD3zeta signaling domain.

In some examples, the nucleic acid encoding the CD8a hinge comprises a nucleotide sequence that is at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to SEQ ID No. 3. In one non-limiting example, the nucleic acid encoding the CD8a hinge includes the sequence of SEQ ID NO. 3.

In some examples, the nucleic acid encoding the transmembrane domain of CD8a comprises a nucleotide sequence that is at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to SEQ ID No. 5. In one non-limiting example, the nucleic acid encoding the transmembrane domain of CD8 α comprises the sequence of SEQ ID NO. 5.

In some examples, the nucleic acid molecule encoding the 4-1BB co-stimulatory domain comprises a nucleotide sequence that is at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to SEQ ID No. 7. In one non-limiting example, a nucleic acid molecule encoding a 4-1BB co-stimulatory domain includes the sequence of SEQ ID NO. 7.

In some examples, the nucleic acid encoding a CD3zeta signaling domain comprises a nucleotide sequence that is at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to SEQ ID No. 9. In one non-limiting example, the nucleic acid encoding the CD3zeta signaling domain comprises the sequence of SEQ ID NO 9.

In some examples, the nucleic acid encoding the first GMCSFRss and the nucleic acid encoding the second GMCSFRss each comprise a nucleotide sequence that is at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to SEQ ID No. 1. In one non-limiting example, the nucleic acid encoding the first GMCSFRss and the nucleic acid encoding the second GMCSFRss each comprise the sequence of SEQ ID No. 1.

In some examples, the self-cleaving 2A peptide is a thioea asigna virus 2A (T2A) peptide. In other examples, the self-cleaving 2A peptide is a foot and mouth disease virus (F2A) peptide, equine rhinitis a virus 2A (E2A) peptide, or porcine teschovirus-1 (P2A) peptide. In particular examples, a nucleic acid encoding a self-cleaving T2A peptide includes a nucleotide sequence that is at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to seq id No. 11. In one non-limiting example, the nucleic acid encoding the self-cleaving T2A peptide includes the sequence of SEQ ID NO. 11.

In some examples, the nucleic acid encoding huEGFRt comprises a nucleotide sequence that is at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to SEQ ID No. 13. In one non-limiting example, the nucleic acid encoding huEGFRT includes the sequence of SEQ ID NO 13.

In some embodiments, the nucleic acid molecule further comprises a human elongation factor 1alpha (EF 1alpha) promoter sequence 5' to the nucleic acid encoding the first GMCSFRss. However, any suitable promoter sequence may be selected by those skilled in the art.

In some embodiments, the antigen-binding fragment is a single chain variable fragment (scFv) or a single domain antibody.

In some embodiments, the antibody or antigen binding fragment specifically binds to a tumor antigen. In particular examples, the tumor antigen is GPC3, GPC2, or mesothelin.

In some examples, wherein the tumor antigen is GPC3, the nucleic acid encoding the antibody binding fragment comprises the heavy chain Variable (VH) domain complementarity determining region 1(CDR1), CDR2 and CDR3 nucleic acid sequences of SEQ ID NO:25(hYP7 VH domain nucleotide sequence) and the light chain Variable (VL) domain CDR1, CDR2 and CDR3 nucleic acid sequences of SEQ ID NO:27 (hYP7VL domain nucleotide sequence). The CDR sequences can be determined using any well-known numbering scheme, such as IMGT, Kabat, or Chothia. In specific examples, the VH domain CDR1, CDR2 and CDR3 nucleic acid sequences each include nucleotides 91-105, 148-204 and 301-318 of SEQ ID NO. 25; and/or the VL domain CDR1, CDR2 and CDR3 nucleic acid sequences each comprise nucleotides 70-120, 166-186 and 283-309 of SEQ ID NO. 27. In other specific examples, the VH domain CDR1, CDR2 and CDR3 nucleic acid sequences each include nucleotides 76-99, 151-180 and 295-318 of SEQ ID NO. 25; and/or the VL domain CDR1, CDR2 and CDR3 nucleic acid sequences each include nucleotides 79-114, 166-174 and 283-309 of SEQ ID NO. 27. In one non-limiting example, the nucleic acid encoding the antibody binding fragment includes the sequence of nucleotides 73-807 of SEQ ID NO. 15.

In other examples, wherein the tumor antigen is GPC3, the nucleic acid encoding the antibody binding fragment comprises the CDR1, CDR2, and CDR3 nucleic acid sequences of SEQ id No. 29(HN3 single domain antibody nucleotide sequence). The CDR sequences can be determined using any well-known numbering scheme, such as IMGT, Kabat, or Chothia. In a specific example, the CDR1, CDR2 and CDR3 nucleic acid sequences each include nucleotides 91-105, 148-195 and 286-315 of SEQ ID NO. 29. In other specific examples, the CDR1, CDR2 and CDR3 nucleic acid sequences each include nucleotides 76-99, 151-171 and 286-315 of SEQ ID NO. 29. In one non-limiting example, the nucleic acid encoding the antibody binding fragment includes the sequence of nucleotides 73-420 of SEQ ID NO. 17.

In other examples, wherein the tumor antigen is GPC3, the nucleic acid encoding the antibody binding fragment comprises the CDR nucleic acid sequences of a GPC 3-specific monoclonal antibody disclosed in WO2013/181543 or WO 2012/145469, which are incorporated herein by reference in their entirety.

In some examples, wherein the tumor antigen is GPC2, the nucleic acid encoding the antibody binding fragment comprises the CDR1, CDR2, and CDR3 nucleic acid sequences of SEQ id no:31(LH7 single domain antibody nucleotide sequence). The CDR sequences can be determined using any well-known numbering scheme, such as IMGT, Kabat, or Chothia. In a specific example, the CDR1, CDR2 and CDR3 nucleic acid sequences each include nucleotides 91-105, 148-195 and 286-327 of SEQ ID NO. 31. In other specific examples, the CDR1, CDR2 and CDR3 nucleic acid sequences each include nucleotides 76-99, 151-171 and 286-327 of SEQ ID NO. 31. In one non-limiting example, the nucleic acid encoding the antibody binding fragment includes the sequence of nucleotides 73-432 of SEQ ID NO 19.

In other examples, wherein the tumor antigen is GPC2, the nucleic acid encoding the antibody binding fragment comprises the CDR1, CDR2, and CDR3 nucleic acid sequences of SEQ ID NO:33(LH4 single domain antibody nucleotide sequence). The CDR sequences can be determined using any well-known numbering scheme, such as IMGT, Kabat, or Chothia. In a specific example, the CDR1, CDR2 and CDR3 nucleic acid sequences each include nucleotides 91-105, 148-195 and 286-327 of SEQ ID NO. 33. In other specific examples, the CDR1, CDR2 and CDR3 nucleic acid sequences each include nucleotides 76-99, 151-171 and 286-327 of SEQ ID NO. 33.

In other examples, wherein the tumor antigen is GPC2, the nucleic acid encoding the antibody binding fragment comprises the CDR1, CDR2, and CDR3 nucleic acid sequences of SEQ ID NO:35(LH6 single domain antibody nucleotide sequence). The CDR sequences can be determined using any well-known numbering scheme, such as IMGT, Kabat, or Chothia. In a specific example, the CDR1, CDR2 and CDR3 nucleic acid sequences each include nucleotides 91-105, 148-198 and 289-330 of SEQ ID NO 35. In other specific examples, the CDR1, CDR2 and CDR3 nucleic acid sequences each include nucleotides 76-99, 151-174 and 289-330 of SEQ ID NO. 35.

In other examples, wherein the tumor antigen is GPC2, the nucleic acid encoding the antibody binding fragment comprises the CDR nucleic acid sequence of the GPC 2-specific monoclonal antibody disclosed in Li et al, Proc Natl Acad Sci USA 114(32): E6623-E6631,2017.

In some examples, wherein the tumor antigen is mesothelin, the nucleic acid encoding the antibody binding fragment comprises the VH domain CDR1, CDR2, and CDR3 nucleic acid sequences of SEQ ID NO 37(YP218 VH domain nucleotide sequences) and the VL domain CDR1, CDR2, and CDR3 nucleic acid sequences of SEQ ID NO 39(YP218 VL domain nucleotide sequences). The CDR sequences can be determined using any well-known numbering scheme, such as IMGT, Kabat, or Chothia. In specific examples, the VH domain CDR1, CDR2 and CDR3 nucleic acid sequences each include nucleotides 91-108, 101-204 and 298-36 of SEQ ID NO: 37; and/or the VL domain CDR1, CDR2 and CDR3 nucleic acid sequences each comprise nucleotides 70-102, 148-168 and 265-303 of SEQ ID NO: 39. In other specific examples, the VH domain CDR1, CDR2 and CDR3 nucleic acid sequences each include nucleotides 79-102, 154-177 and 292-336 of SEQ ID NO: 37; and/or the VL domain CDR1, CDR2 and CDR3 nucleic acid sequences each comprise nucleotides 79-96, 148-156 and 265-303 of SEQ ID NO: 39.

In other examples, wherein the tumor antigen is mesothelin, the nucleic acid encoding the antibody binding fragment comprises the CDR1, CDR2 and CDR3 nucleic acid sequences of SEQ ID NO:41(SD1 single domain antibody nucleotide sequence). The CDR sequences can be determined using any well-known numbering scheme, such as IMGT, Kabat, or Chothia. In a specific example, the CDR1, CDR2 and CDR3 nucleic acid sequences each include nucleotides 91-105, 151-198 and 295-306 of SEQ ID NO. 41. In other specific examples, the CDR1, CDR2 and CDR3 nucleic acid sequences each include nucleotides 78-105, 151-174 and 289-309 of SEQ ID NO. 41. Further provided herein are vectors comprising the CAR-encoding nucleic acid molecules disclosed herein. In some embodiments, the vector is a viral vector, such as, but not limited to, a lentiviral vector.

In other examples, wherein the tumor antigen is mesothelin, the nucleic acid encoding the antibody binding fragment comprises the CDR nucleic acid sequences of mesothelin-specific monoclonal antibodies disclosed in WO2014/031476, WO2014/052064, U.S. patent No. 8,460,660, U.S. patent No. 6,809,184, or U.S. patent No. 7,081,518, each of which is incorporated herein by reference in its entirety.

Also provided are isolated host cells comprising the CAR-encoding nucleic acid molecules disclosed herein. In some embodiments, the isolated host cell is a T lymphocyte.

The present disclosure also provides isolated host cells that co-express a Chimeric Antigen Receptor (CAR) and a truncated human epidermal growth factor receptor (huEGFRt). In some embodiments, the CAR comprises an antigen-specific antibody or antigen-binding fragment thereof, an extracellular hinge region, a transmembrane domain, an intracellular costimulatory domain, and an intracellular signaling domain; and/or the huEGFRT comprises domain III, domain IV and a transmembrane domain from human EGFR, but lacks an Epidermal Growth Factor (EGF) -binding domain and a cytoplasmic domain.

In some embodiments, the extracellular hinge region comprises a CD8a hinge region, a CD28 hinge region, or a sequence from another immunoglobulin molecule such as IgG1, IgG4, or IgD (e.g., from a CH2 and/or CH3 domain of an immunoglobulin molecule).

In some embodiments, the transmembrane domain comprises a CD8a, CD28, CD3, CD45, CD4, CD5, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, or CD154 transmembrane domain. The transmembrane domain may also be a transmembrane region of the α, β or zeta chain of a T cell receptor.

In some embodiments, the intracellular co-stimulatory domain comprises a 4-1BB (CD137, TNFRSF9), CD28, ICOS, OX40(CD134), CD27, CD30, CD40, PD-1, lymphocyte function-associated antigen 1(LFA-1), CD2, CD7, LIGHT, NKG2C, B7-H3, or DAP10 co-stimulatory domain.

In some embodiments, the intracellular signaling domain is a domain with ITAMs, such as a CD3 ζ or FcRI γ signaling domain.

In a specific embodiment, the extracellular hinge region comprises a CD8a hinge region, the transmembrane domain comprises a CD8a transmembrane domain, the intracellular co-stimulatory domain comprises a 4-1BB co-stimulatory domain, and the intracellular signaling domain comprises a CD3zeta signaling domain.

In some examples, the amino acid sequence of the CD8a hinge region is at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to SEQ ID No. 4. In a specific example, the amino acid sequence of the CD8a hinge region includes SEQ ID NO 4.

In some examples, the amino acid sequence of the transmembrane domain of CD8a is at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to SEQ ID No. 6. In a specific example, the amino acid sequence of the transmembrane domain of CD8a includes SEQ ID NO 6.

In some examples, the amino acid sequence of the 4-1BB co-stimulatory domain is at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to SEQ ID No. 8. In a specific example, the amino acid sequence of the 4-1BB co-stimulatory domain includes SEQ ID NO 8.

In some examples, the amino acid sequence of the CD3zeta signaling domain is at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to SEQ ID No. 10. In a specific example, the amino acid sequence of the CD3zeta signaling domain includes SEQ ID NO 10.

In some examples, the amino acid sequence of huEGFRt is at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to SEQ ID No. 14. In a specific example, the amino acid sequence of huEGFRT includes SEQ ID NO 14.

In some embodiments, the antigen-binding fragment is a scFv or a single domain antibody.

In some embodiments, the antibody or antigen binding fragment specifically binds to a tumor antigen. In some examples, the tumor antigen is GPC3, GPC2, or mesothelin.

In some examples, wherein the tumor antigen is GPC3, the amino acid sequence of the antigen-binding fragment comprises the VH domain CDR1, CDR2, and CDR3 sequences of SEQ ID No. 26(hYP7 VH domain) and the VL domain CDR1, CDR2, and CDR3 sequences of SEQ ID No. 28(hYP7VL domain). The CDR sequences can be determined using any well-known numbering scheme, such as IMGT, Kabat, or Chothia. In specific examples, the VH domain CDR1, CDR2 and CDR3 amino acid sequences each include residues 31-35, 50-68 and 101-106 of SEQ ID NO. 26 and/or the VL domain CDR1, CDR2 and CDR3 amino acid sequences each include residues 24-40, 56-62 and 95-103 of SEQ ID NO. 28. In other specific examples, the VH domain CDR1, CDR2, and CDR3 amino acid sequences each include residues 26-33, 51-60, and 99-106 of SEQ ID NO. 26 and/or the VL domain CDR1, CDR2, and CDR3 amino acid sequences each include residues 27-38, 56-58, and 95-103 of SEQ ID NO. 28. In one non-limiting example, the amino acid sequence of the antibody binding fragment includes residues 25-269 of SEQ ID NO 16.

In other examples, wherein the tumor antigen is GPC3, the amino acid sequence of the antigen-binding fragment includes the CDR1, CDR2, and CDR3 sequences of SEQ ID NO:30(HN3 single domain antibody sequence). The CDR sequences can be determined using any well-known numbering scheme, such as IMGT, Kabat, or Chothia. In a specific example, the CDR1, CDR2, and CDR3 amino acid sequences each include residues 31-35,50-65, and 96-105 of SEQ ID NO. 30. In other specific examples, the CDR1, CDR2, and CDR3 amino acid sequences each include residues 26-33, 51-57, and 96-105 of SEQ ID NO. 30. In one non-limiting example, the amino acid sequence of the antibody binding fragment includes residues 25-140 of SEQ ID NO 18.

In other examples, wherein the tumor antigen is GPC3, the amino acid sequence of the antibody binding fragment includes the CDR sequences of a GPC 3-specific monoclonal antibody disclosed in WO2013/181543 or WO 2012/145469, which are incorporated by reference herein in their entirety.

In some examples, wherein the tumor antigen is GPC2, the amino acid sequence of the antigen-binding fragment includes the CDR1, CDR2, and CDR3 sequences of SEQ ID NO:32(LH7 single domain antibody sequence). The CDR sequences can be determined using any well-known numbering scheme, such as IMGT, Kabat, or Chothia. In a specific example, the CDR1, CDR2, and CDR3 amino acid sequences each include residues 26-33, 51-57, and 96-109 of SEQ ID NO. 32. In other specific examples, the CDR1, CDR2, and CDR3 amino acid sequences each include residues 31-35,50-65, and 96-109 of SEQ ID NO. 32. In one non-limiting example, the amino acid sequence of the antibody binding fragment includes residues 25-144 of SEQ ID NO. 20.

In other examples, wherein the tumor antigen is GPC2, the amino acid sequence of the antigen-binding fragment includes the CDR1, CDR2, and CDR3 sequences of SEQ ID NO:34(LH4 single domain antibody sequence). The CDR sequences can be determined using any well-known numbering scheme, such as IMGT, Kabat, or Chothia. In a specific example, the CDR1, CDR2, and CDR3 amino acid sequences each include residues 31-35,50-65, and 96-109 of SEQ ID NO. 34. In other specific examples, the CDR1, CDR2, and CDR3 amino acid sequences each include residues 26-33, 51-57, and 96-109 of SEQ ID NO. 34.

In other examples, wherein the tumor antigen is GPC2, the amino acid sequence of the antigen-binding fragment includes the CDR1, CDR2, and CDR3 sequences of SEQ ID NO:36(LH6 single domain antibody sequence). The CDR sequences can be determined using any well-known numbering scheme, such as IMGT, Kabat, or Chothia. In a specific example, the CDR1, CDR2, and CDR3 amino acid sequences each include residues 31-35, 50-66, and 97-110 of SEQ ID NO. 36. In other specific examples, the CDR1, CDR2, and CDR3 amino acid sequences each include residues 26-33, 51-58, and 97-110 of SEQ ID NO. 36.

In other examples, wherein the tumor antigen is GPC2, the amino acid sequence of the antibody binding fragment includes the CDR sequences of the GPC 2-specific monoclonal antibody disclosed in Li et al, Proc Natl Acad Sci USA 114(32): E6623-E6631,2017.

In some examples, wherein the tumor antigen is mesothelin, the amino acid sequence of the antigen-binding fragment comprises the VH domain CDR1, CDR2, and CDR3 sequences of SEQ ID NO:38(YP218 VH domain) and the VL domain CDR1, CDR2, and CDR3 sequences of SEQ ID NO:40(YP218 VL domain). The CDR sequences can be determined using any well-known numbering scheme, such as IMGT, Kabat, or Chothia. In a specific example, the VH domain CDR1, CDR2 and CDR3 amino acid sequences each include residues 31-36, 51-68 and 100-112 of SEQ ID NO. 38 and/or the VL domain CDR1, CDR2 and CDR3 amino acid sequences each include residues 24-34, 50-56 and 89-101 of SEQ ID NO. 40. In other specific examples, the VH domain CDR1, CDR2, and CDR3 amino acid sequences each include residues 27-34, 52-59, and 98-112 of SEQ ID NO. 38 and/or the VL domain CDR1, CDR2, and CDR3 amino acid sequences each include residues 27-32, 50-52, and 89-101 of SEQ ID NO. 40.

In other examples, wherein the tumor antigen is mesothelin, the amino acid sequence of the antigen-binding fragment includes the CDR1, CDR2 and CDR3 sequences of SEQ ID NO:42(SD1 single domain antibody sequence). The CDR sequences can be determined using any well-known numbering scheme, such as IMGT, Kabat, or Chothia. In a specific example, the CDR1, CDR2, and CDR3 amino acid sequences each include residues 31-35, 51-66, and 99-102 of SEQ ID NO. 42. In other specific examples, the CDR1, CDR2, and CDR3 amino acid sequences each include residues 26-35, 51-58, and 97-103 of SEQ ID NO. 42.

In other examples, wherein the tumor antigen is mesothelin, the amino acid sequence of the antibody binding fragment includes the CDR sequences of mesothelin-specific monoclonal antibodies disclosed in WO2014/031476, WO2014/052064, U.S. patent No. 8,460,660, U.S. patent No. 6,809,184, or U.S. patent No. 7,081,518, each of which is incorporated herein by reference in its entirety.

In some embodiments, the isolated host cell is a T lymphocyte. In some examples, the T lymphocyte is an autologous T lymphocyte. In other examples, the T lymphocytes are allogeneic T lymphocytes.

Also provided herein are compositions comprising an isolated (CAR-expressing-) host cell disclosed herein and a pharmaceutically acceptable carrier.

Further provided herein are methods of treating a GPC 3-positive cancer in a subject. In some embodiments, the method comprises administering to the subject a therapeutically effective amount of an isolated host cell comprising a nucleic acid molecule encoding a GPC 3-targeted CAR disclosed herein, or administering a therapeutically effective amount of an isolated host cell co-expressing a GPC 3-targeted CAR and a huEGFRt, as disclosed herein. In some examples, the GPC 3-positive cancer is hepatocellular carcinoma, melanoma, clear cell carcinoma of the ovary, yolk sac tumor, neuroblastoma, hepatoblastoma, or Wilms tumor.

Also provided are methods of treating a GPC 2-positive cancer in a subject. In some embodiments, the method comprises administering to the subject a therapeutically effective amount of an isolated host cell comprising a nucleic acid molecule encoding a GPC 2-targeted CAR disclosed herein, or using a therapeutically effective amount of an isolated host cell co-expressing a GPC 2-targeted CAR and a huEGFRt, as disclosed herein. In some examples, the GPC 2-positive cancer is neuroblastoma, acute lymphocytic leukemia, embryonal rhabdomyosarcoma, alveolar rhabdomyosarcoma, ewing's sarcoma, desmoplastic small round cell tumor, or osteosarcoma.

Further provided are methods of treating a mesothelin-positive cancer in a subject. In some embodiments, the method comprises administering to the subject a therapeutically effective amount of an isolated host cell comprising a nucleic acid molecule encoding a mesothelin-targeted CAR disclosed herein, or administering a therapeutically effective amount of an isolated host cell co-expressing a mesothelin-targeted CAR and a huEGFRt, as disclosed herein. In some examples, the mesothelin-positive cancer is mesothelioma, prostate cancer, lung cancer, gastric cancer, squamous cell carcinoma, pancreatic cancer, cholangiocarcinoma, triple negative breast cancer, or ovarian cancer.

In some embodiments of the methods of treatment, the isolated host cell is a T lymphocyte. In some examples, the T lymphocyte is an autologous T lymphocyte. In other examples, the T lymphocytes are allogeneic T lymphocytes.

In one embodiment herein, there is provided a nucleic acid molecule encoding a CAR comprising in a5 'to 3' direction a nucleic acid encoding a first GMCSFRss; a nucleic acid encoding an antigen-specific antibody or antigen-binding fragment thereof; a nucleic acid encoding a CD8a hinge region; a nucleic acid encoding a transmembrane domain of CD8 a; a nucleic acid encoding a 4-1BB co-stimulatory domain; a nucleic acid encoding a CD3zeta signaling domain; a nucleic acid encoding a self-cleaving 2A peptide; a nucleic acid encoding a second GMCSFRss; and a nucleic acid encoding huEGFRT. In some examples, the nucleic acid encoding the antibody binding fragment comprises the sequence of nucleotides 73-807 of SEQ ID NO. 15, nucleotides 73-420 of SEQ ID NO. 17, or nucleotides 73-432 of SEQ ID NO. 19. In particular examples, the nucleic acid molecule is at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to SEQ ID No. 15, SEQ ID No. 17, or SEQ ID No. 19. In specific non-limiting examples, the nucleic acid molecule comprises the nucleotide sequence of SEQ ID NO 15, SEQ ID NO 17 or SEQ ID NO 19.

In one embodiment herein, there is provided an isolated host cell co-expressing a CAR and a huEGFRt, wherein the CAR comprises an antigen-specific antibody or antigen-binding fragment thereof, a CD8a hinge region, a CD8a transmembrane domain, a 4-1BB co-stimulatory domain, and a CD3zeta signaling domain; and said huEGFRT comprises domain III, domain IV and a transmembrane domain from human EGFR, but lacks an EGF-binding domain and a cytoplasmic domain. In some examples, the amino acid sequence of the antigen-binding fragment includes residues 25-269 of SEQ ID NO 16, residues 25-140 of SEQ ID NO 18, or residues 25-144 of SEQ ID NO 20. In specific examples, the amino acid sequence of the CAR is at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to residues 25-491 of SEQ ID No. 16, residues 25-362 of SEQ ID No. 18, or residues 25-366 of SEQ ID No. 20, and the amino acid sequence of the huEGFRt is at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to SEQ ID No. 14. In other specific examples, the amino acid sequence of the CAR includes NO more than 10, NO more than 9, NO more than 8, NO more than 7, NO more than 6, NO more than 5, NO more than 4, NO more than 3, NO more than 2, or NO more than 1 amino acid substitution relative to residues 25-491 of SEQ ID No. 16, residues 25-362 of SEQ ID No. 18, or residues 25-366 of SEQ ID No. 20, and the amino acid sequence of the huEGFRt includes NO more than 10, NO more than 9, NO more than 8, NO more than 7, NO more than 6, NO more than 5, NO more than 4, NO more than 3, NO more than 2, or NO more than 1 amino acid substitution relative to SEQ ID No. 14. In a specific non-limiting example, the amino acid sequence of the CAR comprises residues 25-491 of SEQ ID NO:16, residues 25-362 of SEQ ID NO:18, or residues 25-366 of SEQ ID NO:20, and the amino acid sequence of the huEGFRT comprises SEQ ID NO: 14.

Antibodies specific for tumor antigens

The CARs disclosed herein can be targeted to tumor cells expressing or overexpressing a particular antigen by selecting an appropriate tumor antigen-specific monoclonal antibody or antigen-binding fragment thereof. In some embodiments, the antigen binding portion of the CAR is an antigen binding fragment of a monoclonal antibody. In particular examples, the antigen-binding fragment is a scFv or a single domain (VH domain) antibody. Although the CARs disclosed herein can be used with any antigen-specific antibody (or antigen-binding fragment thereof), exemplary antibodies include GPC 3-specific, GPC 2-specific, and mesothelin-specific monoclonal antibodies.

GPC 3-specific antibodies

The CAR constructs disclosed herein can be engineered to include any GPC 3-specific monoclonal antibody or antigen-binding fragment thereof. Several GPC 3-specific monoclonal antibodies are known in the art, including, but not limited to, YP6, YP7, YP8, YP9, and YP9.1 disclosed in PCT publication No. WO2013/181543, and HN3 disclosed in WO 2012/145469, which are incorporated herein by reference in their entirety. In some embodiments herein, the CAR comprises an antigen binding fragment comprising the CDR sequences of GPC 3-specific monoclonal antibody YP7 (disclosed in WO 2013/181543) or a humanized form thereof. The nucleotide and amino acid sequences of YP7 and humanized YP7(hYP7) are provided below. Tables 1A-1D show CDR1, CDR2 and CDR3 of YP7 and hYP 7. In other embodiments, the CAR comprises a single domain monoclonal antibody comprising the CDR sequences of the GPC 3-specific antibody HN3 (disclosed in WO 2012/145469). The nucleotide and amino acid sequence of HN3 is provided below. Tables 2A-2B show the positions of CDR1, CDR2 and CDR3 in HN 3.

YP7 VH nucleotide sequence (SEQ ID NO:21)

GAGGTGCAGCTTGTTGAGACTGGTGGAGGAATGGTGCAGCCTGAAGGGTCATTGAAACTCTCATGTGCAGCCTCTGGATTCACCTTCAATAAGAATGCCATGAATTGGGTCCGCCAGGCTCCAGGAAAGGGTTTGGAATGGGTTGCTCGCATAAGAAATAAAACTAATAATTATGCAACATATTATGCCGATTCAGTGAAAGCCAGGTTTACCATCTCCAGAGATGATTCACAAAGCATGCTCTATCTGCAAATGAACAACTTGAAAATTGAGGACACAGCCATGTACTATTGTGTGGCTGGTAACTCGTTTGCTTACTGGGGCCAAGGGACTCTGGTCACTGTCTCTGCA

YP7 VH amino acid sequence (SEQ ID NO:22)

EVQLVETGGGMVQPEGSLKLSCAASGFTFNKNAMNWVRQAPGKGLEWVARIRNKTNNYATYYADSVKARFTISRDDSQSMLYLQMNNLKIEDTAMYYCVAGNSFAYWGQGTLVTVSA

YP7VL nucleotide sequence (SEQ ID NO:23)

GACATTGTGATGTCACAGTCTCCATCCTCCCTAGTTGTGTCAATTGGAGAGAAGGTTACTATGACCTGCAAGTCCAGTCAGAGCCTTTTATATAGCAGCAATCAAAAGAACTACTTGGCCTGGTACCAACAGAAACCAGGGCAGTCTCCTAAACTGCTGATTTACTGGGCATCCAGTAGGGAATCTGGGGTCCCTGATCGCTTCACAGGCAGTGGATCTGGGACAGATTTCACTCTCACCATCAGCAGTGTGAAGGCTGAAGACCTGGCAGTTTATTACTGTCAGCAATATTATAACTATCCGCTCACGTTCGGTGCTGGGACCAAGTTGGAGCTGAAA

YP7VL amino acid sequence (SEQ ID NO:24)

DIVMSQSPSSLVVSIGEKVTMTCKSSQSLLYSSNQKNYLAWYQQKPGQSPKLLIYWASSRESGVPDRFTGSGSGTDFTLTISSVKAEDLAVYYCQQYYNYPLTFGAGTKLELK

hYP7 VH nucleotide sequence (SEQ ID NO:25)

GAGGTGCAGCTTGTTGAGTCTGGTGGAGGATTGGTGCAGCCTGGAGGGTCATTGAGACTCTCATGTGCAGCCTCTGGATTCACCTTCAATAAGAATGCCATGAATTGGGTCCGCCAGGCTCCAGGAAAGGGTTTGGAATGGGTTGGCCGCATAAGAAATAAAACTAATAATTATGCAACATATTATGCCGATTCAGTGAAAGCCAGGTTTACCATCTCCAGAGATGATTCAAAGAACTCACTCTATCTGCAAATGAACAGCTTGAAAACCGAGGACACAGCCGTGTACTATTGTGTGGCTGGTAACTCGTTTGCTTACTGGGGCCAAGGGACTCTGGTCACTGTCTCTGCA

hYP7 VH amino acid sequence (SEQ ID NO:26)

EVQLVESGGGLVQPGGSLRLSCAASGFTFNKNAMNWVRQAPGKGLEWVGRIRNKTNNYATYYADSVKARFTISRDDSKNSLYLQMNSLKTEDTAVYYCVAGNSFAYWGQGTLVTVSA

hYP7VL nucleotide sequence (SEQ ID NO:27)

GACATTGTGATGACCCAGTCTCCAGACTCCCTAGCTGTGTCACTGGGAGAGAGGGCCACTATCAACTGCAAGTCCAGTCAGAGCCTTTTATATAGCAGCAATCAAAAGAACTACTTGGCCTGGTACCAACAGAAACCAGGGCAGCCTCCTAAACTGCTGATTTACTGGGCATCCAGTAGGGAATCTGGGGTCCCTGATCGCTTCAGTGGCAGTGGATCTGGGACAGATTTCACTCTCACCATCAGCAGTCTGCAGGCTGAAGACGTGGCAGTTTATTACTGTCAGCAATATTATAACTATCCGCTCACGTTCGGTCAGGGGACCAAGTTGGAGATCAAA

hYP7VL amino acid sequence (SEQ ID NO:28)

DIVMTQSPDSLAVSLGERATINCKSSQSLLYSSNQKNYLAWYQQKPGQPPKLLIYWASSRESGVPDRFSGSGSGTDFTLTISSLQAEDVAVYYCQQYYNYPLTFGQGTKLEIK

TABLE 1A. position of CDRs in YP7/hYP7 VH sequence (according to Kabat)

TABLE 1B position of CDRs in YP7/hYP7 VH sequences (according to IMGT)

Figure BDA0002486745430000222

TABLE 1C. position of CDR in YP7/hYP 7VL sequence (according to Kabat)

Figure BDA0002486745430000231

TABLE 1D. position of CDRs in YP7/hYP 7VL sequences (according to IMGT)

Figure BDA0002486745430000232

HN3 DNA sequence (SEQ ID NO:29)

CAGGTGCAGCTGGTGCAGTCTGGGGGAGGCTTGGTACAGCCTGGAGGGTCCCTGAGACTCTCCTGTGCAGCCTCTTATTTCGATTTCGATTCTTATGAAATGAGCTGGGTCCGCCAGGCTCCAGGGAAGGGCCTAGAGTGGATTGGGAGTATCTATCATAGTGGGAGCACCTACTACAACCCGTCCCTCAAGAGTCGAGTCACCATCTCCAGAGACAATTCCAAGAACACGCTGTATCTGCAAATGAACACCCTGAGAGCCGAGGACACAGCCACGTATTACTGTGCGAGAGTAAATATGGACCGATTTGACTACTGGGGCCAGGGAACCCTGGTCACCGTCTCCTCAAGT

HN3 protein sequence (SEQ ID NO:30)

QVQLVQSGGGLVQPGGSLRLSCAASYFDFDSYEMSWVRQAPGKGLEWIGSIYHSGSTYYNPSLKSRVTISRDNSKNTLYLQMNTLRAEDTATYYCARVNMDRFDYWGQGTLVTVSSS

Table 2a. position of CDR in HN3 sequence (according to Kabat)

CDR DNA sequence (SEQ ID NO:29) Protein sequence (SEQ ID NO:30)
CDR1 Nucleotides 91 to 105 Amino acids 31-35
CDR2 Nucleotide 148- Amino acid 50-65
CDR3 Nucleotide 286- Amino acids 96-105

TABLE 2B. position of CDR in HN3 sequence (according to IMGT)

CDR DNA sequence (SEQ ID NO:29) Protein sequence (SEQ ID NO:30)
CDR1 Nucleotides 76 to 99 Amino acids 26-33
CDR2 Nucleotide 151- Amino acids 51-57
CDR3 Nucleotide 286- Amino acids 96-105

GPC 2-specific antibodies

The CAR constructs disclosed herein can also be engineered to include any GPC 2-specific monoclonal antibody or antigen-binding fragment thereof. In some embodiments herein, the CAR comprises an antigen-binding fragment comprising the CDR sequences of GPC 2-specific single domain monoclonal antibodies LH7, LH4, LH6, LH1, LH2, or LH3 (disclosed in Liet al, Proc Natl Acad Sci USA 114(32): E6623-E6631,2017). The nucleotide and amino acid sequences of LH7, LH4 and LH6 are provided below. Tables 3A-5B show the positions of CDR1, CDR2 and CDR3 of H7, LH4 and LH 6.

LH7 DNA(SEQ ID NO:31)

CAGGTGCAGCTGGTGCAGTCTGGGGGAGGCTTGGTACAGCCTGGAGGGTCCCTGAGACTCTCCTGTGCAGCCTCTGATTTCTATTTCTATGATTATGAAATGAGCTGGGTCCGCCAGGCTCCAGGGAAGGGTCTGGAGTGGATTGGGACTGTCTCCTATAGTGGGAGCACCTACTACAACCCGTCCCTCAAGAGTCGAGTCACCATCTCCAGAGACAATTCCAAGAACACGCTGTATCTGCAAATGAACACCCTAAGAGCCGAGGACACAGCCATGTATTACTGTGCGAGAGGTTACAGCTATGATGACTCCCGATATTTTGACTACTGGGGCCAGGGAACCCTGGTCACCGTCTCCTCA

LH7 protein (SEQ ID NO:32)

QVQLVQSGGGLVQPGGSLRLSCAASDFYFYDYEMSWVRQAPGKGLEWIGTVSYSGSTYYNPSLKSRVTISRDNSKNTLYLQMNTLRAEDTAMYYCARGYSYDDSRYFDYWGQGTLVTVSS

TABLE 3A. position of CDR in LH7 sequence (according to Kabat)

CDR DNA sequence (SEQ ID NO:31) Protein sequence (SEQ ID NO:32)
CDR1 91-105 31-35
CDR2 148-195 50-65
CDR3 286-327 96-109

TABLE 3B position of CDR in LH7 sequence (according to IMGT)

CDR DNA sequence (SEQ ID NO:31) Protein sequence (SEQ ID NO:32)
CDR1 76-99 26-33
CDR2 151-171 51-57
CDR3 286-327 96-109

LH4 DNA(SEQ ID NO:33)

CAGGTGCAGCTGGTGCAGTCTGGGGGAGGCTTGGTACAGCCTGGAGGGTCCCTGAGACTCTCCTGTGCAGCCTCTTCTTTCTATTTCGATGATTATGAAATGAGCTGGGTCCGCCAGGCTCCAGGGAAGGCCCTGGAGTGGATTGGGCGTATCTATACCAGTGGGAGCACCAACTACAACCCCTCCCTCAAGAGTCGAGTCACCATCTCCAGAGACAATTCCAAGAACACGCTGTATCTGCAAATGAACACCCTGAGAGCCGAGGACACAGCCACGTATTACTGTGCGAGGGGATATTGTAGTGGTGGTAGCTGCTACTTTGACTACTGGGGCCAGGGAACCCTGGTCACCGTCTCCTCA

LH4 protein (SEQ ID NO:34)

QVQLVQSGGGLVQPGGSLRLSCAASSFYFDDYEMSWVRQAPGKALEWIGRIYTSGSTNYNPSLKSRVTISRDNSKNTLYLQMNTLRAEDTATYYCARGYCSGGSCYFDYWGQGTLVTVSS

TABLE 4A. position of CDR in LH4 sequence (according to Kabat)

CDR DNA sequence (SEQ ID NO:33) Protein sequence (SEQ ID NO:34)
CDR1 91-105 31-35
CDR2 148-195 50-65
CDR3 286-327 96-109

TABLE 4B position of CDR in LH4 sequence (according to IMGT)

CDR DNA sequence (SEQ ID NO:33) Protein sequence (SEQ ID NO:34)
CDR1 76-99 26-33
CDR2 151-171 51-57
CDR3 286-327 96-109

LH6 DNA(SEQ ID NO:35)

CAGGTGCAGCTGGTGCAGTCTGGGGGAGGCTTGGTACAGCCTGGAGGGTCCCTGAGACTCTCCTGTGCAGCCTCTGATTTCTATTTCGATGATTATGAAATGAGCTGGGTCCGCCAGGCTCCAGGGAAGGGGCTGGAGTGGGTCTCAACTATTAGTGGTAGTGGTGGTGGCACATACTACGCAGACTCAGTGAAGGGCCGATTCACCATCTCCAGAGACAATTCCAAGAACACGCTGTATCTGCAAATGAACACCCTGAGAGCCGAGGACACAGCCACATATTACTGTGCGAGAGGTTACAGTTATGACGACTCCCGATATTTTGACTACTGGGGCCAGGGAACCCTGGTCACCGTCTCCTCA

LH6 protein (SEQ ID NO:36)

QVQLVQSGGGLVQPGGSLRLSCAASDFYFDDYEMSWVRQAPGKGLEWVSTISGSGGGTYYADSVKGRFTISRDNSKNTLYLQMNTLRAEDTATYYCARGYSYDDSRYFDYWGQGTLVTVSS

TABLE 5A. position of CDR in LH6 sequence (according to Kabat)

CDR DNA sequence (SEQ ID NO:35) Protein sequence (SEQ ID NO:36)
CDR1 91-105 31-35
CDR2 148-198 50-66
CDR3 289-330 97-110

TABLE 5B position of CDR in LH6 sequence (according to IMGT)

CDR DNA sequence (SEQ ID NO:35) Protein sequence (SEQ ID NO:36)
CDR1 76-99 26-33
CDR2 151-174 51-58
CDR3 289-330 97-110

C. Mesothelin-specific antibodies

The CAR constructs disclosed herein can also be engineered to include any mesothelin-specific monoclonal antibody or antigen-binding fragment thereof. Several mesothelin-specific monoclonal antibodies are known in the art, including, but not limited to YP218, YP223, YP3, YP158, and YP187 disclosed in PCT publication No. WO2014/031476, SD1 disclosed in PCT publication No. WO2014/052064, HN1 disclosed in U.S. patent No. 8,460,660, SS disclosed in U.S. patent No. 6,809,184, and SS1 disclosed in U.S. patent No. 7,081,518, each of which is incorporated herein by reference in its entirety. The nucleotide and amino acid sequences of YP218 and SD1 are provided below. Tables 6A-7B show the positions of the CDRs of YP218 and SD 1.

YP218 VH nucleotide sequence (SEQ ID NO:37)

CAGCAGCAGCTGGAGGAGTCCGGGGGAGGCCTGGTCAAGCCTGAGGGATCCCTGACACTCACCTGCAAAGCCTCTGGATTCGACCTCGGTTTCTACTTTTACGCCTGTTGGGTCCGCCAGGCTCCAGGGAAGGGCCTGGAGTGGATCGCATGCATTTATACTGCTGGTAGTGGTAGCACGTACTACGCGAGCTGGGCGAAAGGCCGATTCACCATCTCCAAAGCCTCGTCGACCACGGTGACTCTGCAAATGACCAGTCTGGCAGCCGCGGACACGGCCACCTATTTCTGTGCGAGATCTACTGCTAATACTAGAAGTACTTATTATCTTAACTTGTGGGGCCCAGGCACCCTGGTCACCGTCTCCTCA

YP218 VH amino acid sequence (SEQ ID NO:38)

QQQLEESGGGLVKPEGSLTLTCKASGFDLGFYFYACWVRQAPGKGLEWIACIYTAGSGSTYYASWAKGRFTISKASSTTVTLQMTSLAAADTATYFCARSTANTRSTYYLNLWGPGTLVTVSS

YP218 VL nucleotide sequence (SEQ ID NO:39)

GACGTCGTGATGACCCAGACTCCAGCCTCCGTGTCTGAACCTGTGGGAGGCACAGTCACCATCAAGTGCCAGGCCAGTCAGAGGATTAGTAGTTACTTATCCTGGTATCAGCAGAAACCAGGGCAGCGTCCCAAGCTCCTGATCTTTGGTGCATCCACTCTGGCATCTGGGGTCCCCTCGCGGTTCAAAGGCAGTGGATCTGGGACAGAATACACTCTCACCATCAGCGACCTGGAGTGTGCCGATGCTGCCACTTACTACTGTCAGAGTTATGCTTATTTTGATAGTAATAATTGGCATGCTTTCGGCGGAGGGACCGAGGTGGTGGTC

YP218 VL amino acid sequence (SEQ ID NO:40)

DVVMTQTPASVSEPVGGTVTIKCQASQRISSYLSWYQQKPGQRPKLLIFGASTLASGVPSRFKGSGSGTEYTLTISDLECADAATYYCQSYAYFDSNNWHAFGGGTEVVV

TABLE 6A. position of CDRs in YP218 VH sequence (according to Kabat)

CDR DNA sequence (SEQ ID NO:37) Protein sequence (SEQ ID NO:38)
CDR1 Nucleotides 91 to 108 Amino acids 31-36
CDR2 Nucleotide 101-204 Amino acids 51-68
CDR3 Nucleotide 298- Amino acid 100-

TABLE 6B position of CDRs in YP218 VH sequence (according to IMGT)

CDR DNA sequence (SEQ ID NO:37) Protein sequence (SEQ ID NO:38)
CDR1 Nucleotides 79 to 102 Amino acids 27 to 34
CDR2 Nucleotide 154-177 Amino acids 52-59
CDR3 Nucleotide 292- Amino acids 98 to 112

TABLE 6C. position of CDR in YP218 VL sequence (according to Kabat)

CDR DNA sequence (SEQ ID NO:39) Protein sequence (SEQ ID NO:40)
CDR1 Nucleotides 70 to 102 Amino acids 24-34
CDR2 Nucleotide 148- Amino acids 50-56
CDR3 Nucleotide 265- Amino acids 89-101

TABLE 6D. position of CDR in YP218 VL sequence (according to IMGT)

CDR DNA sequence (SEQ ID NO:39) Protein sequence (SEQ ID NO:40)
CDR1 Nucleotides 79 to 96 Amino acids 27-32
CDR2 Nucleotide 148- Amino acids 50-52
CDR3 Nucleotide 265- Amino acids 89-101

SD1 nucleotide sequence (SEQ ID NO:41):

CAGGTGCAGCTGGTGCAGTCTGGGGGAGGCTTGGTACAGCCTGGAGGGTCCCTGAGACTCTCCTGTGCAGCCTCTGATTTCGATTTCGCTGCTTATGAAATGAGCTGGGTCCGCCAGGCTCCAGGACAAGGCCTTGAGTGGGTGGCAATTATATCACATGATGGAATCGATAAATACTACACAGACTCCGTGAAGGGCCGATTCACCATCTCCAGAGACAATTCCAAGAACACGCTGTATCTGCAAATGAACACCCTGAGAGCCGAGGACACAGCCACGTATTACTGTTTAAGGCTTGGTGCTGTAGGCCAGGGAACCCTGGTCACCGTCTCCTCAAGT

SD1 amino acid sequence (SEQ ID NO:42):

QVQLVQSGGGLVQPGGSLRLSCAASDFDFAAYEMSWVRQAPGQGLEWVAIISHDGIDKYYTDSVKGRFTISRDNSKNTLYLQMNTLRAEDTATYYCLRLGAVGQGTLVTVSSS

table 7a. position of CDR in SD1 sequence (according to Kabat)

CDR DNA sequence (SEQ ID NO:41) Protein sequence (SEQ ID NO:42)
CDR1 91-105 31-35
CDR2 151-198 51-66
CDR3 295-306 99-102

TABLE 7B position of CDR in SD1 sequence (according to IMGT)

CDR DNA sequence (SEQ ID NO:41) Protein sequence (SEQ ID NO:42)
CDR1 78-105 26-35
CDR2 151-174 51-58
CDR3 289-309 97-103

Chimeric Antigen Receptor (CAR)

Disclosed herein are CARs (also referred to as chimeric T cell receptors, artificial T cell receptors, or chimeric immunoreceptors) and T cells engineered to express CARs. Typically, the CAR comprises a binding moiety, an extracellular hinge/spacer element, a transmembrane region, and an intracellular domain that performs a signaling function (Cartellieri et al, J Biomed Biotechnol 2010:956304,2010; Dai et al, J Natl Cancer Inst 108(7): djv439,2016). In many cases, the binding moiety is an antigen-binding fragment of a monoclonal antibody, such as an scFv or single domain antibody. The spacer/hinge region typically includes sequences from the IgG subclass, such as the IgG1, IgG4, IgD, and CD8 domains. The transmembrane domain may be derived from a variety of different T cell proteins, for example CD3 ζ, CD4, CD8 or CD 28.

Although the entire intracellular T cell signaling domain can be employed in a CAR, in many cases it is not necessary to use the entire chain. For use of a truncated portion of an intracellular T cell signaling domain, the truncated portion can be used in place of the entire chain so long as it transduces the relevant T cell effector function signal. Examples of intracellular T cell signaling domains for CARs include the cytoplasmic sequences of the T Cell Receptor (TCR) and costimulatory molecules that co-initiate signal transduction upon antigen receptor binding, as well as derivatives or variants of these sequences and any synthetic sequences with the same functional capacity. Several different intracellular domains have been used to generate CARs. For example, the intracellular domain may consist of a signaling chain with ITAMs, such as CD3 ζ or FcRI γ. In some cases, the intracellular domain further comprises an intracellular portion of at least one additional costimulatory domain. A costimulatory domain refers to a portion of a CAR that comprises the intracellular domain of a costimulatory molecule. Costimulatory molecules are cell surface molecules other than the antigen receptor or its ligand that are required by lymphocytes to respond effectively to antigens. Costimulatory molecules include, for example, CD28, 4-1BB (CD137, TNFRSF9), OX-40(CD134), ICOS, CD27, and/or DAP 10.

The CAR can also include a signal peptide sequence, e.g., the N-terminus of the antigen binding domain. The signal peptide sequence may be any suitable signal peptide sequence, for example, a signal sequence from granulocyte-macrophage colony stimulating factor receptor (GMCSFR), immunoglobulin light chain kappa, or IL-2. Although the signal peptide sequence may facilitate expression of the CAR on the cell surface, the presence of the signal peptide sequence in the expressed CAR is not necessary for the CAR to function. When the CAR is expressed on the cell surface, the signal peptide sequence may be cleaved from the CAR. Thus, in some embodiments, the CAR lacks a signal peptide sequence.

The CARs disclosed herein are expressed from constructs (e.g., from lentiviral vectors) that also express truncated forms of human EGFR (huEGFRt; as discussed in detail in section VI below). CAR and huEGFRt are separated by a self-cleaving peptide sequence (e.g., T2A) such that upon expression in the transduced cells, the CAR is cleaved from the huEGFRt (see figure 1).

In some embodiments disclosed herein, the CAR construct encodes the following amino acid sequence in an N-terminal to C-terminal direction:

GMCSFRss:MLLLVTSLLLCELPHPAFLLIP(SEQ ID NO:2)

NdeI:HM

antigen binding scFv or Single Domain antibody sequences

SpeI:TS

CD8alpha hinge TTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACD (SEQ ID NO:4)

CD8αTM:IYIWAPLAGTCGVLLLSLVIT(SEQ ID NO:6)

4-1BB:KRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL(SEQ ID NO:8)

CD3ζ:

RVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPR(SEQ ID NO:10)

T2A:EGRGSLLTCGDVEENPGP(SEQ ID NO:12)

GMCSFRss:MLLLVTSLLLCELPHPAFLLIP(SEQ ID NO:2)

huEGFRt:

RKVCNGIGIGEFKDSLSINATNIKHFKNCTSISGDLHILPVAFRGDSFTHTPPLDPQELDILKTVKEITGFLLIQAWPENRTDLHAFENLEIIRGRTKQHGQFSLAVVSLNITSLGLRSLKEISDGDVIISGNKNLCYANTINWKKLFGTSGQKTKIISNRGENSCKATGQVCHALCSPEGCWGPEPRDCVSCRNVSRGRECVDKCNLLEGEPREFVENSECIQCHPECLPQAMNITCTGRGPDNCIQCAHYIDGPHCVKTCPAGVMGENNTLVWKYADAGHVCHLCHPNCTYGCTGPGLEGCPTNGPKIPSIATGMVGALLLLLVVALGIGLFM(SEQ ID NO:14)

The CAR-expressing T cells disclosed herein can be used to target specific cell types, such as tumor cells, e.g., GPC 3-positive, GPC 2-positive, or mesothelin-positive tumor cells. The use of CAR-expressing T cells is more prevalent than standard CTL-based immunotherapy, as CAR-expressing CTLs are HLA-non-restricted and therefore can be used in any patient with a tumor that expresses a target antigen.

Accordingly, provided herein are CARs that include a tumor-specific antibody (or binding fragment thereof), such as a GPC 3-specific antibody, a GPC 2-specific antibody, or a mesothelin-specific antibody. Isolated nucleic acid molecules and vectors encoding the CARs, and host cells, such as T lymphocytes, expressing the CARs are also provided. T cells expressing CARs including GPC 3-specific, GPC 2-specific, or mesothelin-specific monoclonal antibodies are useful for treating cancers expressing GPC3, GPC2, and mesothelin, respectively.

Truncated human EGFR (huEGFRT)

The human epidermal growth factor receptor consists of four extracellular domains, one transmembrane domain and three intracellular domains. The EGFR domain was found in the following N-terminal to C-terminal order: domain I-domain II-domain III-domain IV-Transmembrane (TM) domain-membrane proximal domain-tyrosine kinase domain-C-terminal tail. Domain I and domain III are leucine-rich domains involved in ligand binding. Domain II and domain IV are cysteine-rich domains and are not in contact with EGFR ligands. Domain II mediates homodimerization with similar domains from other EGFR family members, and domain IV may form a disulfide bond with domain II. The EGFR TM domain has a single pass through the cell membrane and may play a role in protein dimerization. The intracellular domain includes the membrane-proximal domain that mediates EGFR signaling, the tyrosine kinase domain, and the C-terminal tail (Wee and Wang, cancer 9(52), doi:10.3390/cancer 9050052; Ferguson, Annu Rev Biophys37:353-373, 2008; Wang et al, Blood118 (5):1255-1263, 2011).

Truncated forms of human EGFR, also referred to herein as "huEGFRt" include only domain III, domain IV and TM domains. Thus, huEGFRt lacks domain I, domain II, and all three intracellular domains. huEGFRt is unable to bind EGF and lacks signaling activity. However, this molecule retains the ability to bind to specific EGFR-specific monoclonal antibodies, such as FDA-approved cetuximab (PCT publication No. WO 2011/056894, which is incorporated herein by reference).

Transduction of T cells with the constructs encoding huEGFRt and tumor antigen specific CARs (e.g., lentiviral vectors) disclosed herein allows for the use of the labeled EGFR monoclonal antibody cetuximab (ERBITUX)TM) Transduced T cells were selected. For example, cetuximab may be labeled with biotin, and transduced T cells may be selected using commercially available anti-biotin magnetic beads (e.g., from Miltenyi Biotec). Co-expression of huEGFRt also allowed for the in vivo tracking of adoptively transferred CAR-expressing T cells. Furthermore, binding of cetuximab to huEGFRT expressing T-cells induces cytotoxicity of ADCC effector cells, thereby providing a mechanism for eliminating transduced T-cells in vivo (Wang et al, Blood118 (5):1255-1263,2011), for example at the end of treatment.

In some embodiments herein, the nucleic acid molecule encoding huEGFRt is at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to SEQ ID No. 13. In some examples, the nucleic acid molecule encoding huEGFRT comprises or consists of the nucleotide sequence of SEQ ID NO 13. In some embodiments, the amino acid sequence of huEGFRt is at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to SEQ ID No. 14. In some examples, the amino acid sequence of huEGFRt comprises or consists of SEQ ID NO: 14. In other embodiments, the amino acid sequence of huEGFRt includes NO more than 10, NO more than 9, NO more than 8, NO more than 7, NO more than 6, NO more than 5, NO more than 4, NO more than 3, NO more than 2, or NO more than 1 amino acid substitution relative to SEQ ID No. 14. In some examples, the amino acid substitution is a conservative substitution.

CAR-expressing cell compositions

Compositions are provided that include CAR-expressing cells in combination with one or more pharmaceutically or physiologically acceptable carriers, diluents, or excipients. The CAR-expressing cell can be a T cell, e.g., CD3+T cells, e.g. CD4+And/or CD8+T cells, and/or NK cells. Such compositions may include buffering agents, such as neutral buffered saline solutions, phosphate buffered saline solutions, and the like; carbohydrates, such as glucose, mannose, sucrose, dextran, or mannitol; a protein; polypeptides or amino acids, such as glycine; an antioxidant; chelating agents, such as EDTA or glutathione; adjuvants (e.g., aluminum hydroxide); and a preservative. The cells may be autologous to the recipient. However, the cells may also be allogeneic (allogeneic).

With respect to cells, various aqueous carriers, such as buffered saline solutions and the like, can be used to introduce the cells. These solutions are sterile and generally free of unwanted substances. These compositions may be sterilized by conventional, well known sterilization techniques. The compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions, such as pH adjusting and buffering agents, toxicity adjusting agents and the like, for example, sodium acetate, sodium chloride, potassium chloride, calcium chloride, sodium lactate and the like. The concentration in these formulations can vary widely and will be selected primarily according to the particular mode of administration selected and the needs of the subject, according to fluid volume, viscosity, body weight, and the like.

The precise amount of the composition to be administered can be determined by a physician taking into account individual differences in age, weight, tumor size, degree of metastasis, and patient (subject) condition. In general, the pharmaceutical compositions described herein comprising CAR-expressing T cells (and/or NK cells) can be administered at the following doses: 104To 109Cells per kg body weight, e.g. 105To 106Cells/kg body weight, including all integer values within these ranges. An exemplary dose is 106Cells/kg to about 108Cells/kg, e.g. about 5X 106Cells/kg to about 7.5x 107Cells/kg, e.g. about 2.5X 107Cells/kg or about 5.0x 107Cells/kg.

The compositions may be administered in one or more of these doses, for example 2, 3, 4, 5, 6, 7, 8, 9 or 10 times. The compositions can be administered by using infusion techniques well known in immunotherapy (see, e.g., Rosenberg et al, new eng.j.of med.319:1676,1988). The composition may be administered daily, weekly, semi-monthly or monthly. In some non-limiting examples, the composition is formulated for intravenous administration and multiple administrations. The amount and frequency of administration will be determined by such factors as the condition of the subject, the type and severity of the subject's disease, although appropriate dosages may be determined by clinical trials.

In some embodiments, the CAR-encoding nucleic acid molecule is introduced into a cell, e.g., a T cell or NK cell, and the subject receives an initial cellular administration and one or more subsequent cellular administrations, wherein the subsequent one or more administrations are administered less than 15 days, e.g., 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, or 2 days after the previous administration. In one embodiment, the subject (e.g., human) is administered more than one administration of CAR-expressing cells per week, e.g., 2, 3, or 4 administrations of CAR-expressing cells of the disclosure per week. In one embodiment, the subject receives more than one administration of CAR-expressing T cells per week (e.g., 2, 3, or 4 administrations per week) (also referred to as cycles), followed by one week without administration of CAR-expressing cells, and then one or more additional administrations of CAR-expressing cells are administered to the subject (e.g., more than one administration of CAR T cells per week). In another embodiment, the subject (e.g., a human subject) receives more than one CAR-expressing cell cycle and the time between each cycle is less than 10, 9, 8, 7, 6, 5, 4, or 3 days. In one embodiment, the CAR-expressing cells are administered every other day, 3 times per week. In another embodiment, the CAR-expressing cells are administered for at least two weeks, three weeks, four weeks, five weeks, six weeks, seven weeks, eight weeks, or more. The dosage of the above treatments administered to a patient will vary with the exact nature of the condition being treated and the recipient of the treatment. The dosage for human administration may be scaled according to art-recognized practice.

In some embodiments, the CAR-expressing T cell is capable of replicating in vivo, resulting in long-term persistence, such that tumor control can be sustained. In various aspects, following administration of T cells to a subject, the T cells administered to the subject, or progeny of such cells, persist in the subject for at least 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 12 months, 13 months, 14 months, 15 months, 16 months, 17 months, 18 months, 19 months, 20 months, 21 months, 22 months, 23 months, or years. In other embodiments, the cell and its progeny are present for less than 6 months, 5 months, 4 months, 3 months, 2 months, or 1 month, e.g., 3 weeks, 2 weeks, 1 week, after administration of the CAR-expressing T cell to the subject.

Administration of the subject compositions may be carried out in any convenient manner, including by injection, ingestion, infusion, implantation or transplantation. The disclosed compositions can be administered to a patient by intravenous injection, intra-arterially, subcutaneously, intradermally, intratumorally, intranodal, intramedullary, intramuscularly, or intraperitoneally. In some embodiments, the composition is administered to the patient by intradermal or subcutaneous injection. In other embodiments, the compositions of the present invention are administered by intravenous injection. The composition may also be injected directly into the tumor or lymph nodes.

In some embodiments, the subject may undergo leukapheresis (leukapheresis), wherein leukocytes are collected, enriched, or depleted in vitro, to select and/or isolate cells of interest, e.g., T cells and/or NK cells. These cell isolates can be expanded and processed by methods known in the art such that one or more CAR constructs can be introduced, thereby generating autologous cells that express the CAR. In some embodiments herein, CAR-expressing cells are generated using lentiviral vectors expressing CAR and truncated forms of human egfr (huegfrt). Co-expression of huEGFRt allows selection and purification of CAR-expressing T cells using antibodies that recognize huEGFRt (e.g., cetuximab, see PCT publication No. WO 2011/056894, which is incorporated herein by reference), as described in section VI above.

In some embodiments, by lysing erythrocytes and depleting monocytes, e.g., by PERCOLLTMGradient centrifugation or convection centrifugation elutriation (counterflow centrifugation) to separate T cells from peripheral blood lymphocytes specific subpopulations of T cells, such as CD3+, CD28+, CD4+, CD8+, CD45RA + and CD45RO + T cells, may be further isolated by positive or negative selection techniquesM-450CD3/CD28T was incubated for a time sufficient to positively select for the desired T cells, which could be isolated, see U.S. published application No. US20140271635A 1. In a non-limiting example, the time is about 30 minutes. In other non-limiting examples, the time ranges from 30 minutes to 36 hours or more, and integer values in between. In further non-limiting examples, the time is at least 1,2, 3, 4, 5, 6 hours, 10 to 24 hours, or more. In any case where T cells are rare compared to other cell types (e.g., isolated from immunocompromised individuals), longer incubation times can be used to isolate T cells. Furthermore, the efficiency of capturing CD8+ T cells can be improved using longer incubation times. Due to the fact thatThis, by simply shortening or extending the time allowed for T cells to bind to CD3/CD28 beads and/or by increasing or decreasing the bead to T cell ratio (as described further herein), a subset of T cells can be preferentially selected or deselected at the start of culture or at other time points in the process. In addition, by increasing or decreasing the proportion of anti-CD 3 and/or anti-CD 28 antibodies on the beads or other surfaces, a subset of T cells can be preferentially selected or deselected at the start of culture or at other desired time points. Multiple rounds of selection may also be used.

Enrichment of T cell populations by negative selection can be accomplished using a combination of antibodies to surface markers specific to the negative selection cells. One approach is cell sorting and/or selection by negative magnetic immunoadhesion or flow cytometry using a mixture of monoclonal antibodies directed against cell surface markers present on negatively selected cells. For example, to enrich for CD4+ cells by negative selection, the monoclonal antibody cocktail typically includes antibodies against CD14, CD20, CD11b, CD16, HLA-DR, and CD 8. A population of T cells expressing one or more cytokines may be selected. Methods for screening for cell expression are disclosed in PCT publication No. WO 2013/126712.

To isolate a desired cell population by positive or negative selection, the cell concentration and surface (e.g., particles such as beads) can be varied to ensure maximum contact of cells and beads in some embodiments, 1 × 10 is used9Concentration of cells/ml in other embodiments, greater than 1 × 10 is used8In other embodiments, 10, 15,20, 25, 30, 35, 40, 45, 50, 65, 70, 75, 80, 85, 90, 95, or 100 × 10 is used6Cell concentration of cells/ml. Without being bound by theory, the use of high concentrations can result in increased cell yield, cell activation, and cell expansion. Lower concentrations of cells may also be used. Without being bound by theory, the mixture of T cells and surfaces (e.g., particles such as beads) is significantly diluted, minimizing particle-to-cell interactions. This allows selection of cells expressing a large amount of the desired antigen to be bound to the particles. For example, CD4+ T cells express higher levels of CD28 and are specific at dilute concentrationsCD8+ T cells are more efficiently captured in some embodiments, the cell concentration used is 5 × 106In other embodiments, the concentration used may be about 1 × 105From ml to 1 × 106Ml, and any integer value in between.

Methods of treatment

Provided herein are methods of treating cancer in a subject by administering to the subject a therapeutically effective amount of the tumor-targeted CAR T cells disclosed herein. Also provided herein are methods of inhibiting tumor growth or metastasis in a subject by administering to the subject a therapeutically effective amount of the tumor-targeting CAR T cells disclosed herein.

Specifically provided are methods of treating a GPC 3-positive cancer in a subject. In some embodiments, the method comprises administering to the subject a therapeutically effective amount of an isolated host cell comprising a nucleic acid molecule encoding a GPC 3-targeted CAR and a huEGFRt, or administering a therapeutically effective amount of an isolated host cell co-expressing a GPC 3-targeted CAR and a huEGFRt. In some embodiments, the GPC 3-positive cancer is HCC, melanoma, clear ovarian cell carcinoma, YST, neuroblastoma, hepatoblastoma, or Wilms tumor.

Also provided are methods of treating a GPC 2-positive cancer in a subject. In some embodiments, the method comprises administering to the subject a therapeutically effective amount of an isolated host cell comprising a nucleic acid molecule encoding a GPC 2-targeted CAR and a huEGFRt, or administering a therapeutically effective amount of an isolated host cell co-expressing a GPC 2-targeted CAR and a huEGFRt. In some embodiments, the GPC 2-positive cancer is neuroblastoma, acute lymphocytic leukemia, embryonal rhabdomyosarcoma, alveolar rhabdomyosarcoma, ewing's sarcoma, desmoplastic small round cell tumor, or osteosarcoma.

Further provided are methods of treating a mesothelin-positive cancer in a subject. In some embodiments, the method comprises administering to the subject a therapeutically effective amount of an isolated host cell comprising a nucleic acid molecule encoding a mesothelin-targeted CAR and a huEGFRt, or administering a therapeutically effective amount of an isolated host cell co-expressing a mesothelin-targeted CAR and a huEGFRt. In some embodiments, the mesothelin-positive cancer is mesothelioma, prostate cancer, lung cancer, gastric cancer, squamous cell carcinoma, pancreatic cancer, cholangiocarcinoma, triple negative breast cancer, or ovarian cancer.

In some embodiments of the methods disclosed herein, the isolated host cell is a T lymphocyte. In some examples, the T lymphocyte is an autologous T lymphocyte.

A therapeutically effective amount of CAR-expressing T cells will depend on the severity of the disease, the type of disease, and the overall health of the patient. A therapeutically effective amount of CAR-expressing T cells and compositions thereof is that which provides subjective relief or an objectively determinable improvement in symptoms (e.g., reduction in tumor volume or metastasis) as indicated by a clinician or other qualified observer.

Administration of the CAR-expressing T cells and compositions disclosed herein can also be accompanied by administration of other anti-cancer drugs or therapeutic treatments (e.g., surgical resection of a tumor). Any suitable anti-cancer agent can be administered in combination with the compositions disclosed herein. Exemplary anticancer agents include, but are not limited to, chemotherapeutic agents, e.g., mitotic inhibitors, alkylating agents, antimetabolites, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzymes, topoisomerase inhibitors, anti-survival agents, biological response modifiers, anti-hormones (e.g., anti-androgens), and anti-angiogenic agents. Other anti-cancer therapies include radiation therapy and other antibodies that specifically target cancer cells.

Non-limiting examples of alkylating agents include nitrogen mustards (such as dichloromethyldiethanolamine (mechlororethamine), cyclophosphamide, melphalan, uracil mustard or chlorambucil), alkyl sulfonates (such as busulfan), nitrosoureas (such as carmustine, lomustine, semustine, streptozotocin or dacarbazine).

Non-limiting examples of antimetabolites include folic acid analogs (e.g., methotrexate), pyrimidine analogs (e.g., 5-FU or cytarabine), and purine analogs, such as mercaptopurine or thioguanine.

Non-limiting examples of natural products include vinblastines (e.g., vinblastine, vincristine, or vindesine), epipodophyllotoxins (e.g., etoposide or teniposide), antibiotics (e.g., actinomycin d, daunorubicin, doxorubicin, bleomycin, plicamycin, or mitomycin C), and enzymes (L-asparaginase).

Non-limiting examples of heteroreagents include platinum coordination complexes (e.g., cis-diamine-dichloroplatinum II, also known as cisplatin), substituted ureas (e.g., hydroxyurea), methylhydrazine derivatives (e.g., procarbazine), and adrenocortical suppressants (e.g., mitotane and aminoglutethimide).

Non-limiting examples of hormones and antagonists include adrenocorticosteroids (such as prednisone), progestins (such as hydroxyprogesterone caproate, medroxyprogesterone acetate and megestrol acetate), estrogens (such as diethylstilbestrol and ethinylestradiol), antiestrogens (such as tamoxifen), and androgens (such as testosterone propionate and fluoxymesterone). Examples of the most commonly used chemotherapeutic drugs include doxorubicin, melphalan (Alkeran), cytarabine (Ara-C), BiCNU, busulfan, CCNU, carboplatin (carboplatin), cisplatin, cyclophosphamide (Cytoxan), daunomycin, DTIC, 5-FU, fludarabine, hydroxyurea (Hydrea), Idarubicin (Idarubicin), ifosfamide, methotrexate, Mithramycin (Mithramycin), mitomycin, mitoxantrone, mechlorethamine, paclitaxel (or other taxanes such as docetaxel), vinblastine (Velban), vincristine, VP-16, and some newer drugs include gemcitabine (Gemzar), Herceptin (Herceptin), Irinotecan (Irinotecan) (Camptosar, CPT-11), cladribine (Leustatin), Navelbine (Navelbine), rituximab STI-571, taxotere, topotecan (Hycamtin), Hirodar (capecitabine), Zevelin (Zevelin), and calcitriol.

Non-limiting examples of immunomodulators that can be used include AS-101(Wyeth-Ayerst Labs.), bromopirine (bropirimine) (Upjohn), interferon-gamma (Genentech), GM-CSF (granulocyte macrophage colony stimulating factor; Genetics Institute), IL-2(Cetus or Hoffman-LaRoche), human immunoglobulin (cuter biological), IMREG (from Imreg of New orans, La.), SK & F106528, and TNF (tumor necrosis factor; Genentech).

Another common treatment for certain types of cancer is surgical treatment, such as surgical removal of the cancer or a portion thereof. Another example of treatment is radiation therapy, such as the application of radioactive substances or energy (e.g., external beam therapy) to the tumor site prior to surgical resection to help eradicate or shrink the tumor.

The following examples are provided to illustrate certain specific features and/or embodiments. These examples should not be construed as limiting the disclosure to the specific features or embodiments described.

Examples

126页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:半胱氨酸工程化的抗原结合分子

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!